1
|
Gurashi K, Wang YH, Amaral FMR, Spence K, Cant R, Yao CY, Lin CC, Wirth C, Wedge DC, Montalban-Bravo G, Colla S, Tien HF, Somervaille TCP, Batta K, Wiseman DH. An integrative multiparametric approach stratifies putative distinct phenotypes of blast phase chronic myelomonocytic leukemia. Cell Rep Med 2025; 6:101933. [PMID: 39892394 DOI: 10.1016/j.xcrm.2025.101933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/19/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Approximately 30% of patients with chronic myelomonocytic leukemia (CMML) undergo transformation to a chemo-refractory blastic phase (BP-CMML). Seeking novel therapeutic approaches, we profiled blast transcriptomes from 42 BP-CMMLs, observing extensive transcriptional heterogeneity and poor alignment to current acute myeloid leukemia (AML) classifications. BP-CMMLs display distinctive transcriptomic profiles, including enrichment for quiescence and variability in drug response signatures. Integrating clinical, immunophenotype, and transcriptome parameters, Random Forest unsupervised clustering distinguishes immature and mature subtypes characterized by differential expression of transcriptional modules, oncogenes, apoptotic regulators, and patterns of surface marker expression. Subtypes differ in predicted response to AML drugs, validated ex vivo in primary samples. Iteratively refined stratification resolves a classification structure comprising five subtypes along a maturation spectrum, predictive of response to novel agents including consistent patterns for receptor tyrosine kinase (RTK), cyclin-dependent kinase (CDK), mechanistic target of rapamycin (MTOR), and mitogen-activated protein kinase (MAPK) inhibitors. Finally, we generate a prototype decision tree to stratify BP-CMML with high specificity and sensitivity, requiring validation but with potential clinical applicability to guide personalized drug selection for improved outcomes.
Collapse
Affiliation(s)
- Kristian Gurashi
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Yu-Hung Wang
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Fabio M R Amaral
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Katherine Spence
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Rachel Cant
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Chi-Yuan Yao
- The National University Hospital of Taiwan, Taipei, Taiwan
| | - Chien-Chin Lin
- The National University Hospital of Taiwan, Taipei, Taiwan
| | - Christopher Wirth
- Wedge Group, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - David C Wedge
- Wedge Group, Manchester Cancer Research Centre, University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester, UK
| | | | - Simona Colla
- Departments of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Hwei-Fang Tien
- The National University Hospital of Taiwan, Taipei, Taiwan
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK; The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Kiran Batta
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| | - Daniel H Wiseman
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK; The Christie Hospital NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
2
|
Burban A, Tessier C, Larroquette M, Guyon J, Lubiato C, Pinglaut M, Toujas M, Galvis J, Dartigues B, Georget E, Luchman HA, Weiss S, Cappellen D, Nicot N, Klink B, Nikolski M, Brisson L, Mathivet T, Bikfalvi A, Daubon T, Sharanek A. Exploiting metabolic vulnerability in glioblastoma using a brain-penetrant drug with a safe profile. EMBO Mol Med 2025:10.1038/s44321-025-00195-6. [PMID: 39901019 DOI: 10.1038/s44321-025-00195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/05/2025] Open
Abstract
Glioblastoma is one of the most treatment-resistant and lethal cancers, with a subset of self-renewing brain tumour stem cells (BTSCs), driving therapy resistance and relapse. Here, we report that mubritinib effectively impairs BTSC stemness and growth. Mechanistically, bioenergetic assays and rescue experiments showed that mubritinib targets complex I of the electron transport chain, thereby impairing BTSC self-renewal and proliferation. Gene expression profiling and Western blot analysis revealed that mubritinib disrupts the AMPK/p27Kip1 pathway, leading to cell-cycle impairment. By employing in vivo pharmacokinetic assays, we established that mubritinib crosses the blood-brain barrier. Using preclinical patient-derived and syngeneic models, we demonstrated that mubritinib delays glioblastoma progression and extends animal survival. Moreover, combining mubritinib with radiotherapy or chemotherapy offers survival advantage to animals. Notably, we showed that mubritinib alleviates hypoxia, thereby enhancing ROS generation, DNA damage, and apoptosis in tumours when combined with radiotherapy. Encouragingly, toxicological and behavioural studies revealed that mubritinib is well tolerated and spares normal cells. Our findings underscore the promising therapeutic potential of mubritinib, warranting its further exploration in clinic for glioblastoma therapy.
Collapse
Affiliation(s)
- Audrey Burban
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Cloe Tessier
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | | | - Joris Guyon
- CHU of Bordeaux, Service de Pharmacologie Médicale, Bordeaux, France
- University of Bordeaux, INSERM, BPH, U1219, Bordeaux, France
| | - Cloe Lubiato
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Mathis Pinglaut
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Maxime Toujas
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Johanna Galvis
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Benjamin Dartigues
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux, France
| | - Emmanuelle Georget
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - H Artee Luchman
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - David Cappellen
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Nathalie Nicot
- LuxGen Genome Center, Luxembourg Institute of Health, Laboratoire national de santé, Dudelange, Luxembourg
| | - Barbara Klink
- LuxGen Genome Center, Luxembourg Institute of Health, Laboratoire national de santé, Dudelange, Luxembourg
- National Center of Genetics (NCG), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Luxembourg, 1526, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Macha Nikolski
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux, France
| | - Lucie Brisson
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Thomas Mathivet
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Andreas Bikfalvi
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France.
| | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France.
| | - Ahmad Sharanek
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France.
| |
Collapse
|
3
|
Tjahjono E, Daneman MR, Meika B, Revtovich AV, Kirienko NV. Mitochondrial abnormalities as a target of intervention in acute myeloid leukemia. Front Oncol 2025; 14:1532857. [PMID: 39902131 PMCID: PMC11788353 DOI: 10.3389/fonc.2024.1532857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy; it is the most common acute leukemia in adults. AML prognosis is often poor, and relapse often occurs after initial remission. Recurrent genetic abnormalities underlying this disease and the presence of leukemic stem cells complicate disease treatment. However, the complex metabolic reprogramming that enables the unrestrained cell growth seen in these cells may also be their Achilles' heel. In these cells, mitophagy operates as a double-edged sword. On one hand, it provides a source of building blocks for further cell division and serves as a method for removing damaged organelles, promoting cell survival. However, the profound metabolic changes to mitochondria also render these organelles more sensitive to damage and place them precariously close to excess mitophagic activation. This review discusses the dual role mitophagy plays in AML survival, the importance of targeting mitophagy to treat AML, and current progress in the area. The discovery and mechanism of action of multiple compounds that were used to inhibit or stimulate mitophagy and their effects on AML survival are also described. Further, we explore the combination strategy of mitophagy-targeting compounds with existing and/or novel chemotherapeutics to eradicate AML and discuss strategies to uncover new drug targets and novel mitochondria-targeting drugs.
Collapse
|
4
|
Sokei J, Kanefsky J, Sykes SM. Reprogramming of Fatty Acid Metabolism in Acute Leukemia. J Cell Physiol 2025; 240:e70000. [PMID: 39835485 DOI: 10.1002/jcp.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Fatty acids are essential biomolecules that support several cellular processes, such as membrane structures, energy storage and production, as well as signal transduction. Accordingly, changes in fatty acid metabolism can have a significant impact on cell behavior, such as growth, survival, proliferation, differentiation, and motility. Therefore, it is not surprising that many aspects of fatty acid metabolism are frequently dysregulated in human cancer, including in highly aggressive blood cancers such as acute leukemia. The aims of this review are to summarize the aspects of fatty acid metabolism that are specifically coopted in acute leukemia as well as current preclinical strategies for targeting fatty acid metabolism in these cancers.
Collapse
Affiliation(s)
- Judith Sokei
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Joice Kanefsky
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Stephen M Sykes
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University in Saint Louis, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Salomoni P, Flanagan AM, Cottone L. (B)On(e)-cohistones and the epigenetic alterations at the root of bone cancer. Cell Death Differ 2025; 32:66-77. [PMID: 37828086 PMCID: PMC11748643 DOI: 10.1038/s41418-023-01227-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Identification of mutations in histones in a number of human neoplasms and developmental syndromes represents the most compelling evidence to date for a causal role of epigenetic perturbations in human disease. In most cases, these mutations have gain of function properties that cause deviation from normal developmental processes leading to embryo defects and/or neoplastic transformation. These exciting discoveries represent a step-change in our understanding of the role of chromatin (dys)regulation in development and disease. However, the mechanisms of action of oncogenic histone mutations (oncohistones) remain only partially understood. Here, we critically assess existing literature on oncohistones focussing mainly on bone neoplasms. We show how it is possible to draw parallels with some of the cell-autonomous mechanisms of action described in paediatric brain cancer, although the functions of oncohistones in bone tumours remain under-investigated. In this respect, it is becoming clear that histone mutations targeting the same residues display, at least in part, tissue-specific oncogenic mechanisms. Furthermore, it is emerging that cancer cells carrying oncohistones can modify the surrounding microenvironment to support growth and/or alter differentiation trajectories. A better understanding of oncohistone function in different neoplasms provide potential for identification of signalling that could be targeted therapeutically. Finally, we discuss some of the main concepts and future directions in this research area, while also drawing possible connections and parallels with other cancer epigenetic mechanisms.
Collapse
Affiliation(s)
- Paolo Salomoni
- Nuclear Function Group, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.
| | - Adrienne M Flanagan
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, UK
- Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Lucia Cottone
- Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
6
|
Tatarata QZ, Wang Z, Konopleva M. BCL-2 inhibition in acute myeloid leukemia: resistance and combinations. Expert Rev Hematol 2024; 17:935-946. [PMID: 39552410 DOI: 10.1080/17474086.2024.2429604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The introduction of venetoclax has revolutionized the treatment landscape of acute myeloid leukemia, offering new therapeutic opportunities. However, the clinical response to venetoclax varies significantly between patients, with many experiencing limited duration of response. AREAS COVERED Identified resistance mechanisms include both intrinsic and acquired resistance to VEN. The former is associated with cell lineage and differentiation state. The latter includes dependency on alternative BCL-2 family anti-apoptotic protein(s) mediated by genetic, epigenetic, or post-translational mechanisms, mitochondrial and metabolic involvement, as well as microenvironment. Understanding these mechanisms is crucial for optimizing venetoclax-based therapies and enhancing treatment outcomes for patients with acute myeloid leukemia. This review aims to elucidate the primary mechanisms underlying resistance to venetoclax and explore current therapeutic strategies to overcome this challenge. EXPERT OPINION In patients with venetoclax resistance, alternative options include targeted combination therapies tailored to individual cases based on cytogenetics and prior treatments. Many of these therapies require further clinical investigation to validate their safety and efficacy.
Collapse
Affiliation(s)
- Qi Zhang Tatarata
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- The Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhe Wang
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
7
|
Chen Y, Xian M, Ying W, Liu J, Bing S, Wang X, Yu J, Xu X, Xiang S, Shao X, Cao J, He Q, Yang B, Ying M. Succinate dehydrogenase deficiency-driven succinate accumulation induces drug resistance in acute myeloid leukemia via ubiquitin-cullin regulation. Nat Commun 2024; 15:9820. [PMID: 39537588 PMCID: PMC11560925 DOI: 10.1038/s41467-024-53398-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Drug resistance is vital for the poor prognosis of acute myeloid leukemia (AML) patients, but the underlying mechanism remains poorly understood. Given the unique microenvironment of bone marrow, we reasoned that drug resistance of AML might rely on distinct metabolic processes. Here, we identify succinate dehydrogenase (SDH) deficiency and over-cumulative succinate as typical features in AML, with a marked function in causing the resistance of AML cells to various anti-cancer therapies. Mechanistically, succinate promotes the accumulation of oncogenic proteins in a manner that precedes transcriptional activation. This function is mediated by succinate-triggered upregulation of ubiquitin-conjugating enzyme E2M (UBC12) phosphorylation, which impairs its E2 function in cullins neddylation. Notably, decreasing succinate by fludarabine can restore the sensitivity of anti-cancer drugs in SDH-deficient AML. Together, we uncover the function of succinate in driving drug resistance by regulating p-UBC12/cullin activity, and indicate reshaping succinate metabolism as a promising treatment for SDH-deficient AML.
