1
|
Gabellier L, Bosetta E, Heiblig M, Sarry JE. Metabolism and therapeutic response in acute myeloid leukemia with IDH1/2 mutations. Trends Cancer 2025; 11:475-490. [PMID: 39955197 DOI: 10.1016/j.trecan.2025.01.011] [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: 12/04/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/17/2025]
Abstract
Pathogenic variants of isocitrate dehydrogenase 1 and 2 (IDH1/2) genes are present in approximately 20% of acute myeloid leukemia (AML) cases, resulting in the oncometabolite R-2-hydroxyglutarate (R-2-HG). The accumulation of R-2-HG in leukemic cells and in their niche induces epigenetic modifications, profound rewiring of the cellular metabolism, and microenvironmental remodeling. These changes promote cellular differentiation bias, enhancing the survival and proliferation of leukemic cells, and thus playing a pivotal role in leukemogenesis and resistance to standard AML therapy. This review focuses on the different perspectives offered by studying metabolism and resistance to standard treatments in AML with IDH1 or IDH2 pathogenic variants, for the development of new biomarkers and therapeutic solutions.
Collapse
MESH Headings
- Humans
- Isocitrate Dehydrogenase/genetics
- Isocitrate Dehydrogenase/metabolism
- Isocitrate Dehydrogenase/antagonists & inhibitors
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mutation
- Glutarates/metabolism
- Drug Resistance, Neoplasm/genetics
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Epigenesis, Genetic
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
Collapse
Affiliation(s)
- Ludovic Gabellier
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France; Team «Ubiquitin family in hematological malignancies», Institut de Génétique Moléculaire de Montpellier, CNRS UMR5535, Université de Montpellier, Montpellier, France
| | - Enzo Bosetta
- Centre de Recherches en Cancérologie de Toulouse, U1037, Inserm, Université de Toulouse, Toulouse, France
| | - Maël Heiblig
- Service d'Hématologie Clinique, Hôpital Lyon Sud Pierre-Bénite, Lyon, France; Team «Lymphoma Immuno-Biology», Inserm U1111, CNRS UMR5308, Université Claude Bernard, Lyon I - ENS de Lyon, Faculté de Médecine Lyon-Sud, Lyon, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, U1037, Inserm, Université de Toulouse, Toulouse, France.
| |
Collapse
|
2
|
Addanki S, Kim L, Stevens A. Understanding and Targeting Metabolic Vulnerabilities in Acute Myeloid Leukemia: An Updated Comprehensive Review. Cancers (Basel) 2025; 17:1355. [PMID: 40282531 PMCID: PMC12025543 DOI: 10.3390/cancers17081355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by aggressive proliferation and metabolic reprogramming that support its survival and resistance to therapy. This review explores the metabolic distinctions between AML cells and normal hematopoietic stem cells (HSCs), emphasizing the role of altered mitochondrial function, oxidative phosphorylation (OXPHOS), and biosynthetic pathways in leukemic progression. AML cells exhibit distinct metabolic vulnerabilities, including increased mitochondrial biogenesis, reliance on glycolysis and amino acid metabolism, and unique signaling interactions that sustain leukemic stem cells (LSCs). These dependencies provide potential therapeutic targets, as metabolic inhibitors have demonstrated efficacy in disrupting AML cell survival while sparing normal hematopoietic cells. We examine current and emerging metabolic therapies, such as inhibitors targeting glycolysis, amino acid metabolism, and lipid biosynthesis, highlighting their potential in overcoming drug resistance. However, challenges remain in translating these strategies into clinical practice due to AML's heterogeneity and adaptability. Further research into AML's metabolic plasticity and precision medicine approaches is crucial for improving treatment outcomes. Understanding and exploiting AML's metabolic vulnerabilities could pave the way for novel, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Sridevi Addanki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Alexandra Stevens
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
3
|
Liu Y, Bollino DR, Bah OM, Strovel ET, Le TV, Zarrabi J, Philip S, Lapidus RG, Baer MR, Niyongere S, Duong VH, Dougherty CC, Beumer JH, Caprinolo KD, Kamangar F, Emadi A. A phase 1 study of the amino acid modulator pegcrisantaspase and venetoclax for relapsed or refractory acute myeloid leukemia. Blood 2025; 145:486-496. [PMID: 39437546 PMCID: PMC11826518 DOI: 10.1182/blood.2024024837] [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: 04/01/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT Glutamine dependency has been shown to be a metabolic vulnerability in acute myeloid leukemia (AML). Prior studies using several in vivo AML models showed that depletion of plasma glutamine, induced by long-acting crisantaspase (pegcrisantaspase [PegC]) was synergistic with the B-cell lymphoma-2 (BCL-2) inhibitor venetoclax (Ven), resulting in significantly reduced leukemia burden and enhanced survival. Here, we report a phase 1 study of the combination of Ven and PegC (VenPegC) for treating adult patients with relapsed or refractory AML, including patients who had previously received Ven. The primary end points were the incidence of regimen-limiting toxicities (RLTs) and the maximum tolerated dose (MTD). Twenty-five patients received at least 1 PegC dose with Ven, and 18 efficacy-evaluable patients completed at least 1 VenPegC cycle; 12 (67%) had previously received Ven. Hyperbilirubinemia was the RLT and occurred in 60% of patients treated with VenPegC; 20% had grade ≥3 bilirubin elevations. MTD was determined to be Ven 400 mg daily with biweekly PegC 750 IU/m2. The most common treatment-related adverse events of any grade in 25 patients who received VenPegC included antithrombin III decrease (52%), elevated transaminases (36%-48%), fatigue (28%), and hypofibrinogenemia (24%). No thromboembolic or hemorrhagic adverse events or clinical pancreatitis were observed. The overall complete remission rate in efficacy-evaluable patients was 33%. Response correlated with alterations in proteins involved in messenger RNA translation. In patients with RUNX1 mutations, the composite complete remission rate was 100%. This study was registered at www.ClinicalTrials.gov as #NCT04666649.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Sulfonamides/administration & dosage
- Sulfonamides/adverse effects
- Sulfonamides/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Female
- Male
- Middle Aged
- Aged
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Asparaginase/administration & dosage
- Asparaginase/adverse effects
- Asparaginase/therapeutic use
- Maximum Tolerated Dose
- Aged, 80 and over
- Recurrence
- Young Adult
- Polyethylene Glycols
Collapse
Affiliation(s)
- Yuchen Liu
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Dominique R. Bollino
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Osman M. Bah
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Erin T. Strovel
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Tien V. Le
- Cancer Therapeutics Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Jinoos Zarrabi
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Sunita Philip
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Rena G. Lapidus
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Maria R. Baer
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Sandrine Niyongere
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Vu H. Duong
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | | | - Jan H. Beumer
- Cancer Therapeutics Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Farin Kamangar
- Department of Biology, School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD
| | - Ashkan Emadi
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
4
|
Ma X, Sun C, Ding X, Xu J, Zhang Y, Deng T, Wang Y, Yang H, Ding R, Li H, Wang D, Zheng M. Mechanism analysis and targeted therapy of IDH gene mutation in glioma. Am J Cancer Res 2025; 15:248-270. [PMID: 39949933 PMCID: PMC11815359 DOI: 10.62347/nsxc2205] [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: 09/16/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Isocitrate dehydrogenase (IDH) is a pivotal enzyme responsible for catalyzing the oxidative decarboxylation of isocitrate into α-ketoglutarate (α-KG). This enzyme serves as a crucial regulator in the tricarboxylic acid cycle (TCA cycle), acting as a rate-limiting step. Its role extends beyond mere metabolic function, influencing cellular homeostasis and overall cell function. In the past decade, prominent research in cancer genetics has revealed that genes responsible for encoding isocitrate dehydrogenase are commonly mutated across various human malignancies. Significant research in the field has shown that these mutations are commonly found in diseases like glioma, acute myeloid leukemia (AML), cholangiocarcinoma (CCA), chondrosarcoma, and thyroid cancer (TC). As research on IDH progresses, deeper insights into the biological effects of IDH mutations have been gained, unveiling their potential role in tumorigenesis. In addition, IDH mutants' unique activities creates new pathways in tumor metabolism, gene rearrangement, and therapeutic resistance. Currently, innovative molecular targeting strategies for genes bearing mutations in IDH have been devised to enhance the therapeutic efficacy against cancers harboring IDH mutations. These methods represent a promising avenue for improving treatment outcomes in IDH-mutated malignancies. This article mainly summarizes the related research on glioma caused by IDH mutation, and focuses on the biological characteristics and transformation of IDH.