Collapse
Affiliation(s)
- Yifan Chen
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Miao Xian
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wenwen Ying
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiayi Liu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shaowei Bing
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaomin Wang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiayi Yu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaojun Xu
- Division of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Senfeng Xiang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xuejing Shao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Meidan Ying
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Division of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
8
|
Qureshi Z, Altaf F, Jamil A, Siddique R. Safety, Efficacy, and Predictive Factors of Venetoclax-Based Regimens in Elderly Acute Myeloid Leukemia Patients: A Meta-Analysis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:e835-e851. [PMID: 39218712 DOI: 10.1016/j.clml.2024.07.004] [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: 06/14/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024]
Abstract
Venetoclax synergizes with low-intensity regimens such as hypomethylating agents (HMAs) and low-dose cytarabine (LDAC). However, less is known about the clinical activity of venetoclax combined with HMAs or LDAC. Therefore, the current study focused on assessing the clinical efficacy, safety, and predictive factors for response to these venetoclax-based regimens in elderly patients with acute myeloid leukemia (AML). A comprehensive search for literature related to our study objective was performed on PubMed, Embase, Web of Science, and Google Scholar databases. The statistical analyses were performed using Review Manager or the Comprehensive Meta-Analysis software. In addition, methodological quality evaluation of nonrandomized studies was conducted using the Newcastle Ottawa Scale, while bias assessment of randomized studies was performed with Cochrane's risk of bias tool. Twelve studies, including 1432 elderly AML patients treated with venetoclax-based regimens, were identified for review and analysis. The pooled analysis showed that the rate of complete response with or without incomplete blood count recovery (CR/CRi) and overall response rate (ORR) among patients treated with venetoclax and HMAs was 59% and 64%, respectively. On the other hand, an CR/CRi of 50% was observed in patients treated with venetoclax and LDAC. Furthermore, venetoclax combined with HMAs demonstrated a significant survival benefit over HMAs alone and intensive chemotherapy (HR: 0.57; 95% CI: 0.47-0.68; P < .00001). The most common grade ≥ 3 hematologic disorder, nonhematological event, and infection in AML patients treated with venetoclax and HMAs were febrile neutropenia (39%), hypokalemia (12%), and pneumonia (19%), respectively. Conversely, thrombocytopenia, hypokalemia, and pneumonia were more common in patients treated with venetoclax and LDAC (41%, 15%, and 12%, respectively). Venetoclax combined with HMAs or LDAC has good clinical activity and a manageable safety profile in elderly patients with AML.
Collapse
Affiliation(s)
- Zaheer Qureshi
- Department of Medicine, The Frank H. Netter MD School of Medicine at Quinnipiac University, Bridgeport, CT.
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre Watertown, Watertown, NY
| | | |
Collapse
|
9
|
Boreel DF, Beerkens AP, Heskamp S, Boswinkel M, Peters JP, Adema GJ, Span PN, Bussink J. Inhibition of OXPHOS induces metabolic rewiring and reduces hypoxia in murine tumor models. Clin Transl Radiat Oncol 2024; 49:100875. [PMID: 39469146 PMCID: PMC11513494 DOI: 10.1016/j.ctro.2024.100875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Tumor hypoxia is a feature of many solid malignancies and is known to cause radio resistance. In recent years it has become clear that hypoxic tumor regions also foster an immunosuppressive phenotype and are involved in immunotherapy resistance. It has been proposed that reducing the tumors' oxygen consumption will result in an increased oxygen concentration in the tissue and improve radio- and immunotherapy efficacy. The aim of this study is to investigate the metabolic rewiring of cancer cells by pharmacological attenuation of oxidative phosphorylation (OXPHOS) and subsequently reduce tumor hypoxia. Material and methods The metabolic effects of three OXPHOS inhibitors IACS-010759, atovaquone and metformin were explored by measuring oxygen consumption rate, extra cellular acidification rate, and [18F]FDG uptake in 2D and 3D cell culture. Tumor cell growth in 2D cell culture and hypoxia in 3D cell culture were analyzed by live cell imaging. Tumor hypoxia and [18F]FDG uptake in vivo following treatment with IACS-010759 was determined by immunohistochemistry and ex vivo biodistribution respectively. Results In vitro experiments show that tumor cell metabolism is heterogeneous between different models. Upon OXPHOS inhibition, metabolism shifts from oxygen consumption through OXPHOS towards glycolysis, indicated by increased acidification and glucose uptake. Inhibition of OXPHOS by IACS-010759 treatment reduced diffusion limited tumor hypoxia in both 3D cell culture and in vivo. Although immune cell presence was lower in hypoxic areas compared with normoxic areas, it is not altered following short term OXPHOS inhibition. Discussion These results show that inhibition of OXPHOS causes a metabolic shift from OXPHOS towards increased glycolysis in 2D and 3D cell culture. Moreover, inhibition of OXPHOS reduces diffusion limited hypoxia in 3D cell culture and murine tumor models. Reduced hypoxia by OXPHOS inhibition might enhance therapy efficacy in future studies. However, caution is warranted as systemic metabolic rewiring can cause adverse effects.
Collapse
Affiliation(s)
- Daan F. Boreel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
- Department of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Anne P.M. Beerkens
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
- Department of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Milou Boswinkel
- Department of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Johannes P.W. Peters
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
| | - Gosse J. Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
| | - Paul N. Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
| |
Collapse
|
10
|
Yang J, Wang Z, Wu K, Nie B, Li L, Ruan J, Zhou Q, Zeng Y, Shi M. Isocitrate dehydrogenase 2 mutation promotes cytarabine resistance in acute myeloid leukemia by Warburg effect. Hematol Oncol 2024; 42:e3316. [PMID: 39526588 PMCID: PMC11590050 DOI: 10.1002/hon.3316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
Mutation of isocitrate dehydrogenase 2 (IDH2) is a key factor in promoting cytarabine (Ara-C) resistance in acute myeloid leukemia (AML), however the underly mechanism remains unclear. Acute myeloid leukemia cells, were cultured with either IDH2 knockdown (KD-IDH2) or overexpression (OE-IDH2) to elucidate the role of IDH2 in these leukemic cell lines. Additionally, mutant cell lines were engineered to replicate clinically relevant IDH2 mutations. To investigate cellular responses, the glycolytic inhibitor 2-deoxy-D-glucose (2-DG) was administered to the cells. Cell proliferation was quantified using a Cell Counting Kit-8 (CCK-8), while apoptosis was evaluated through propidium iodide staining followed by flow cytometry. Glycolytic metabolism levels were measured using a specific reagent kit, and Western blotting was employed to determine the expression levels of glycolysis-related proteins. Transcriptome sequencing was conducted to elucidate the mechanisms by which IDH2 mutations influence glycolysis. Furthermore, both in vitro cell experiments and in vivo subcutaneous transplantation tumor models in nude mice were utilized to validate these mechanisms. OE-IDH2 in AML cells, enhances resistance to the Ara-C, promotes cell proliferation and glycolysis, and inhibits apoptosis. KD-IDH2 exhibits opposite effects. Both IDH2 mutations and OE-IDH2 produce similar effects on these cellular processes. The increase in glycolysis levels following IDH2 mutation may contribute to the reduced efficacy of Enasidenib in inhibiting the proliferation of IDH-mutant AML cells. Transcriptome sequencing results indicate an enrichment of the PI3K/Akt signaling pathway in IDH2-mutant AML cells. BEZ235 significantly inhibits the expression of phosphorylated PI3K (p-PI3K), phosphorylated Akt (p-Akt), mTOR, glycolytic metabolism, and Ara-C resistance both in vitro and in vivo. Overexpression and mutation of IDH2 coordinate with the Warburg effect through the PI3K/Akt/mTOR pathway to promote Ara-C resistance in AML.
Collapse
MESH Headings
- Cytarabine/pharmacology
- Isocitrate Dehydrogenase/genetics
- Isocitrate Dehydrogenase/metabolism
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Animals
- Mice
- Drug Resistance, Neoplasm
- Mutation
- Warburg Effect, Oncologic
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Mice, Nude
- Glycolysis/drug effects
- Antimetabolites, Antineoplastic/pharmacology
Collapse
Affiliation(s)
- Jinrong Yang
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Zixu Wang
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Kun Wu
- Department of Clinical LaboratoryThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Yunnan Key Laboratory of Laboratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Bo Nie
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Liyin Li
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Jingyan Ruan
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Qiang Zhou
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Yun Zeng
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Mingxia Shi
- Department of HematologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Hematology Research Center of Yunnan ProvinceThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
11
|
Shahswar R, Ganser A. Relapse and resistance in acute myeloid leukemia post venetoclax: improving second lines therapy and combinations. Expert Rev Hematol 2024; 17:723-739. [PMID: 39246164 DOI: 10.1080/17474086.2024.2402283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION The combined use of the BCL-2 inhibitor venetoclax with azacitidine now is the standard of care for patients with acute myeloid leukemia (AML) unfit for intensive chemotherapy with outcomes exceeding those achieved with hypomethylating agents alone. Venetoclax in combination with intensive chemotherapy is also increasingly used both as frontline as well as salvage therapy. However, resistance to and relapse after venetoclax-based therapies are of major concern and outcomes after treatment failure remain poor. AREAS COVERED A comprehensive search was performed using PubMed database (up to April 2024). Studies evaluating venetoclax-based combination treatments in AML and studies assessing markers of response and resistance to venetoclax were investigated. We summarize the status of venetoclax-based therapies in the frontline and relapsed/refractory setting with focus on the main mechanisms of resistance to BCL-2 inhibition. Further, strategies to overcome resistance including combinatorial regimens of hypomethylating agent (HMA) + venetoclax + inhibitors targeting actionable mutations like IDH1/2 or FLT3-ITD and the introduction of novel agents like menin-inhibitors are addressed. EXPERT OPINION Although venetoclax is reshaping the treatment of unfit and fit AML patients, prognosis of patients after HMA/VEN failure remains dismal, and strategies to abrogate primary and secondary resistance are an unmet clinical need.
Collapse
Affiliation(s)
- Rabia Shahswar
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Boët E, Saland E, Skuli S, Griessinger E, Sarry JE. [ Mitohormesis: a key driver of the therapy resistance in cancer cells]. C R Biol 2024; 347:59-75. [PMID: 39171610 DOI: 10.5802/crbiol.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 08/23/2024]
Abstract
A large body of literature highlights the importance of energy metabolism in the response of haematological malignancies to therapy. In this review, we are particularly interested in acute myeloid leukaemia, where mitochondrial metabolism plays a key role in response and resistance to treatment. We describe the new concept of mitohormesis in the response to therapy-induced stress and in the initiation of relapse in this disease.
Collapse
|
13
|
Álvarez-González E, Sierra LM. Tricarboxylic Acid Cycle Relationships with Non-Metabolic Processes: A Short Story with DNA Repair and Its Consequences on Cancer Therapy Resistance. Int J Mol Sci 2024; 25:9054. [PMID: 39201738 PMCID: PMC11355010 DOI: 10.3390/ijms25169054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Metabolic changes involving the tricarboxylic acid (TCA) cycle have been linked to different non-metabolic cell processes. Among them, apart from cancer and immunity, emerges the DNA damage response (DDR) and specifically DNA damage repair. The oncometabolites succinate, fumarate and 2-hydroxyglutarate (2HG) increase reactive oxygen species levels and create pseudohypoxia conditions that induce DNA damage and/or inhibit DNA repair. Additionally, by influencing DDR modulation, they establish direct relationships with DNA repair on at least four different pathways. The AlkB pathway deals with the removal of N-alkylation DNA and RNA damage that is inhibited by fumarate and 2HG. The MGMT pathway acts in the removal of O-alkylation DNA damage, and it is inhibited by the silencing of the MGMT gene promoter by 2HG and succinate. The other two pathways deal with the repair of double-strand breaks (DSBs) but with opposite effects: the FH pathway, which uses fumarate to help with the repair of this damage, and the chromatin remodeling pathway, in which oncometabolites inhibit its repair by impairing the homologous recombination repair (HRR) system. Since oncometabolites inhibit DNA repair, their removal from tumor cells will not always generate a positive response in cancer therapy. In fact, their presence contributes to longer survival and/or sensitization against tumor therapy in some cancer patients.
Collapse
Affiliation(s)
- Enol Álvarez-González
- Departamento de Biología Funcional, Área de Genética, University of Oviedo, C/Julián Clavería s/n, 33006 Oviedo, Spain;
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avda. HUCA s/n, 33011 Oviedo, Spain
| | - Luisa María Sierra
- Departamento de Biología Funcional, Área de Genética, University of Oviedo, C/Julián Clavería s/n, 33006 Oviedo, Spain;
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avda. HUCA s/n, 33011 Oviedo, Spain
| |
Collapse
|
14
|
He J, Qiu Z, Fan J, Xie X, Sheng Q, Sui X. Drug tolerant persister cell plasticity in cancer: A revolutionary strategy for more effective anticancer therapies. Signal Transduct Target Ther 2024; 9:209. [PMID: 39138145 PMCID: PMC11322379 DOI: 10.1038/s41392-024-01891-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 08/15/2024] Open
Abstract
Non-genetic mechanisms have recently emerged as important drivers of anticancer drug resistance. Among these, the drug tolerant persister (DTP) cell phenotype is attracting more and more attention and giving a predominant non-genetic role in cancer therapy resistance. The DTP phenotype is characterized by a quiescent or slow-cell-cycle reversible state of the cancer cell subpopulation and inert specialization to stimuli, which tolerates anticancer drug exposure to some extent through the interaction of multiple underlying mechanisms and recovering growth and proliferation after drug withdrawal, ultimately leading to treatment resistance and cancer recurrence. Therefore, targeting DTP cells is anticipated to provide new treatment opportunities for cancer patients, although our current knowledge of these DTP cells in treatment resistance remains limited. In this review, we provide a comprehensive overview of the formation characteristics and underlying drug tolerant mechanisms of DTP cells, investigate the potential drugs for DTP (including preclinical drugs, novel use for old drugs, and natural products) based on different medicine models, and discuss the necessity and feasibility of anti-DTP therapy, related application forms, and future issues that will need to be addressed to advance this emerging field towards clinical applications. Nonetheless, understanding the novel functions of DTP cells may enable us to develop new more effective anticancer therapy and improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Jun He
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zejing Qiu
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jingjing Fan
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xiaohong Xie
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Qinsong Sheng
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xinbing Sui
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
15
|
Wang J, Han L, Liu Z, Zhang W, Zhang L, Jing J, Gao A. Targeting IGF2BP1 alleviated benzene hematotoxicity by reprogramming BCAA metabolism and fatty acid oxidation. Chem Biol Interact 2024; 398:111107. [PMID: 38866309 DOI: 10.1016/j.cbi.2024.111107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
Benzene is the main environmental pollutant and risk factor of childhood leukemia and chronic benzene poisoning. Benzene exposure leads to hematopoietic stem and progenitor cell (HSPC) dysfunction and abnormal blood cell counts. However, the key regulatory targets and mechanisms of benzene hematotoxicity are unclear. In this study, we constructed a benzene-induced hematopoietic damage mouse model to explore the underlying mechanisms. We identified that Insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) was significantly reduced in benzene-exposed mice. Moreover, targeting IGF2BP1 effectively mitigated damages to hematopoietic function and hematopoietic molecule expression caused by benzene in mice. On the mechanics, by metabolomics and transcriptomics, we discovered that branched-chain amino acid (BCAA) metabolism and fatty acid oxidation were key metabolic pathways, and Branched-chain amino acid transaminase 1 (BCAT1) and Carnitine palmitoyltransferase 1a (CPT1A) were critical metabolic enzymes involved in IGF2BP1-mediated hematopoietic injury process. The expression of the above molecules in the benzene exposure population was also examined and consistent with animal experiments. In conclusion, targeting IGF2BP1 alleviated hematopoietic injury caused by benzene exposure, possibly due to the reprogramming of BCAA metabolism and fatty acid oxidation via BCAT1 and CPT1A metabolic enzymes. IGF2BP1 is a potential regulatory and therapeutic target for benzene hematotoxicity.
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lin Han
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Ziyan Liu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Wei Zhang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lei Zhang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Jiaru Jing
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Ai Gao
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
16
|
Savino AM, Stuani L. Targeting glycolysis to rescue 2-hydroxyglutarate immunosuppressive effects in dendritic cells and acute myeloid leukemia. Haematologica 2024; 109:2388-2390. [PMID: 38497144 PMCID: PMC11290541 DOI: 10.3324/haematol.2023.284893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024] Open
MESH Headings
- Glycolysis/drug effects
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Dendritic Cells/metabolism
- Dendritic Cells/immunology
- Humans
- Glutarates/metabolism
- Animals
- Mice
Collapse
Affiliation(s)
- Angela Maria Savino
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca
| | - Lucille Stuani
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Univ Montpellier, Institut régional du Cancer de Montpellier (ICM), INSERM U1194, Montpellier, France; Equipe Labellisée Ligue Contre le Cancer, Paris.
| |
Collapse
|
17
|
Hammon K, Renner K, Althammer M, Voll F, Babl N, Decking SM, Siska PJ, Matos C, Conejo ZEC, Mendes K, Einwag F, Siegmund H, Iberl S, Berger RS, Dettmer K, Schoenmehl R, Brochhausen C, Herr W, Oefner PJ, Rehli M, Thomas S, Kreutz M. D-2-hydroxyglutarate supports a tolerogenic phenotype with lowered major histocompatibility class II expression in non-malignant dendritic cells and acute myeloid leukemia cells. Haematologica 2024; 109:2500-2514. [PMID: 38235501 PMCID: PMC11290548 DOI: 10.3324/haematol.2023.283597] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
D-2-hydroxyglutarate (D-2-HG) accumulates in patients with acute myeloid leukemia (AML) with mutated isocitrate dehydrogenase (IDH) and in other malignancies. D-2-HG suppresses antitumor T-cell immunity but little is known about potential effects on non-malignant myeloid cells. Here we show that D-2-HG impairs human but not murine dendritic cell differentiation, resulting in a tolerogenic phenotype with low major histocompatibility class II expression. In line with this, IDH-mutated AML blasts exhibited lower expression of HLA-DP and were less susceptible to lysis by HLA-DP-specific T cells. Interestingly, besides its expected impact on DNA demethylation, D-2-HG reprogrammed metabolism towards increased lactate production in dendritic cells and AML. Vitamin C accelerated DNA demethylation, but only the combination of vitamin C and glycolytic inhibition lowered lactate levels and supported major histocompatibility complex class II expression. Our results indicate an unexpected link between the immunosuppressive metabolites 2-HG and lactic acid and suggest a potentially novel therapeutic strategy with combinations of anti-glycolytic drugs and epigenetic modulators (hypomethylating agents) or other therapeutics for the treatment of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/drug effects
- Glutarates/metabolism
- Glutarates/pharmacology
- Mice
- Animals
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Phenotype
- Cell Differentiation/drug effects
- Lactic Acid/metabolism
- Immune Tolerance/drug effects
- Isocitrate Dehydrogenase/genetics
Collapse
Affiliation(s)
- Kathrin Hammon
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg, Germany; Department of Otorhinolaryngology, University Hospital Regensburg, Regensburg
| | - Michael Althammer
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Florian Voll
- LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Nathalie Babl
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Sonja-Maria Decking
- Department of Otorhinolaryngology, University Hospital Regensburg, Regensburg
| | - Peter J Siska
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Carina Matos
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | | | - Karina Mendes
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; Present address: Universidade Católica Portuguesa, Center for Interdisciplinary Research in Health (CIIS), Institute of Health Sciences (ICS); Viseu
| | - Friederike Einwag
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Heiko Siegmund
- Institute of Pathology, University of Regensburg; Regensburg
| | - Sabine Iberl
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Raffaela S Berger
- Institute of Functional Genomics, University of Regensburg; Regensburg
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg; Regensburg
| | - Rebecca Schoenmehl
- Institute of Pathology, University Medical Center Mannheim, University Heidelberg, Mannheim
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg; Regensburg, Germany; Institute of Pathology, University Medical Center Mannheim, University Heidelberg, Mannheim
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg; Regensburg
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Simone Thomas
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg; Regensburg, Germany; LIT - Leibniz Institute for Immunotherapy; Regensburg.
| |
Collapse
|
18
|
Kowalczyk A, Zarychta J, Lejman M, Latoch E, Zawitkowska J. Clinical Implications of Isocitrate Dehydrogenase Mutations and Targeted Treatment of Acute Myeloid Leukemia with Mutant Isocitrate Dehydrogenase Inhibitors-Recent Advances, Challenges and Future Prospects. Int J Mol Sci 2024; 25:7916. [PMID: 39063158 PMCID: PMC11276768 DOI: 10.3390/ijms25147916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Despite the better understanding of the molecular mechanisms contributing to the pathogenesis of acute myeloid leukemia (AML) and improved patient survival in recent years, AML therapy still remains a clinical challenge. For this reason, it is important to search for new therapies that will enable the achievement of remission. Recently, the Food and Drug Administration approved three mutant IDH (mIDH) inhibitors for the treatment of AML. However, the use of mIDH inhibitors in monotherapy usually leads to the development of resistance and the subsequent recurrence of the cancer, despite the initial effectiveness of the therapy. A complete understanding of the mechanisms by which IDH mutations influence the development of leukemia, as well as the processes that enable resistance to mIDH inhibitors, may significantly improve the efficacy of this therapy through the use of an appropriate synergistic approach. The aim of this literature review is to present the role of IDH1/IDH2 mutations in the pathogenesis of AML and the results of clinical trials using mIDH1/IDH2 inhibitors in AML and to discuss the challenges related to the use of mIDH1/IDH2 inhibitors in practice and future prospects related to the potential methods of overcoming resistance to these agents.
Collapse
Affiliation(s)
- Adrian Kowalczyk
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (J.Z.)
| | - Julia Zarychta
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (J.Z.)
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Eryk Latoch
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Bialystok, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
19
|
Khalaf A, de Beauchamp L, Kalkman E, Rattigan K, Himonas E, Jones J, James D, Shokry ESA, Scott MT, Dunn K, Tardito S, Copland M, Sumpton D, Shanks E, Helgason GV. Nutrient-sensitizing drug repurposing screen identifies lomerizine as a mitochondrial metabolism inhibitor of chronic myeloid leukemia. Sci Transl Med 2024; 16:eadi5336. [PMID: 38865484 DOI: 10.1126/scitranslmed.adi5336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
In chronic myeloid leukemia (CML), the persistence of leukemic stem cells (LSCs) after treatment with tyrosine kinase inhibitors (TKIs), such as imatinib, can lead to disease relapse. It is known that therapy-resistant LSCs rely on oxidative phosphorylation (OXPHOS) for their survival and that targeting mitochondrial respiration sensitizes CML LSCs to imatinib treatment. However, current OXPHOS inhibitors have demonstrated limited efficacy or have shown adverse effects in clinical trials, highlighting that identification of clinically safe oxidative pathway inhibitors is warranted. We performed a high-throughput drug repurposing screen designed to identify mitochondrial metabolism inhibitors in myeloid leukemia cells. This identified lomerizine, a US Food and Drug Administration (FDA)-approved voltage-gated Ca2+ channel blocker now used for the treatment of migraines, as one of the top hits. Transcriptome analysis revealed increased expression of voltage-gated CACNA1D and receptor-activated TRPC6 Ca2+ channels in CML LSCs (CD34+CD38-) compared with normal counterparts. This correlated with increased endoplasmic reticulum (ER) mass and increased ER and mitochondrial Ca2+ content in CML stem/progenitor cells. We demonstrate that lomerizine-mediated inhibition of Ca2+ uptake leads to ER and mitochondrial Ca2+ depletion, with similar effects seen after CACNA1D and TRPC6 knockdown. Through stable isotope-assisted metabolomics and functional assays, we observe that lomerizine treatment inhibits mitochondrial isocitrate dehydrogenase activity and mitochondrial oxidative metabolism and selectively sensitizes CML LSCs to imatinib treatment. In addition, combination treatment with imatinib and lomerizine reduced CML tumor burden, targeted CML LSCs, and extended survival in xenotransplantation model of human CML, suggesting this as a potential therapeutic strategy to prevent disease relapse in patients.
Collapse
MESH Headings
- Drug Repositioning
- Humans
- Mitochondria/metabolism
- Mitochondria/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Animals
- Cell Line, Tumor
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum/drug effects
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Mice
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Calcium/metabolism
- Oxidative Phosphorylation/drug effects
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
Collapse
Affiliation(s)
- Ahmed Khalaf
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Lucie de Beauchamp
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Eric Kalkman
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Kevin Rattigan
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Ekaterini Himonas
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Joe Jones
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Daniel James
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | | | - Mary T Scott
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Karen Dunn
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Saverio Tardito
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Mhairi Copland
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - David Sumpton
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Emma Shanks
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - G Vignir Helgason
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| |
Collapse
|
20
|
Li Z, Sai K, Ma G, Chen F, Xu X, Chen L, Wang S, Li W, Huang G, Cui P. Diterpenoid honatisine overcomes temozolomide resistance in glioblastoma by inducing mitonuclear protein imbalance through disruption of TFAM-mediated mtDNA transcription. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155328. [PMID: 38522316 DOI: 10.1016/j.phymed.2023.155328] [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: 08/30/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 03/26/2024]
Abstract
BACKGROUND Glioblastoma (GBM) represents as the most formidable intracranial malignancy. The systematic exploration of natural compounds for their potential applications in GBM therapy has emerged as a pivotal and fruitful avenue of research. PURPOSE In the present study, a panel of 96 diterpenoids was systematically evaluated as a repository of potential antitumour agents. The primary objective was to discern their potency in overcoming resistance to temozolomide (TMZ). Through an extensive screening process, honatisine, a heptacyclic diterpenoid alkaloid, emerged as the most robust candidate. Notably, honatisine exhibited remarkable efficacy in patient-derived primary and recurrent GBM strains. Subsequently, we subjected this compound to comprehensive scrutiny, encompassing GBM cultured spheres, GBM organoids (GBOs), TMZ-resistant GBM cell lines, and orthotopic xenograft mouse models of GBM cells. RESULTS Our investigative efforts delved into the mechanistic underpinnings of honatisine's impact. It was discerned that honatisine prompted mitonuclear protein imbalance and elicited the mitochondrial unfolded protein response (UPRmt). This effect was mediated through the selective depletion of mitochondrial DNA (mtDNA)-encoded subunits, with a particular emphasis on the diminution of mitochondrial transcription factor A (TFAM). The ultimate outcome was the instigation of deleterious mitochondrial dysfunction, culminating in apoptosis. Molecular docking and surface plasmon resonance (SPR) experiments validated honatisine's binding affinity to TFAM within its HMG-box B domain. This binding may promote phosphorylation of TFAM and obstruct the interaction of TFAM bound to heavy strand promoter 1 (HSP1), thereby enhancing Lon-mediated TFAM degradation. Finally, in vivo experiments confirmed honatisine's antiglioma properties. Our comprehensive toxicological assessments underscored its mild toxicity profile, emphasizing the necessity for a thorough evaluation of honatisine as a novel antiglioma agent. CONCLUSION In summary, our data provide new insights into the therapeutic mechanisms underlying honatisine's selective inducetion of apoptosis and its ability to overcome chemotherapy resistance in GBM. These actions are mediated through the disruption of mitochondrial proteostasis and function, achieved by the inhibition of TFAM-mediated mtDNA transcription. This study highlights honatisine's potential as a promising agent for glioblastoma therapy, underscoring the need for further exploration and investigation.