Collapse
Affiliation(s)
- Xingyuan Ma
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Chao Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical UniversityBeijing 100070, China
| | - Xiao Ding
- The Third Department of Surgery, Armed Police Hospital of TianjinTianjin 300163, China
| | - Jiaqi Xu
- Edinburgh Medical School, The University of EdinburghEdinburgh EH16 4SB, Scotland, UK
| | - Yuhang Zhang
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Tingzhen Deng
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Yatao Wang
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Haijun Yang
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Ruiwen Ding
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Haotian Li
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Dawen Wang
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| | - Maohua Zheng
- The First School of Clinical Medicine, Lanzhou UniversityLanzhou 730000, Gansu, China
- Department of Neurosurgery, The First Hospital of Lanzhou UniversityLanzhou 730000, Gansu, China
| |
Collapse
|
5
|
de la Fuente MI, Touat M, van den Bent MJ, Preusser M, Peters KB, Young RJ, Huang RY, Ellingson BM, Capper D, Phillips JJ, Halasz LM, Shih HA, Rudà R, Lim-Fat MJ, Blumenthal DT, Weller M, Arakawa Y, Whittle JR, Ducray F, Reardon DA, Bi WL, Minniti G, Rahman R, Hervey-Jumper S, Chang SM, Wen PY. The role of vorasidenib in the treatment of isocitrate dehydrogenase-mutant glioma. Neuro Oncol 2024:noae259. [PMID: 39723472 DOI: 10.1093/neuonc/noae259] [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] [Indexed: 12/28/2024] Open
Abstract
Isocitrate dehydrogenase (IDH)-mutant gliomas are the most common malignant primary brain tumors in young adults. This condition imposes a substantial burden on patients and their caregivers, marked by neurocognitive deficits and high mortality rates due to tumor progression, coupled with significant morbidity from current treatment modalities. Although surgery, radiation therapy, and chemotherapy improve survival, these treatments can adversely affect cognitive function, quality of life, finances, employment status, and overall independence. Consequently, there is an urgent need for innovative strategies that delay progression and the use of radiation therapy and chemotherapy. The recent Federal Drug Administration (FDA) approval of vorasidenib, a brain-penetrant small molecule targeting mutant IDH1/2 proteins, heralds a shift in the therapeutic landscape for IDH-mutant gliomas. In this review, we address the role of vorasidenib in the treatment of IDH-mutant gliomas, providing a roadmap for its incorporation into daily practice. We discuss ongoing clinical trials with vorasidenib and other IDH inhibitors, as single-agent or in combination with other therapies, as well as current challenges and future directions.
Collapse
Affiliation(s)
- Macarena I de la Fuente
- Department of Neurology, University of Miami, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Mehdi Touat
- Service de Neuro-oncologie, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, Paris Brain Institute, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Paris, France
- Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Martin J van den Bent
- Service de Neuro-oncologie, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, Paris Brain Institute, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Paris, France
- Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Katherine B Peters
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina, USA
| | - Robert J Young
- Service Neuroradiology, Department of Radiology, Memorial Sloan Kettering Cancer, New York, New York, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David Capper
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Joanna J Phillips
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Lia M Halasz
- Department of Radiation Oncology, University of Washington/Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Helen A Shih
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Mary Jane Lim-Fat
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - James R Whittle
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Personalised Oncology Division, WEHI, Parkville, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - François Ducray
- Department of Neuro-Oncology, East Group Hospital, Hospices Civils de Lyon, Université de Lyon, Université Claude Bernard, Lyon, France
| | - David A Reardon
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Giuseppe Minniti
- IRCCS Neuromed, Pozzilli, Isernia, Italy
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Rifaquat Rahman
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shawn Hervey-Jumper
- Department of Neurological Surgery, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Susan M Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Capelletti MM, Montini O, Ruini E, Tettamanti S, Savino AM, Sarno J. Unlocking the Heterogeneity in Acute Leukaemia: Dissection of Clonal Architecture and Metabolic Properties for Clinical Interventions. Int J Mol Sci 2024; 26:45. [PMID: 39795903 PMCID: PMC11719665 DOI: 10.3390/ijms26010045] [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: 11/21/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Genetic studies of haematological cancers have pointed out the heterogeneity of leukaemia in its different subpopulations, with distinct mutations and characteristics, impacting the treatment response. Next-generation sequencing (NGS) and genome-wide analyses, as well as single-cell technologies, have offered unprecedented insights into the clonal heterogeneity within the same tumour. A key component of this heterogeneity that remains unexplored is the intracellular metabolome, a dynamic network that determines cell functions, signalling, epigenome regulation, immunity and inflammation. Understanding the metabolic diversities among cancer cells and their surrounding environments is therefore essential in unravelling the complexities of leukaemia and improving therapeutic strategies. Here, we describe the currently available methodologies and approaches to addressing the dynamic heterogeneity of leukaemia progression. In the second section, we focus on metabolic leukaemic vulnerabilities in acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL). Lastly, we provide a comprehensive overview of the most interesting clinical trials designed to target these metabolic dependencies, highlighting their potential to advance therapeutic strategies in leukaemia treatment. The integration of multi-omics data for cancer identification with the metabolic states of tumour cells will enable a comprehensive "micro-to-macro" approach for the refinement of clinical practices and delivery of personalised therapies.