Collapse
Affiliation(s)
- Zongyang Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Ke Sai
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Guoxu Ma
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Fanfan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Xudong Xu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Lei Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Sicen Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Weiping Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Guodong Huang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China.
| | - Ping Cui
- Department of pharmacy, Shenzhen Children's Hospital, Shenzhen 518038, China.
| |
Collapse
|
21
|
Zheng J, Wang Q, Chen J, Cai G, Zhang Z, Zou H, Zou JX, Liu Q, Ji S, Shao G, Li H, Li S, Chen HW, Lu L, Yuan Y, Liu P, Wang J. Tumor mitochondrial oxidative phosphorylation stimulated by the nuclear receptor RORγ represents an effective therapeutic opportunity in osteosarcoma. Cell Rep Med 2024; 5:101519. [PMID: 38692271 PMCID: PMC11148566 DOI: 10.1016/j.xcrm.2024.101519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 12/12/2023] [Accepted: 03/27/2024] [Indexed: 05/03/2024]
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor with a poor prognosis. Here, we show that the nuclear receptor RORγ may serve as a potential therapeutic target in OS. OS exhibits a hyperactivated oxidative phosphorylation (OXPHOS) program, which fuels the carbon source to promote tumor progression. We found that RORγ is overexpressed in OS tumors and is linked to hyperactivated OXPHOS. RORγ induces the expression of PGC-1β and physically interacts with it to activate the OXPHOS program by upregulating the expression of respiratory chain component genes. Inhibition of RORγ strongly inhibits OXPHOS activation, downregulates mitochondrial functions, and increases ROS production, which results in OS cell apoptosis and ferroptosis. RORγ inverse agonists strongly suppressed OS tumor growth and progression and sensitized OS tumors to chemotherapy. Taken together, our results indicate that RORγ is a critical regulator of the OXPHOS program in OS and provides an effective therapeutic strategy for this deadly disease.
Collapse
Affiliation(s)
- Jianwei Zheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Qianqian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Jianghe Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Guodi Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhenhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hongye Zou
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - June X Zou
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Qianqian Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Shufeng Ji
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Guoli Shao
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Hong Li
- Biomedical Laboratory, Guangzhou Jingke Life Science Institute, Guangzhou, Guangdong 510145, P.R. China
| | - Sheng Li
- Biomedical Laboratory, Guangzhou Jingke Life Science Institute, Guangzhou, Guangdong 510145, P.R. China
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - LinLin Lu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, SAR, China
| | - Yanqiu Yuan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China.
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China; National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China.
| | - Junjian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China; National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China.
| |
Collapse
|
22
|
Hong JH, Yong CH, Heng HL, Chan JY, Lau MC, Chen J, Lee JY, Lim AH, Li Z, Guan P, Chu PL, Boot A, Ng SR, Yao X, Wee FYT, Lim JCT, Liu W, Wang P, Xiao R, Zeng X, Sun Y, Koh J, Kwek XY, Ng CCY, Klanrit P, Zhang Y, Lai J, Tai DWM, Pairojkul C, Dima S, Popescu I, Hsieh SY, Yu MC, Yeong J, Kongpetch S, Jusakul A, Loilome W, Tan P, Tan J, Teh BT. Integrative multiomics enhancer activity profiling identifies therapeutic vulnerabilities in cholangiocarcinoma of different etiologies. Gut 2024; 73:966-984. [PMID: 38050079 DOI: 10.1136/gutjnl-2023-330483] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023]
Abstract
OBJECTIVES Cholangiocarcinoma (CCA) is a heterogeneous malignancy with high mortality and dismal prognosis, and an urgent clinical need for new therapies. Knowledge of the CCA epigenome is largely limited to aberrant DNA methylation. Dysregulation of enhancer activities has been identified to affect carcinogenesis and leveraged for new therapies but is uninvestigated in CCA. Our aim is to identify potential therapeutic targets in different subtypes of CCA through enhancer profiling. DESIGN Integrative multiomics enhancer activity profiling of diverse CCA was performed. A panel of diverse CCA cell lines, patient-derived and cell line-derived xenografts were used to study identified enriched pathways and vulnerabilities. NanoString, multiplex immunohistochemistry staining and single-cell spatial transcriptomics were used to explore the immunogenicity of diverse CCA. RESULTS We identified three distinct groups, associated with different etiologies and unique pathways. Drug inhibitors of identified pathways reduced tumour growth in in vitro and in vivo models. The first group (ESTRO), with mostly fluke-positive CCAs, displayed activation in estrogen signalling and were sensitive to MTOR inhibitors. Another group (OXPHO), with mostly BAP1 and IDH-mutant CCAs, displayed activated oxidative phosphorylation pathways, and were sensitive to oxidative phosphorylation inhibitors. Immune-related pathways were activated in the final group (IMMUN), made up of an immunogenic CCA subtype and CCA with aristolochic acid (AA) mutational signatures. Intratumour differences in AA mutation load were correlated to intratumour variation of different immune cell populations. CONCLUSION Our study elucidates the mechanisms underlying enhancer dysregulation and deepens understanding of different tumourigenesis processes in distinct CCA subtypes, with potential significant therapeutics and clinical benefits.
Collapse
Affiliation(s)
- Jing Han Hong
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore
| | - Chern Han Yong
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- Department of Computer Science, National University of Singapore, Singapore
| | - Hong Lee Heng
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Mai Chan Lau
- Singapore Immunology Network, Agency for Science Technology and Research (A*STAR), Singapore
- Bioinformatics Institute (BII), Agency for Science Technology and Research (A*STAR), Singapore
| | - Jianfeng Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Yi Lee
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
| | - Abner Herbert Lim
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
| | - Zhimei Li
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
| | - Peiyong Guan
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore
| | - Pek Lim Chu
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore
| | - Arnoud Boot
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore
| | - Sheng Rong Ng
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
| | - Xiaosai Yao
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
| | - Felicia Yu Ting Wee
- Institute of Molecular and Cell Biology, Integrative Biology for Theranostics Lab, Agency for Science Technology and Research (A*STAR), Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology, Integrative Biology for Theranostics Lab, Agency for Science Technology and Research (A*STAR), Singapore
| | - Wei Liu
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
| | - Peili Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rong Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xian Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yichen Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Joanna Koh
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
| | - Xiu Yi Kwek
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
| | - Cedric Chuan Young Ng
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
| | - Poramate Klanrit
- Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong
| | - Jiaming Lai
- Department of Pancreaticobiliary Surgery, Sun Yat-sen University, Guangzhou, China
| | - David Wai Meng Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Simona Dima
- Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucuresti, Romania
| | - Irinel Popescu
- Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucuresti, Romania
| | - Sen-Yung Hsieh
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Ming-Chin Yu
- Department of General Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Joe Yeong
- Institute of Molecular and Cell Biology, Integrative Biology for Theranostics Lab, Agency for Science Technology and Research (A*STAR), Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
- Pathology Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Sarinya Kongpetch
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Patrick Tan
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jing Tan
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- State Key Laboratory of Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bin Tean Teh
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
| |
Collapse
|
23
|
Xiang X, Dai Z, Luo B, Zhao N, Liu S, Sui J, Huang J, Zhou Y, Gu J, Zhang J, Yang T, Bao R, Luo Y. Rational Design of a Novel Class of Human ClpP Agonists through a Ring-Opening Strategy with Enhanced Antileukemia Activity. J Med Chem 2024; 67:6769-6792. [PMID: 38620134 DOI: 10.1021/acs.jmedchem.4c00338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The activation of Homo sapiens Casein lysing protease P (HsClpP) by a chemical or genetic strategy has been proved to be a new potential therapy in acute myeloid leukemia (AML). However, limited efficacy has been achieved with classic agonist imipridone ONC201. Here, a novel class of HsClpP agonists is designed and synthesized using a ring-opening strategy based on the lead compound 1 reported in our previous study. Among these novel scaffold agonists, compound 7k exhibited remarkably enhanced proteolytic activity of HsClpP (EC50 = 0.79 ± 0.03 μM) and antitumor activity in vitro (IC50 = 0.038 ± 0.003 μM). Moreover, the intraperitoneal administration of compound 7k markedly suppressed tumor growth in Mv4-11 xenograft models, achieving a tumor growth inhibition rate of 88%. Concurrently, 7k displayed advantageous pharmacokinetic properties in vivo. This study underscores the promise of compound 7k as a significant HsClpP agonist and an antileukemia drug candidate, warranting further exploration for AML treatment.
Collapse
Affiliation(s)
- Xinrong Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baozhu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ninglin Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Song Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Sui
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiasheng Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanzheng Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinlong Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiangnan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Human Diseases and Immunotherapies, West China Hospital and Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui Bao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
24
|
Nachmias B, Aumann S, Haran A, Schimmer AD. Venetoclax resistance in acute myeloid leukaemia-Clinical and biological insights. Br J Haematol 2024; 204:1146-1158. [PMID: 38296617 DOI: 10.1111/bjh.19314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 04/11/2024]
Abstract
Venetoclax, an oral BCL-2 inhibitor, has been widely incorporated in the treatment of acute myeloid leukaemia. The combination of hypomethylating agents and venetoclax is the current standard of care for elderly and patient's ineligible for aggressive therapies. However, venetoclax is being increasingly used with aggressive chemotherapy regimens both in the front line and in the relapse setting. Our growing experience and intensive research demonstrate that certain genetic abnormalities are associated with venetoclax sensitivity, while others with resistance, and that resistance can emerge during treatment leading to disease relapse. In the current review, we provide a summary of the known mechanisms of venetoclax cytotoxicity, both regarding the inhibition of BCL-2-mediated apoptosis and its effect on cell metabolism. We describe how these pathways are linked to venetoclax resistance and are associated with specific mutations. Finally, we provide the rationale for novel drug combinations in current and future clinical trials.
Collapse
Affiliation(s)
- Boaz Nachmias
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shlomzion Aumann
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arnon Haran
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Hurrish KH, Su Y, Patel S, Ramage CL, Zhao J, Temby BR, Carter JL, Edwards H, Buck SA, Wiley SE, Hüttemann M, Polin L, Kushner J, Dzinic SH, White K, Bao X, Li J, Yang J, Boerner J, Hou Z, Al-Atrash G, Konoplev SN, Busquets J, Tiziani S, Matherly LH, Taub JW, Konopleva M, Ge Y, Baran N. Enhancing anti-AML activity of venetoclax by isoflavone ME-344 through suppression of OXPHOS and/or purine biosynthesis in vitro. Biochem Pharmacol 2024; 220:115981. [PMID: 38081370 PMCID: PMC11149698 DOI: 10.1016/j.bcp.2023.115981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/16/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Venetoclax (VEN), in combination with low dose cytarabine (AraC) or a hypomethylating agent, is FDA approved to treat acute myeloid leukemia (AML) in patients who are over the age of 75 or cannot tolerate standard chemotherapy. Despite high response rates to these therapies, most patients succumb to the disease due to relapse and/or drug resistance, providing an unmet clinical need for novel therapies to improve AML patient survival. ME-344 is a potent isoflavone with demonstrated inhibitory activity toward oxidative phosphorylation (OXPHOS) and clinical activity in solid tumors. Given that OXPHOS inhibition enhances VEN antileukemic activity against AML, we hypothesized that ME-344 could enhance the anti-AML activity of VEN. Here we report that ME-344 enhanced VEN to target AML cell lines and primary patient samples while sparing normal hematopoietic cells. Cooperative suppression of OXPHOS was detected in a subset of AML cell lines and primary patient samples. Metabolomics analysis revealed a significant reduction of purine biosynthesis metabolites by ME-344. Further, lometrexol, a purine biosynthesis inhibitor, synergistically enhanced VEN-induced apoptosis in AML cell lines. Interestingly, AML cells with acquired AraC resistance showed significantly increased purine biosynthesis metabolites and sensitivities to ME-344. Furthermore, synergy between ME-344 and VEN was preserved in these AraC-resistant AML cells. In vivo studies revealed significantly prolonged survival upon combination therapy of ME-344 and VEN in NSGS mice bearing parental or AraC-resistant MV4-11 leukemia compared to the vehicle control. This study demonstrates that ME-344 enhances VEN antileukemic activity against preclinical models of AML by suppressing OXPHOS and/or purine biosynthesis.
Collapse
Affiliation(s)
- Katie H Hurrish
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yongwei Su
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shraddha Patel
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cassandra L Ramage
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianlei Zhao
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Brianna R Temby
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Steven A Buck
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA
| | | | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Juiwanna Kushner
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sijana H Dzinic
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kathryn White
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xun Bao
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jay Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Julie Boerner
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhanjun Hou
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sergej N Konoplev
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Jonathan Busquets
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Larry H Matherly
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA; Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Marina Konopleva
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Natalia Baran
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA.
| |
Collapse
|
26
|
Tomar MS, Kumar A, Shrivastava A. Mitochondrial metabolism as a dynamic regulatory hub to malignant transformation and anti-cancer drug resistance. Biochem Biophys Res Commun 2024; 694:149382. [PMID: 38128382 DOI: 10.1016/j.bbrc.2023.149382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Glycolysis is the fundamental cellular process that permits cancer cells to convert energy and grow anaerobically. Recent developments in molecular biology have made it evident that mitochondrial respiration is critical to tumor growth and treatment response. As the principal organelle of cellular energy conversion, mitochondria can rapidly alter cellular metabolic processes, thereby fueling malignancies and contributing to treatment resistance. This review emphasizes the significance of mitochondrial biogenesis, turnover, DNA copy number, and mutations in bioenergetic system regulation. Tumorigenesis requires an intricate cascade of metabolic pathways that includes rewiring of the tricarboxylic acid (TCA) cycle, electron transport chain and oxidative phosphorylation, supply of intermediate metabolites of the TCA cycle through amino acids, and the interaction between mitochondria and lipid metabolism. Cancer recurrence or resistance to therapy often results from the cooperation of several cellular defense mechanisms, most of which are connected to mitochondria. Many clinical trials are underway to assess the effectiveness of inhibiting mitochondrial respiration as a potential cancer therapeutic. We aim to summarize innovative strategies and therapeutic targets by conducting a comprehensive review of recent studies on the relationship between mitochondrial metabolism, tumor development and therapeutic resistance.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, 462020, Madhya Pradesh, India
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India.
| |
Collapse
|
27
|
Rodríguez-Medina C, Stuckey R, Bilbao-Sieyro C, Gómez-Casares MT. Biomarkers of Response to Venetoclax Therapy in Acute Myeloid Leukemia. Int J Mol Sci 2024; 25:1421. [PMID: 38338698 PMCID: PMC10855565 DOI: 10.3390/ijms25031421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Recent progress in the use of massive sequencing technologies has greatly enhanced our understanding of acute myeloid leukemia (AML) pathology. This knowledge has in turn driven the development of targeted therapies, such as venetoclax, a BCL-2 inhibitor approved for use in combination with azacitidine, decitabine, or low-dose cytarabine for the treatment of newly diagnosed adult patients with AML who are not eligible for intensive chemotherapy. However, a significant number of AML patients still face the challenge of disease relapse. In this review, we will explore biomarkers that may predict disease progression in patients receiving venetoclax-based therapy, considering both clinical factors and genetic changes. Despite the many advances, we conclude that the identification of molecular profiles for AML patients who will respond optimally to venetoclax therapy remains an unmet clinical need.
Collapse
Affiliation(s)
- Carlos Rodríguez-Medina
- Hematology Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35019 Las Palmas de Gran Canaria, Spain; (C.R.-M.); (R.S.); (C.B.-S.)
| | - Ruth Stuckey
- Hematology Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35019 Las Palmas de Gran Canaria, Spain; (C.R.-M.); (R.S.); (C.B.-S.)
| | - Cristina Bilbao-Sieyro
- Hematology Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35019 Las Palmas de Gran Canaria, Spain; (C.R.-M.); (R.S.); (C.B.-S.)
- Morphology Department, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
| | - María Teresa Gómez-Casares
- Hematology Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35019 Las Palmas de Gran Canaria, Spain; (C.R.-M.); (R.S.); (C.B.-S.)
- Department of Medical Sciences, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
| |
Collapse
|
28
|
Forsberg M, Konopleva M. SOHO State of the Art Updates and Next Questions: Understanding and Overcoming Venetoclax Resistance in Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:1-14. [PMID: 38007372 DOI: 10.1016/j.clml.2023.10.006] [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: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/27/2023]
Abstract
The discovery of Venetoclax (VEN) has transformed the therapeutic landscape of acute myeloid leukemia (AML) and chronic lymphocytic leukemia (CLL). However, the response is heterogeneous with 10% to 50% of newly diagnosed AML patients not responding to hypomethylating agent (HMA) and VEN. Furthermore, up to 40% of responding patients relapse shortly. This review discusses the mechanism of action of Venetoclax and the major mechanisms of inherent and acquired resistance to VEN. VEN is highly specific to BCL-2 binding, as such other antiapoptotic proteins in BCL-2 family induce resistance. These antiapoptotic proteins can also be upregulated via a number of compensatory cell signaling pathways including PI3K/AKT/mTOR, the MAPK/ERK pathway, and mutant FLT3-ITD. Mutations can occur in BCL-2 and BAX proteins, or they can be silenced by TP53 mutations and other epigenetic changes. Changes to mitochondrial structure and metabolism can induce resistance. Key metabolic regulators include OXPHOS and alternative amino acid metabolism. Finally microenvironmental factors can influence VEN responses. This paper evaluates subsets of AML by differentiation, histology, cytogenetics and molecular markers and their different responses to VEN; with spliceosome mutations, ASXL1, NPM1 and IDH1/2 being favorable while others such as FLT3, TP53 and BCL-2 mutations being less responsive. Currently intensive multiagent chemotherapy and Venetoclax combinations such as 7+3+VEN are favored in fit younger AML patients. However, with resistant patients' subsets targeted combination therapies are becoming an increasingly attractive option. We explore the incorporation of non-BCL-2 inhibitors, next-generation BCL-2 and multi-protein agents, other inhibitors most prominently FLT-3 inhibitors in addition to Venetoclax, and other novel approaches for resolving Venetoclax resistance.
Collapse
Affiliation(s)
- Mark Forsberg
- Department of Oncology, Montefiore Einstein Cancer Center, Bronx, NY
| | - Marina Konopleva
- Department of Oncology, Montefiore Einstein Cancer Center, Bronx, NY.
| |
Collapse
|
29
|
Morishima T, Takahashi K, Chin DWL, Wang Y, Tokunaga K, Arima Y, Matsuoka M, Suda T, Takizawa H. Phospholipid metabolic adaptation promotes survival of IDH2 mutant acute myeloid leukemia cells. Cancer Sci 2024; 115:197-210. [PMID: 37882467 PMCID: PMC10823289 DOI: 10.1111/cas.15994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023] Open
Abstract
Genetic mutations in the isocitrate dehydrogenase (IDH) gene that result in a pathological enzymatic activity to produce oncometabolite have been detected in acute myeloid leukemia (AML) patients. While specific inhibitors that target mutant IDH enzymes and normalize intracellular oncometabolite level have been developed, refractoriness and resistance has been reported. Since acquisition of pathological enzymatic activity is accompanied by the abrogation of the crucial WT IDH enzymatic activity in IDH mutant cells, aberrant metabolism in IDH mutant cells can potentially persist even after the normalization of intracellular oncometabolite level. Comparisons of isogenic AML cell lines with and without IDH2 gene mutations revealed two mutually exclusive signalings for growth advantage of IDH2 mutant cells, STAT phosphorylation associated with intracellular oncometabolite level and phospholipid metabolic adaptation. The latter came to light after the oncometabolite normalization and increased the resistance of IDH2 mutant cells to arachidonic acid-mediated apoptosis. The release of this metabolic adaptation by FDA-approved anti-inflammatory drugs targeting the metabolism of arachidonic acid could sensitize IDH2 mutant cells to apoptosis, resulting in their eradication in vitro and in vivo. Our findings will contribute to the development of alternative therapeutic options for IDH2 mutant AML patients who do not tolerate currently available therapies.
Collapse
Affiliation(s)
- Tatsuya Morishima
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Laboratory of Hematopoietic Stem Cell Engineering, IRCMSKumamoto UniversityKumamotoJapan
| | - Koichi Takahashi
- Departments of Leukemia and Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Desmond Wai Loon Chin
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Yuxin Wang
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Department of Hematology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Kenji Tokunaga
- Department of Hematology, Rheumatology, and Infectious Diseases, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yuichiro Arima
- Laboratory of Developmental Cardiology, IRCMSKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA)Kumamoto UniversityKumamotoJapan
| | - Masao Matsuoka
- Department of Hematology, Rheumatology, and Infectious Diseases, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Toshio Suda
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
- Laboratory of Stem Cell Regulation, IRCMSKumamoto UniversityKumamotoJapan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA)Kumamoto UniversityKumamotoJapan
| |
Collapse
|
30
|
Landberg N, Köhnke T, Feng Y, Nakauchi Y, Fan AC, Linde MH, Karigane D, Lim K, Sinha R, Malcovati L, Thomas D, Majeti R. IDH1-mutant preleukemic hematopoietic stem cells can be eliminated by inhibition of oxidative phosphorylation. Blood Cancer Discov 2023; 5:731701. [PMID: 38091010 PMCID: PMC10905513 DOI: 10.1158/2643-3230.bcd-23-0195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/03/2023] [Accepted: 12/06/2023] [Indexed: 01/25/2024] Open
Abstract
Rare preleukemic hematopoietic stem cells (pHSCs) harboring only the initiating mutations can be detected at the time of AML diagnosis. pHSCs are the origin of leukemia and a potential reservoir for relapse. Using primary human samples and gene-editing to model isocitrate dehydrogenase 1 (IDH1) mutant pHSCs, we show epigenetic, transcriptional, and metabolic differences between pHSCs and healthy hematopoietic stem cells (HSCs). We confirm that IDH1 driven clonal hematopoiesis is associated with cytopenia, suggesting an inherent defect to fully reconstitute hematopoiesis. Despite giving rise to multilineage engraftment, IDH1-mutant pHSCs exhibited reduced proliferation, blocked differentiation, downregulation of MHC Class II genes, and reprogramming of oxidative phosphorylation metabolism. Critically, inhibition of oxidative phosphorylation resulted in complete eradication of IDH1-mutant pHSCs but not IDH2-mutant pHSCs or wildtype HSCs. Our results indicate that IDH1-mutant preleukemic clones can be targeted with complex I inhibitors, offering a potential strategy to prevent development and relapse of leukemia.
Collapse
Affiliation(s)
- Niklas Landberg
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Thomas Köhnke
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Yang Feng
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Yusuke Nakauchi
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Amy C. Fan
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
- Immunology Graduate Program, Stanford University, Stanford, California
| | - Miles H. Linde
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
- Immunology Graduate Program, Stanford University, Stanford, California
| | - Daiki Karigane
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Kelly Lim
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Precision Medicine, South Australian Health and Medical Research Institute, The University of Adelaide, Adelaide, Australia
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Daniel Thomas
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Precision Medicine, South Australian Health and Medical Research Institute, The University of Adelaide, Adelaide, Australia
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| |
Collapse
|
31
|
Zhang YW, Schönberger K, Cabezas‐Wallscheid N. Bidirectional interplay between metabolism and epigenetics in hematopoietic stem cells and leukemia. EMBO J 2023; 42:e112348. [PMID: 38010205 PMCID: PMC10711668 DOI: 10.15252/embj.2022112348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 11/29/2023] Open
Abstract
During the last decades, remarkable progress has been made in further understanding the complex molecular regulatory networks that maintain hematopoietic stem cell (HSC) function. Cellular and organismal metabolisms have been shown to directly instruct epigenetic alterations, and thereby dictate stem cell fate, in the bone marrow. Epigenetic regulatory enzymes are dependent on the availability of metabolites to facilitate DNA- and histone-modifying reactions. The metabolic and epigenetic features of HSCs and their downstream progenitors can be significantly altered by environmental perturbations, dietary habits, and hematological diseases. Therefore, understanding metabolic and epigenetic mechanisms that regulate healthy HSCs can contribute to the discovery of novel metabolic therapeutic targets that specifically eliminate leukemia stem cells while sparing healthy HSCs. Here, we provide an in-depth review of the metabolic and epigenetic interplay regulating hematopoietic stem cell fate. We discuss the influence of metabolic stress stimuli, as well as alterations occurring during leukemic development. Additionally, we highlight recent therapeutic advancements toward eradicating acute myeloid leukemia cells by intervening in metabolic and epigenetic pathways.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | | | | |
Collapse
|
32
|
Zhang H, Sun C, Sun Q, Li Y, Zhou C, Sun C. Susceptibility of acute myeloid leukemia cells to ferroptosis and evasion strategies. Front Mol Biosci 2023; 10:1275774. [PMID: 37818101 PMCID: PMC10561097 DOI: 10.3389/fmolb.2023.1275774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/15/2023] [Indexed: 10/12/2023] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematologic malignancy with a 5-year survival rate of less than 30%. Continuous updating of diagnostic and therapeutic strategies has not been effective in improving the clinical benefit of AML. AML cells are prone to iron metabolism imbalance due to their unique pathological characteristics, and ferroptosis is a novel cell death mode that is dominated by three cellular biological processes: iron metabolism, oxidative stress and lipid metabolism. An in-depth exploration of the unique ferroptosis mechanism in AML can provide new insights for the diagnosis and treatment of this disease. This study summarizes recent studies on ferroptosis in AML cells and suggests that the metabolic characteristics, gene mutation patterns, and dependence on mitochondria of AML cells greatly increase their susceptibility to ferroptosis. In addition, this study suggests that AML cells can establish a variety of strategies to evade ferroptosis to maintain their survival during the process of occurrence and development, and summarizes the related drugs targeting ferroptosis pathway in AML treatment, which provides development directions for the subsequent mechanism research and clinical treatment of AML.