Collapse
Affiliation(s)
- Martina Maria Capelletti
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Orsola Montini
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Emilio Ruini
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Angela Maria Savino
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Jolanda Sarno
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
7
|
Duan H, Lai Q, Jiang Y, Yang L, Deng M, Lin Z, Shan W, Zhong M, Yao J, Zhang L, Xu B, Zha J. Chiglitazar diminishes the warburg effect through PPARγ/mTOR/PKM2 and increases the sensitivity of imatinib in chronic myeloid leukemia. Exp Hematol Oncol 2024; 13:121. [PMID: 39696470 DOI: 10.1186/s40164-024-00589-1] [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: 09/03/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND A tyrosine kinase inhibitor (TKI) such as Imatinib (IM) is the preferred treatment for Chronic Myeloid Leukemia (CML). However, the emergence of IM resistance presents a significant challenge to disease management. A characteristic of cancer cells, including IM-resistant CMLs, are characterized by heightened uptake of glucose and aberrant glycolysis in the cytosol, which is known as the Warburg effect. In addition to its potential to modulate the Warburg effect, Chiglitazar (Chi), a compound that regulates glucose metabolism, has also been investigated for its implication in cancer treatment. This suggests that combining Chi with IM may be a therapeutic strategy for overcoming IM resistance in CML. METHODS Sensitive and IM-resistance CML cells were treated with Chi in vitro, followed by detecting of extracellular acidification rate (ECAR) using a Seahorse XF Analyzer. CML cell proliferation, cell cycle distribution, and apoptosis were tested by CCK-8 assay and flow cytometry. RNA sequencing was utilized to investigate potential transcriptional changes induced by Chi usage. In vivo studies were conducted on immunodeficient mice implanted with CML cells and given Chi and/or IM later. Tumor growth was monitored, as well as tumor burden and survival rates between groups. RESULTS Our metabonomic, transcriptomic, and molecular biology studies demonstrated that Chi, in part, diminished the Warburg effect by reducing glucose and lactate production in imatinib-resistant CML cells through the PPARγ/mTOR/PKM2 pathway. This modulation of glucose metabolism resulted in reduced cell proliferation and enhanced sensitivity to IM in imatinib-resistant CML cells in vitro. Rescue assay by introducing shPPARγ or mTOR activator verified the underlying regulatory pathway. Also, the combination of Chi and IM synergistically increased the sensitivity of IM in vivo and prolonged the survival of imatinib-resistance CML transplanted mice. CONCLUSIONS Our results demonstrated the potential of Chi to overcome IM resistance in vitro and in vivo. By inhibiting the Warburg effect through the PPARγ/mTOR/PKM2 pathway, Chi resensitizes CML cells towards imatinib treatment. Combining IM with Chi is an alternative therapeutic option for CML management, especially for IM-resistant CML patients.
Collapse
Affiliation(s)
- Hongpeng Duan
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Qian Lai
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Liuzhen Yang
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Manman Deng
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Zhijuan Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Weihang Shan
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jingwei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Li Zhang
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China.
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Zhenhai Road, Xiamen, 361003, Fujian, People's Republic of China.
| |
Collapse
|
8
|
Zhou H, Xiang W, Zhou G, Rodrigues-Lima F, Guidez F, Wang L. Metabolic dysregulation in myelodysplastic neoplasm: impact on pathogenesis and potential therapeutic targets. Med Oncol 2024; 42:23. [PMID: 39644425 DOI: 10.1007/s12032-024-02575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Despite significant advancements in the research of the pathogenesis mechanisms of Myelodysplastic Neoplasm (MDS) in recent years, there are still many gaps to fill. The advancement of metabolomics studies has led to a research booming in clarifying the impact of metabolic abnormalities during the pathogenesis of MDS. The present review primarily focuses on the dysregulated metabolic pathways, exploring the influences on the pathogenesis of MDS and their roles during the course of the disease. Furthermore, we discuss the potential of relevant metabolic pathways as therapeutic targets, along with the latest metabolic-related treatment drugs and approaches.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wenqiong Xiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Guangyu Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fernando Rodrigues-Lima
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle Et Adaptative, 75013, Paris, France
| | - Fabien Guidez
- Université Paris Cité, Institut de Recherche Saint Louis INSERM UMR_S1131, 75010, Paris, France
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
9
|
Baek C, Laurenge A, Touat M. Advances in the treatment of IDH-mutant gliomas. Curr Opin Neurol 2024; 37:708-716. [PMID: 39253756 DOI: 10.1097/wco.0000000000001316] [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: 09/11/2024]
Abstract
PURPOSE OF REVIEW Isocitrate dehydrogenase (IDH) mutation is a defining molecular driver of WHO grade 2-4 astrocytomas and oligodendrogliomas. In this article, we review the recent therapeutic approaches specifically targeting IDH-mutant gliomas and summarize ongoing clinical trials in this population. RECENT FINDINGS The IDH inhibitor vorasidenib recently demonstrated its efficacy after surgical resection in grade 2 IDH-mutated gliomas. Several studies in patients with IDH-mutant gliomas are currently exploring various strategies to target IDH mutations, including the use of small-molecule inhibitors, immunotherapies, peptide vaccines and agents targeting metabolic and epigenomic vulnerabilities. SUMMARY Mutant-IDH targeting holds significant promise in treating progressive or recurrent IDH-mutant gliomas. Recent results with IDH inhibitors will change practice and influence the existing guidelines in a near future.
Collapse
Affiliation(s)
- Chooyoung Baek
- Service de Neuro-oncologie, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université
| | - Alice Laurenge
- Service de Neuro-oncologie, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université
- Institut du Cerveau, Paris Brain Institute (ICM), Inserm, CNRS, Sorbonne Université, AP-HP, SIRIC CURAMUS, Paris, France
| | - Mehdi Touat
- Service de Neuro-oncologie, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université
- Institut du Cerveau, Paris Brain Institute (ICM), Inserm, CNRS, Sorbonne Université, AP-HP, SIRIC CURAMUS, Paris, France
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Chen Y. Dissecting L-glutamine metabolism in acute myeloid leukemia: single-cell insights and therapeutic implications. J Transl Med 2024; 22:1002. [PMID: 39506790 PMCID: PMC11539756 DOI: 10.1186/s12967-024-05779-3] [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: 07/10/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a rapidly progressing blood cancer. The prognosis of AML can be challenging, emphasizing the need for ongoing research and innovative approaches to improve outcomes in individuals affected by this formidable hematologic malignancy. METHODS In this study, we used single-cell RNA sequencing (scRNA-seq) from AML patients to investigate the impact of L-glutamine metabolism-related genes on disease progression. RESULTS Our analysis revealed increased glutamine-related activity in CD34 + pre-B cells, suggesting a potential regulatory role in tumorigenesis and AML progression. Furthermore, intercellular communication analysis revealed a significant signaling pathway involving macrophage migration inhibitory factor signaling through CD74 + CD44 within CD34 + pre-B cells, which transmit signals to pre-dendritic cells and monocytes. Ligands for this pathway were predominantly expressed in stromal cells, naïve T cells, and CD34 + pre-B cells. CD74, the pertinent receptor, was predominantly detected in a variety of cellular components, including stromal cells, pre-dendritic cells, plasmacytoid dendritic cells, and hematopoietic progenitors. The study's results provide insights into the possible interplay among these cell types and their collective contribution to the pathogenesis of AML. Moreover, we identified 10 genes associated with AML prognosis, including CCL5, CD52, CFD, FABP5, LGALS1, NUCB2, PSAP, S100A4, SPINK2, and VCAN. Among these, CCL5 and CD52 have been implicated in AML progression and are potential therapeutic targets. CONCLUSIONS This thorough examination of AML biology significantly deepens our grasp of the disease and presents pivotal information that could guide the creation of innovative treatment strategies for AML patients.