Collapse
Affiliation(s)
- Hanyun Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunjie Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ye Li
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chao Zhou
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| |
Collapse
|
33
|
Turtoi E, Jeudy J, Henry S, Dadi I, Valette G, Enjalbal C, Turtoi A. Analysis of polar primary metabolites in biological samples using targeted metabolomics and LC-MS. STAR Protoc 2023; 4:102400. [PMID: 37590149 PMCID: PMC10440349 DOI: 10.1016/j.xpro.2023.102400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/17/2023] [Accepted: 06/01/2023] [Indexed: 08/19/2023] Open
Abstract
Primary metabolites are molecules of essential biochemical reactions that define the biological phenotype. All primary metabolites cannot be measured in a single analysis. In this protocol, we outline the multiplexed and quantitative measurement of 106 metabolites that cover the central part of primary metabolism. The protocol includes several sample preparation techniques and one liquid chromatography-mass spectrometry method. Then, we describe the steps of the bioinformatic data analysis to better understand the metabolic perturbations that may occur in a biological system. For complete details on the use and execution of this protocol, please refer to: Costanza et al.,1 Blomme et al.,2 Blomme et al.,3 Guillon et al.,4 Stuani et al.5.
Collapse
Affiliation(s)
- Evgenia Turtoi
- Platform for Translational Oncometabolomics, Biocampus, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | | | - Sylvain Henry
- Platform for Translational Oncometabolomics, Biocampus, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Ikrame Dadi
- Tumor Microenvironment and Resistance to Therapy Laboratory, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM U1194, Montpellier, France
| | - Gilles Valette
- Laboratoire de Mesures Physiques, Université de Montpellier, Montpellier, France
| | - Christine Enjalbal
- Laboratoire de Mesures Physiques, Université de Montpellier, Montpellier, France
| | - Andrei Turtoi
- Platform for Translational Oncometabolomics, Biocampus, CNRS, INSERM, Université de Montpellier, Montpellier, France; Tumor Microenvironment and Resistance to Therapy Laboratory, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM U1194, Montpellier, France; Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan.
| |
Collapse
|
34
|
Feng L, Zhang PY, Gao W, Yu J, Robson SC. Targeting chemoresistance and mitochondria-dependent metabolic reprogramming in acute myeloid leukemia. Front Oncol 2023; 13:1244280. [PMID: 37746249 PMCID: PMC10513429 DOI: 10.3389/fonc.2023.1244280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Chemoresistance often complicates the management of cancer, as noted in the instance of acute myeloid leukemia (AML). Mitochondrial function is considered important for the viability of AML blasts and appears to also modulate chemoresistance. As mitochondrial metabolism is aberrant in AML, any distinct pathways could be directly targeted to impact both cell viability and chemoresistance. Therefore, identifying and targeting those precise rogue elements of mitochondrial metabolism could be a valid therapeutic strategy in leukemia. Here, we review the evidence for abnormalities in mitochondria metabolic processes in AML cells, that likely impact chemoresistance. We further address several therapeutic approaches targeting isocitrate dehydrogenase 2 (IDH2), CD39, nicotinamide phosphoribosyl transferase (NAMPT), electron transport chain (ETC) complex in AML and also consider the roles of mesenchymal stromal cells. We propose the term "mitotherapy" to collectively refer to such regimens that attempt to override mitochondria-mediated metabolic reprogramming, as used by cancer cells. Mounting evidence suggests that mitotherapy could provide a complementary strategy to overcome chemoresistance in liquid cancers, as well as in solid tumors.
Collapse
Affiliation(s)
- Lili Feng
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Philip Y. Zhang
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Wenda Gao
- Antagen Institute for Biomedical Research, Canton, MA, United States
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Simon C. Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Department of Medicine, Division of Gastroenterology/Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
35
|
Bruzzese A, Labanca C, Martino EA, Mendicino F, Lucia E, Olivito V, Neri A, Imovilli A, Morabito F, Vigna E, Gentile M. Ivosidenib in acute myeloid leukemia. Expert Opin Pharmacother 2023; 24:2093-2100. [PMID: 37874005 DOI: 10.1080/14656566.2023.2272659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION Traditional treatment strategies for acute myeloid leukemia (AML) have primarily relied on standard chemotherapy regimens for four decades. Indeed, the landscape of AML therapy has evolved substantially in recent years, mainly due to the introduction of hypomethylating agents and small molecules.Bcl2 inhibitor venetoclax, Fms-like tyrosine kinase 3 (FLT3) inhibitors such as midostaurin and gilteritinib, and isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) inhibitors ivosidenib and enasidenib, as well as hedgehog (HH) pathway inhibitor glasdegib represented a significant step forward in AML therapeutic armamentarium. Smoothened (SMO) inhibitor in combination with low-dose cytarabine marks a recent milestone. AREAS COVERED Ivosidenib, the first-in-class, selective, allosteric IDH1R132 inhibitor, showed the capability to induce in vitro differentiation of primary mIDH1 AML blasts. Clinical data highlighted its exceptional safety profile, as a standalone therapy and in combination strategy. Additionally, comprehensive studies consistently demonstrated its effectiveness, both in monotherapy and in association with chemotherapy. EXPERT OPINION The identified ivosidenib's strengths, including its remarkable safety record and ability to yield positive therapeutic outcomes, position it as an ideal partner for both classic chemotherapy and biological treatments, i.e. hypometilant agents and/or venetoclax. Further studies are warranted to explore strategies for overcoming the occurrence of ivosidenib resistance.
Collapse
Affiliation(s)
- Antonella Bruzzese
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Caterina Labanca
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Enrica Antonia Martino
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Francesco Mendicino
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Eugenio Lucia
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Virginia Olivito
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Antonino Neri
- Scientific Directorate IRCCS di Reggio Emilia, EmiliaRomagna, Reggio Emilia, Italy
| | - Annalisa Imovilli
- Department of Hematology, Azienda USL-IRCCS di Reggio Emilia, Emilia Romagna, Reggio Emilia, Italy
| | | | - Ernesto Vigna
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Massimo Gentile
- Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
36
|
Tangella AV, Gajre A, Kantheti VV. Isocitrate Dehydrogenase 1 Mutation and Ivosidenib in Patients With Acute Myeloid Leukemia: A Comprehensive Review. Cureus 2023; 15:e44802. [PMID: 37692182 PMCID: PMC10483130 DOI: 10.7759/cureus.44802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2023] [Indexed: 09/12/2023] Open
Abstract
Acute myeloid leukemia (AML) arises from immature myeloid progenitors, resulting in a stem-cell-like proliferative state. This leads to excessive pools of immature cells that cannot function, which usually happens at the cost of the production of mature functional cells, leading to deleterious consequences. The management of AML has intensified as newer targeted therapies have come into existence owing to deeper genetic analysis of the disease and patients. Isocitrate dehydrogenase (IDH) is a cytosolic enzyme that is a part of the Krebs cycle and is extremely important in maintaining the homeostasis of the cell. It is produced by two different genes: IDH1 and IDH2. Ivosidenib has been associated with IDH1 inhibition and has been studied in numerous cancers. This review highlights the studies that have dealt with ivosidenib, an IDH1 inhibitor, in AML, the side effect profile, and the possible future course of the drug. After a scoping review of the available literature, we have identified that studies have consistently shown positive outcomes and that ivosidenib is a promising avenue for the management of AML. But it also has to be kept in mind that resistance to IDH inhibitors is on the rise, and the need to identify ways to circumvent this is to be addressed.
Collapse
Affiliation(s)
| | - Ashwin Gajre
- Internal Medicine, Lokmanya Tilak Municipal Medical College, Mumbai, IND
| | | |
Collapse
|
37
|
Behnam B, Taghizadeh-Hesary F. Mitochondrial Metabolism: A New Dimension of Personalized Oncology. Cancers (Basel) 2023; 15:4058. [PMID: 37627086 PMCID: PMC10452105 DOI: 10.3390/cancers15164058] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Energy is needed by cancer cells to stay alive and communicate with their surroundings. The primary organelles for cellular metabolism and energy synthesis are mitochondria. Researchers recently proved that cancer cells can steal immune cells' mitochondria using nanoscale tubes. This finding demonstrates the dependence of cancer cells on normal cells for their living and function. It also denotes the importance of mitochondria in cancer cells' biology. Emerging evidence has demonstrated how mitochondria are essential for cancer cells to survive in the harsh tumor microenvironments, evade the immune system, obtain more aggressive features, and resist treatments. For instance, functional mitochondria can improve cancer resistance against radiotherapy by scavenging the released reactive oxygen species. Therefore, targeting mitochondria can potentially enhance oncological outcomes, according to this notion. The tumors' responses to anticancer treatments vary, ranging from a complete response to even cancer progression during treatment. Therefore, personalized cancer treatment is of crucial importance. So far, personalized cancer treatment has been based on genomic analysis. Evidence shows that tumors with high mitochondrial content are more resistant to treatment. This paper illustrates how mitochondrial metabolism can participate in cancer resistance to chemotherapy, immunotherapy, and radiotherapy. Pretreatment evaluation of mitochondrial metabolism can provide additional information to genomic analysis and can help to improve personalized oncological treatments. This article outlines the importance of mitochondrial metabolism in cancer biology and personalized treatments.
Collapse
Affiliation(s)
- Babak Behnam
- Department of Regulatory Affairs, Amarex Clinical Research, NSF International, Germantown, MD 20874, USA
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 1445613131, Iran
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran 1445613131, Iran
| |
Collapse
|
38
|
Taghizadeh-Hesary F, Houshyari M, Farhadi M. Mitochondrial metabolism: a predictive biomarker of radiotherapy efficacy and toxicity. J Cancer Res Clin Oncol 2023; 149:6719-6741. [PMID: 36719474 DOI: 10.1007/s00432-023-04592-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Radiotherapy is a mainstay of cancer treatment. Clinical studies revealed a heterogenous response to radiotherapy, from a complete response to even disease progression. To that end, finding the relative prognostic factors of disease outcomes and predictive factors of treatment efficacy and toxicity is essential. It has been demonstrated that radiation response depends on DNA damage response, cell cycle phase, oxygen concentration, and growth rate. Emerging evidence suggests that altered mitochondrial metabolism is associated with radioresistance. METHODS This article provides a comprehensive evaluation of the role of mitochondria in radiotherapy efficacy and toxicity. In addition, it demonstrates how mitochondria might be involved in the famous 6Rs of radiobiology. RESULTS In terms of this idea, decreasing the mitochondrial metabolism of cancer cells may increase radiation response, and enhancing the mitochondrial metabolism of normal cells may reduce radiation toxicity. Enhancing the normal cells (including immune cells) mitochondrial metabolism can potentially improve the tumor response by enhancing immune reactivation. Future studies are invited to examine the impacts of mitochondrial metabolism on radiation efficacy and toxicity. Improving radiotherapy response with diminishing cancer cells' mitochondrial metabolism, and reducing radiotherapy toxicity with enhancing normal cells' mitochondrial metabolism.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Clinical Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Houshyari
- Clinical Oncology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Arévalo CM, Cruz-Rodriguez N, Quijano S, Fiorentino S. Plant-derived extracts and metabolic modulation in leukemia: a promising approach to overcome treatment resistance. Front Mol Biosci 2023; 10:1229760. [PMID: 37520325 PMCID: PMC10382028 DOI: 10.3389/fmolb.2023.1229760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Leukemic cells acquire complex and often multifactorial mechanisms of resistance to treatment, including various metabolic alterations. Although the use of metabolic modulators has been proposed for several decades, their use in clinical practice has not been established. Natural products, the so-called botanical drugs, are capable of regulating tumor metabolism, particularly in hematopoietic tumors, which could partly explain the biological activity attributed to them for a long time. This review addresses the most recent findings relating to metabolic reprogramming-Mainly in the glycolytic pathway and mitochondrial activity-Of leukemic cells and its role in the generation of resistance to conventional treatments, the modulation of the tumor microenvironment, and the evasion of immune response. In turn, it describes how the modulation of metabolism by plant-derived extracts can counteract resistance to chemotherapy in this tumor model and contribute to the activation of the antitumor immune system.
Collapse
Affiliation(s)
- Cindy Mayerli Arévalo
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
40
|
Sabatier M, Birsen R, Lauture L, Mouche S, Angelino P, Dehairs J, Goupille L, Boussaid I, Heiblig M, Boet E, Sahal A, Saland E, Santos JC, Armengol M, Fernández-Serrano M, Farge T, Cognet G, Simonetta F, Pignon C, Graffeuil A, Mazzotti C, Avet-Loiseau H, Delos O, Bertrand-Michel J, Chedru A, Dembitz V, Gallipoli P, Anstee NS, Loo S, Wei AH, Carroll M, Goubard A, Castellano R, Collette Y, Vergez F, Mansat-De Mas V, Bertoli S, Tavitian S, Picard M, Récher C, Bourges-Abella N, Granat F, Kosmider O, Sujobert P, Colsch B, Joffre C, Stuani L, Swinnen JV, Guillou H, Roué G, Hakim N, Dejean AS, Tsantoulis P, Larrue C, Bouscary D, Tamburini J, Sarry JE. C/EBPα Confers Dependence to Fatty Acid Anabolic Pathways and Vulnerability to Lipid Oxidative Stress-Induced Ferroptosis in FLT3-Mutant Leukemia. Cancer Discov 2023; 13:1720-1747. [PMID: 37012202 DOI: 10.1158/2159-8290.cd-22-0411] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 01/19/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Although transcription factor CCAAT-enhancer binding protein α (C/EBPα) is critical for normal and leukemic differentiation, its role in cell and metabolic homeostasis is largely unknown in cancer. Here, multiomics analyses uncovered a coordinated activation of C/EBPα and Fms-like tyrosine kinase 3 (FLT3) that increased lipid anabolism in vivo and in patients with FLT3-mutant acute myeloid leukemia (AML). Mechanistically, C/EBPα regulated the fatty acid synthase (FASN)-stearoyl-CoA desaturase (SCD) axis to promote fatty acid (FA) biosynthesis and desaturation. We further demonstrated that FLT3 or C/EBPα inactivation decreased monounsaturated FA incorporation to membrane phospholipids through SCD downregulation. Consequently, SCD inhibition enhanced susceptibility to lipid redox stress that was exploited by combining FLT3 and glutathione peroxidase 4 inhibition to trigger lipid oxidative stress, enhancing ferroptotic death of FLT3-mutant AML cells. Altogether, our study reveals a C/EBPα function in lipid homeostasis and adaptation to redox stress, and a previously unreported vulnerability of FLT3-mutant AML to ferroptosis with promising therapeutic application. SIGNIFICANCE FLT3 mutations are found in 30% of AML cases and are actionable by tyrosine kinase inhibitors. Here, we discovered that C/EBPα regulates FA biosynthesis and protection from lipid redox stress downstream mutant-FLT3 signaling, which confers a vulnerability to ferroptosis upon FLT3 inhibition with therapeutic potential in AML. This article is highlighted in the In This Issue feature, p. 1501.
Collapse
Affiliation(s)
- Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Rudy Birsen
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, Paris, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| | - Laura Lauture
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Sarah Mouche
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
| | - Paolo Angelino
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, LKI-Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Léa Goupille
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Ismael Boussaid
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, Paris, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Maël Heiblig
- Hospices Civils de Lyon, Hôpital Lyon Sud, Lyon, France
- CIRI, Inserm U1111 CNRS 5308, Université Lyon 1, Lyon, France
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Juliana C Santos
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Marc Armengol
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | | | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Guillaume Cognet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Federico Simonetta
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
| | - Corentin Pignon
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Antoine Graffeuil
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Céline Mazzotti
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Hervé Avet-Loiseau
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Océane Delos
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, University Paul Sabatier, Toulouse, France
| | - Justine Bertrand-Michel
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, University Paul Sabatier, Toulouse, France
| | - Amélie Chedru
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, MetaboHUB, Gif sur Yvette, France
| | - Vilma Dembitz
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Paolo Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Natasha S Anstee
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Sun Loo
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | - Andrew H Wei
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | - Martin Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Armelle Goubard
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Rémy Castellano
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Yves Collette
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - François Vergez
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Véronique Mansat-De Mas
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Sarah Bertoli
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Suzanne Tavitian
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | - Muriel Picard
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service de Réanimation, Toulouse, France
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, Toulouse, France
| | | | - Fanny Granat
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Olivier Kosmider
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, Paris, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Pierre Sujobert
- Hospices Civils de Lyon, Hôpital Lyon Sud, Lyon, France
- CIRI, Inserm U1111 CNRS 5308, Université Lyon 1, Lyon, France
| | - Benoit Colsch
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, MetaboHUB, Gif sur Yvette, France
| | - Carine Joffre
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Lucille Stuani
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, LKI-Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Hervé Guillou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, University Paul Sabatier, Toulouse, France
| | - Gael Roué
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Nawad Hakim
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITy), Inserm UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse, France
| | - Anne S Dejean
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITy), Inserm UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse, France
| | - Petros Tsantoulis
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Clément Larrue
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
| | - Didier Bouscary
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, Paris, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| | - Jerome Tamburini
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, Paris, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, Toulouse, France
- LabEx Toucan, Toulouse, France
- Équipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| |
Collapse
|
41
|
Wang J, Tomlinson B, Lazarus HM. Update on Small Molecule Targeted Therapies for Acute Myeloid Leukemia. Curr Treat Options Oncol 2023; 24:770-801. [PMID: 37195589 DOI: 10.1007/s11864-023-01090-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/18/2023]
Abstract
OPINION STATEMENT The search for effective therapies for the highly heterogenous disease acute myeloid leukemia (AML) has remained elusive. While cytotoxic therapies can induce complete remission and even, at times, long-term survival, this approach is associated with significant toxic effects to visceral organs and worsening of immune dysfunction and marrow suppression leading to death. Sophisticated molecular studies have revealed defects within the AML cell that can be exploited by utilizing small molecule agents to target these defects, often dubbed "target therapy." Several medications have already established new standards of care for many patients with AML, including FDA-approved agents that inhibitor IDH1, IDH2, FLT3, and BCL-2. Emerging small molecules hold additional to add to the armamentarium of AML treatment options including MCL-1 inhibitors, TP53 inhibitors, menin inhibitors, and E-selectin antagonists. Moreover, the increasing options also mean that future combinations of these agents need to be explored, including with cytotoxic drugs and other newer emerging strategies such as immunotherapies for AML. Recent investigations continue to show that overcoming many of the challenges of treating AML finally is on the horizon.
Collapse
Affiliation(s)
- Jiasheng Wang
- Division of Hematology, Department of Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11000 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Benjamin Tomlinson
- Division of Hematology, Department of Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11000 Euclid Avenue, Cleveland, OH, 44106, USA.
| | - Hillard M Lazarus
- Division of Hematology, Department of Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11000 Euclid Avenue, Cleveland, OH, 44106, USA
| |
Collapse
|
42
|
Saulle E, Spinello I, Quaranta MT, Labbaye C. Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting. Cells 2023; 12:1553. [PMID: 37296673 PMCID: PMC10252746 DOI: 10.3390/cells12111553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Autophagy is a highly conserved cellular degradation process that regulates cellular metabolism and homeostasis under normal and pathophysiological conditions. Autophagy and metabolism are linked in the hematopoietic system, playing a fundamental role in the self-renewal, survival, and differentiation of hematopoietic stem and progenitor cells, and in cell death, particularly affecting the cellular fate of the hematopoietic stem cell pool. In leukemia, autophagy sustains leukemic cell growth, contributes to survival of leukemic stem cells and chemotherapy resistance. The high frequency of disease relapse caused by relapse-initiating leukemic cells resistant to therapy occurs in acute myeloid leukemia (AML), and depends on the AML subtypes and treatments used. Targeting autophagy may represent a promising strategy to overcome therapeutic resistance in AML, for which prognosis remains poor. In this review, we illustrate the role of autophagy and the impact of its deregulation on the metabolism of normal and leukemic hematopoietic cells. We report updates on the contribution of autophagy to AML development and relapse, and the latest evidence indicating autophagy-related genes as potential prognostic predictors and drivers of AML. We review the recent advances in autophagy manipulation, combined with various anti-leukemia therapies, for an effective autophagy-targeted therapy for AML.
Collapse
Affiliation(s)
- Ernestina Saulle
- Correspondence: (E.S.); (C.L.); Tel.: +39-0649902422 (E.S.); +39-0649902418 (C.L.)
| | | | | | - Catherine Labbaye
- Correspondence: (E.S.); (C.L.); Tel.: +39-0649902422 (E.S.); +39-0649902418 (C.L.)
| |
Collapse
|
43
|
Proteogenomic analysis of acute myeloid leukemia associates relapsed disease with reprogrammed energy metabolism both in adults and children. Leukemia 2023; 37:550-559. [PMID: 36572751 PMCID: PMC9991901 DOI: 10.1038/s41375-022-01796-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/27/2022]
Abstract
Despite improvement of current treatment strategies and novel targeted drugs, relapse and treatment resistance largely determine the outcome for acute myeloid leukemia (AML) patients. To identify the underlying molecular characteristics, numerous studies have been aimed to decipher the genomic- and transcriptomic landscape of AML. Nevertheless, further molecular changes allowing malignant cells to escape treatment remain to be elucidated. Mass spectrometry is a powerful tool enabling detailed insights into proteomic changes that could explain AML relapse and resistance. Here, we investigated AML samples from 47 adult and 22 pediatric patients at serial time-points during disease progression using mass spectrometry-based in-depth proteomics. We show that the proteomic profile at relapse is enriched for mitochondrial ribosomal proteins and subunits of the respiratory chain complex, indicative of reprogrammed energy metabolism from diagnosis to relapse. Further, higher levels of granzymes and lower levels of the anti-inflammatory protein CR1/CD35 suggest an inflammatory signature promoting disease progression. Finally, through a proteogenomic approach, we detected novel peptides, which present a promising repertoire in the search for biomarkers and tumor-specific druggable targets. Altogether, this study highlights the importance of proteomic studies in holistic approaches to improve treatment and survival of AML patients.
Collapse
|
44
|
Thomas D, Wu M, Nakauchi Y, Zheng M, Thompson-Peach CA, Lim K, Landberg N, Köhnke T, Robinson N, Kaur S, Kutyna M, Stafford M, Hiwase D, Reinisch A, Peltz G, Majeti R. Dysregulated Lipid Synthesis by Oncogenic IDH1 Mutation Is a Targetable Synthetic Lethal Vulnerability. Cancer Discov 2023; 13:496-515. [PMID: 36355448 PMCID: PMC9900324 DOI: 10.1158/2159-8290.cd-21-0218] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/18/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022]
Abstract
Isocitrate dehydrogenase 1 and 2 (IDH) are mutated in multiple cancers and drive production of (R)-2-hydroxyglutarate (2HG). We identified a lipid synthesis enzyme [acetyl CoA carboxylase 1 (ACC1)] as a synthetic lethal target in mutant IDH1 (mIDH1), but not mIDH2, cancers. Here, we analyzed the metabolome of primary acute myeloid leukemia (AML) blasts and identified an mIDH1-specific reduction in fatty acids. mIDH1 also induced a switch to b-oxidation indicating reprogramming of metabolism toward a reliance on fatty acids. Compared with mIDH2, mIDH1 AML displayed depletion of NADPH with defective reductive carboxylation that was not rescued by the mIDH1-specific inhibitor ivosidenib. In xenograft models, a lipid-free diet markedly slowed the growth of mIDH1 AML, but not healthy CD34+ hematopoietic stem/progenitor cells or mIDH2 AML. Genetic and pharmacologic targeting of ACC1 resulted in the growth inhibition of mIDH1 cancers not reversible by ivosidenib. Critically, the pharmacologic targeting of ACC1 improved the sensitivity of mIDH1 AML to venetoclax. SIGNIFICANCE Oncogenic mutations in both IDH1 and IDH2 produce 2-hydroxyglutarate and are generally considered equivalent in terms of pathogenesis and targeting. Using comprehensive metabolomic analysis, we demonstrate unexpected metabolic differences in fatty acid metabolism between mutant IDH1 and IDH2 in patient samples with targetable metabolic interventions. See related commentary by Robinson and Levine, p. 266. This article is highlighted in the In This Issue feature, p. 247.
Collapse
Affiliation(s)
- Daniel Thomas
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
- Adelaide Medical School, University of Adelaide, South Australia and Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Manhong Wu
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Yusuke Nakauchi
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Ming Zheng
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Chloe A.L. Thompson-Peach
- Adelaide Medical School, University of Adelaide, South Australia and Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Kelly Lim
- Adelaide Medical School, University of Adelaide, South Australia and Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Niklas Landberg
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Thomas Köhnke
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Nirmal Robinson
- Centre for Cancer Biology, University of South Australia, South Australia, Australia
| | - Satinder Kaur
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Monika Kutyna
- Adelaide Medical School, University of Adelaide, South Australia and Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Melissa Stafford
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Devendra Hiwase
- Adelaide Medical School, University of Adelaide, South Australia and Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andreas Reinisch
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
- Division of Hematology and Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Gary Peltz
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, California
- Corresponding Author: Ravindra Majeti, Department of Medicine, Division of Hematology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Lokey Stem Cell Building, 265 Campus Drive, Stanford, CA 94305. Phone: 650-721-6376; Fax: 650-736-2961; E-mail:
| |
Collapse
|
45
|
Huang Z, Shen Y, Liu W, Yang Y, Guo L, Yan Q, Wei C, Guo Q, Fan X, Ma W. Berberine targets the electron transport chain complex I and reveals the landscape of OXPHOS dependency in acute myeloid leukemia with IDH1 mutation. Chin J Nat Med 2023; 21:136-145. [PMID: 36871981 DOI: 10.1016/s1875-5364(23)60391-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Indexed: 03/07/2023]
Abstract
Metabolic reprogramming, a newly recognized trait of tumor biology, is an intensively studied prospect for oncology medicines. For numerous tumors and cancer cell subpopulations, oxidative phosphorylation (OXPHOS) is essential for their biosynthetic and bioenergetic functions. Cancer cells with mutations in isocitrate dehydrogenase 1 (IDH1) exhibit differentiation arrest, epigenetic and transcriptional reprogramming, and sensitivity to mitochondrial OXPHOS inhibitors. In this study, we report that berberine, which is widely used in China to treat intestinal infections, acted solely at the mitochondrial electron transport chain (ETC) complex I, and that its association with IDH1 mutant inhibitor (IDH1mi) AG-120 decreased mitochondrial activity and enhanced antileukemic effect in vitro andin vivo. Our study gives a scientific rationale for the therapy of IDH1 mutant acute myeloid leukemia (AML) patients using combinatory mitochondrial targeted medicines, particularly those who are resistant to or relapsing from IDH1mi.
Collapse
Affiliation(s)
- Zhe Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Yunfu Shen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Wenjun Liu
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Yan Yang
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Ling Guo
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Qin Yan
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Chengming Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qulian Guo
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China.
| |
Collapse
|
46
|
Mecklenbrauck R, Heuser M. Resistance to targeted therapies in acute myeloid leukemia. Clin Exp Metastasis 2023; 40:33-44. [PMID: 36318439 PMCID: PMC9898349 DOI: 10.1007/s10585-022-10189-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/12/2022] [Indexed: 02/04/2023]
Abstract
The introduction of new targeted therapies to the treatment algorithm of acute myeloid leukemia (AML) offers new opportunities, but also presents new challenges. Patients diagnosed with AML receiving targeted therapies as part of lower intensity regimens will relapse inevitably due to primary or secondary resistance mechanisms. In this review, we summarize the current knowledge on the main mechanisms of resistance to targeted therapies in AML. Resistance to FLT3 inhibitors is mainly mediated by on target mutations and dysregulation of downstream pathways. Switching the FLT3 inhibitor has a potential therapeutic benefit. During treatment with IDH inhibitors resistance can develop due to aberrant cell metabolism or secondary site IDH mutations. As a unique resistance mechanism the mutated IDH isotype may switch from IDH1 to IDH2 or vice versa. Resistance to gemtuzumab-ozogamicin is determined by the CD33 isotype and the degradation of the cytotoxin. The main mechanisms of resistance to venetoclax are the dysregulation of alternative pathways especially the upregulation of the BCL-2-analogues MCL-1 and BCL-XL or the induction of an aberrant cell metabolism. The introduction of therapies targeting immune processes will lead to new forms of therapy resistance. Knowing those mechanisms will help to develop strategies that can overcome resistance to treatment.
Collapse
Affiliation(s)
- Rabea Mecklenbrauck
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
47
|
Yap TA, Daver N, Mahendra M, Zhang J, Kamiya-Matsuoka C, Meric-Bernstam F, Kantarjian HM, Ravandi F, Collins ME, Francesco MED, Dumbrava EE, Fu S, Gao S, Gay JP, Gera S, Han J, Hong DS, Jabbour EJ, Ju Z, Karp DD, Lodi A, Molina JR, Baran N, Naing A, Ohanian M, Pant S, Pemmaraju N, Bose P, Piha-Paul SA, Rodon J, Salguero C, Sasaki K, Singh AK, Subbiah V, Tsimberidou AM, Xu QA, Yilmaz M, Zhang Q, Li Y, Bristow CA, Bhattacharjee MB, Tiziani S, Heffernan TP, Vellano CP, Jones P, Heijnen CJ, Kavelaars A, Marszalek JR, Konopleva M. Complex I inhibitor of oxidative phosphorylation in advanced solid tumors and acute myeloid leukemia: phase I trials. Nat Med 2023; 29:115-126. [PMID: 36658425 DOI: 10.1038/s41591-022-02103-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/21/2022] [Indexed: 01/21/2023]
Abstract
Although targeting oxidative phosphorylation (OXPHOS) is a rational anticancer strategy, clinical benefit with OXPHOS inhibitors has yet to be achieved. Here we advanced IACS-010759, a highly potent and selective small-molecule complex I inhibitor, into two dose-escalation phase I trials in patients with relapsed/refractory acute myeloid leukemia (NCT02882321, n = 17) and advanced solid tumors (NCT03291938, n = 23). The primary endpoints were safety, tolerability, maximum tolerated dose and recommended phase 2 dose (RP2D) of IACS-010759. The PK, PD, and preliminary antitumor activities of IACS-010759 in patients were also evaluated as secondary endpoints in both clinical trials. IACS-010759 had a narrow therapeutic index with emergent dose-limiting toxicities, including elevated blood lactate and neurotoxicity, which obstructed efforts to maintain target exposure. Consequently no RP2D was established, only modest target inhibition and limited antitumor activity were observed at tolerated doses, and both trials were discontinued. Reverse translational studies in mice demonstrated that IACS-010759 induced behavioral and physiological changes indicative of peripheral neuropathy, which were minimized with the coadministration of a histone deacetylase 6 inhibitor. Additional studies are needed to elucidate the association between OXPHOS inhibition and neurotoxicity, and caution is warranted in the continued development of complex I inhibitors as antitumor agents.
Collapse
Affiliation(s)
- Timothy A Yap
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Naval Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mikhila Mahendra
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jixiang Zhang
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meghan E Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Maria Emilia Di Francesco
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sisi Gao
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason P Gay
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonal Gera
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Han
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias J Jabbour
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Jennifer R Molina
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalia Baran
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maro Ohanian
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carolina Salguero
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anand K Singh
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Quanyun A Xu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Zhang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christopher A Bristow
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meenakshi B Bhattacharjee
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA
| | - Timothy P Heffernan
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher P Vellano
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip Jones
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cobi J Heijnen
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Psychological Sciences, Rice University, Houston, TX, USA
| | - Annemieke Kavelaars
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph R Marszalek
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Marina Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
48
|
The Effect of Oxidative Phosphorylation on Cancer Drug Resistance. Cancers (Basel) 2022; 15:cancers15010062. [PMID: 36612059 PMCID: PMC9817696 DOI: 10.3390/cancers15010062] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Recent studies have shown that oxidative phosphorylation (OXPHOS) is a target for the effective attenuation of cancer drug resistance. OXPHOS inhibitors can improve treatment responses to anticancer therapy in certain cancers, such as melanomas, lymphomas, colon cancers, leukemias and pancreatic ductal adenocarcinoma (PDAC). However, the effect of OXPHOS on cancer drug resistance is complex and associated with cell types in the tumor microenvironment (TME). Cancer cells universally promote OXPHOS activity through the activation of various signaling pathways, and this activity is required for resistance to cancer therapy. Resistant cancer cells are prevalent among cancer stem cells (CSCs), for which the main metabolic phenotype is increased OXPHOS. CSCs depend on OXPHOS to survive targeting by anticancer drugs and can be selectively eradicated by OXPHOS inhibitors. In contrast to that in cancer cells, mitochondrial OXPHOS is significantly downregulated in tumor-infiltrating T cells, impairing antitumor immunity. In this review, we summarize novel research showing the effect of OXPHOS on cancer drug resistance, thereby explaining how this metabolic process plays a dual role in cancer progression. We highlight the underlying mechanisms of metabolic reprogramming in cancer cells, as it is vital for discovering new drug targets.
Collapse
|
49
|
Jiang X, Huang X, Zheng G, Jia G, Li Z, Ding X, Lei L, Yuan L, Xu S, Gao N. Targeting PI4KA sensitizes refractory leukemia to chemotherapy by modulating the ERK/AMPK/OXPHOS axis. Am J Cancer Res 2022; 12:6972-6988. [PMID: 36276647 PMCID: PMC9576605 DOI: 10.7150/thno.76563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022] Open
Abstract
Background: The emergence of chemoresistance in leukemia markedly impedes chemotherapeutic efficacy and dictates poor prognosis. Recent evidence has revealed that phosphatidylinositol 4 kinase-IIIα (PI4KA) plays a critical role in tumorigenesis. However, the molecular mechanisms of PI4KA-regulated chemoresistance and leukemogenesis remain largely unknown. Methods: Liquid chromatography-mass spectrometry (LC-MS), patient samples and leukemia xenograft mouse models were used to investigate whether PI4KA was an effective target to overcome chemoresistance in leukemia. Enzyme-linked immunosorbent assay (ELISA) and molecular mechanics/generalized born surface area (MM/GBSA) method were employed to identify cepharanthine (CEP) as a novel PI4KA inhibitor. Results: High expression of PI4KA was observed in drug-resistant leukemia cells or in relapsed leukemia patients, which was correlated with poor overall survival. Depletion of PI4KA sensitized drug-resistant leukemia cells to chemotherapeutic drugs in vitro and in vivo by regulating ERK/AMPK/OXPHOS axis. We also identified cepharanthine (CEP) as a novel PI4KA inhibitor, which could undermine the stability of the PI4KA/TTC7/FAM126 complex, enhancing the sensitivity of drug-resistant leukemia cells to chemotherapeutic drugs in vitro and in vivo. Conclusions: Our study underscored the potential of therapeutic targeting of PI4KA to overcome chemoresistance in leukemia. A combination of the PI4KA inhibitor with classic chemotherapeutic agents could represent a novel therapeutic strategy for the treatment of refractory leukemia.
Collapse
Affiliation(s)
- Xiuxing Jiang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Xiangtao Huang
- Department of Hematology, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Guoxun Zheng
- Shanghai StoneWise AI Technology Co. Ltd. Shanghai 201210, China
| | - Guanfei Jia
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Zhiqiang Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Xin Ding
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Ling Lei
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Liang Yuan
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Shuangnian Xu
- Department of Hematology, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Ning Gao
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, China.,Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, China
| |
Collapse
|
50
|
Sharma A, Nair R, Achreja A, Mittal A, Gupta P, Balakrishnan K, Edgar CL, Animasahun O, Dwivedi B, Barwick BG, Gupta VA, Matulis SM, Bhasin M, Lonial S, Nooka AK, Wiita AP, Boise LH, Nagrath D, Shanmugam M. Therapeutic implications of mitochondrial stress-induced proteasome inhibitor resistance in multiple myeloma. SCIENCE ADVANCES 2022; 8:eabq5575. [PMID: 36170375 PMCID: PMC9519052 DOI: 10.1126/sciadv.abq5575] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The connections between metabolic state and therapy resistance in multiple myeloma (MM) are poorly understood. We previously reported that electron transport chain (ETC) suppression promotes sensitivity to the BCL-2 antagonist venetoclax. Here, we show that ETC suppression promotes resistance to proteasome inhibitors (PIs). Interrogation of ETC-suppressed MM reveals integrated stress response-dependent suppression of protein translation and ubiquitination, leading to PI resistance. ETC and protein translation gene expression signatures from the CoMMpass trial are down-regulated in patients with poor outcome and relapse, corroborating our in vitro findings. ETC-suppressed MM exhibits up-regulation of the cystine-glutamate antiporter SLC7A11, and analysis of patient single-cell RNA-seq shows that clusters with low ETC gene expression correlate with higher SLC7A11 expression. Furthermore, erastin or venetoclax treatment diminishes mitochondrial stress-induced PI resistance. In sum, our work demonstrates that mitochondrial stress promotes PI resistance and underscores the need for implementing combinatorial regimens in MM cognizant of mitochondrial metabolic state.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Remya Nair
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Abhinav Achreja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Anjali Mittal
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pulkit Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kamakshi Balakrishnan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Claudia L. Edgar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Olamide Animasahun
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Bhakti Dwivedi
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Vikas A. Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Shannon M. Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ajay K. Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Arun P. Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
- Corresponding author.
| |
Collapse
|