Collapse
Affiliation(s)
- Yanli Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
11
|
Shen S, Zhuang H. Homoharringtonine in the treatment of acute myeloid leukemia: A review. Medicine (Baltimore) 2024; 103:e40380. [PMID: 39496012 PMCID: PMC11537654 DOI: 10.1097/md.0000000000040380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by the accumulation of immature myeloid precursor cells. Over half of AML patients fail to achieve long-term disease-free survival under existing therapy, and the overall prognosis is poor, necessitating the urgent development of novel therapeutic approaches. The plant alkaloid homoharringtonine (HHT), which has anticancer properties, was first identified more than 40 years ago. It works in a novel method of action that prevents the early elongation phase of protein synthesis. HHT has been widely utilized in the treatment of AML, with strong therapeutic effects, few toxic side effects, and the ability to enhance AML patients' prognoses. In AML, HHT can induce cell apoptosis through multiple pathways, exerting synergistic antitumor effects, according to clinical and pharmacological research. About its modes of action, some findings have been made recently. This paper reviews the development of research on the mechanisms of HHT in treating AML to offer insights for further research and clinical therapy.
Collapse
Affiliation(s)
- Siyu Shen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Haifeng Zhuang
- Department of Clinical Hematology and Transfusion, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
12
|
DiNardo CD, Verma D, Baran N, Bhagat TD, Skwarska A, Lodi A, Saxena K, Cai T, Su X, Guerra VA, Poigaialwar G, Kuruvilla VM, Konoplev S, Gordon-Mitchell S, Pradhan K, Aluri S, Hackman GL, Chaudhry S, Collins M, Sweeney SR, Busquets J, Rathore AS, Deng Q, Green MR, Grant S, Demo S, Choudhary GS, Sahu S, Agarwal B, Spodek M, Thiruthuvanathan V, Will B, Steidl U, Tippett GD, Burger J, Borthakur G, Jabbour E, Pemmaraju N, Kadia T, Kornblau S, Daver NG, Naqvi K, Short NJ, Garcia-Manero G, Tiziani S, Verma A, Konopleva M. Glutaminase inhibition in combination with azacytidine in myelodysplastic syndromes: a phase 1b/2 clinical trial and correlative analyses. NATURE CANCER 2024; 5:1515-1533. [PMID: 39300320 DOI: 10.1038/s43018-024-00811-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 07/24/2024] [Indexed: 09/22/2024]
Abstract
Malignancies are reliant on glutamine as an energy source and a facilitator of aberrant DNA methylation. We demonstrate preclinical synergy of telaglenastat (CB-839), a selective glutaminase inhibitor, combined with azacytidine (AZA), followed by a single-arm, open-label, phase 1b/2 study in persons with advanced myelodysplastic syndrome (MDS). The dual primary endpoints evaluated clinical activity, safety and tolerability; secondary endpoints evaluated pharmacokinetics, pharmacodynamics, overall survival, event-free survival and duration of response. The dose-escalation study included six participants and the dose-expansion study included 24 participants. Therapy was well tolerated and led to an objective response rate of 70% with (marrow) complete remission in 53% of participants and a median overall survival of 11.6 months, with evidence of myeloid differentiation in responders determined by single-cell RNA sequencing. Glutamine transporter solute carrier family 38 member 1 in MDS stem cells was associated with clinical responses and predictive of worse prognosis in a large MDS cohort. These data demonstrate the safety and efficacy of CB-839 and AZA as a combined metabolic and epigenetic approach in MDS. ClinicalTrials.gov identifier: NCT03047993 .
Collapse
Affiliation(s)
- Courtney D DiNardo
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Divij Verma
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Natalia Baran
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Section of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Tushar D Bhagat
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna Skwarska
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Dell Pediatric Research Institute, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Kapil Saxena
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tianyu Cai
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Veronica A Guerra
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gowri Poigaialwar
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vinitha M Kuruvilla
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shanisha Gordon-Mitchell
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kith Pradhan
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Srinivas Aluri
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - G Lavender Hackman
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Dell Pediatric Research Institute, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Sovira Chaudhry
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Meghan Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Dell Pediatric Research Institute, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Shannon R Sweeney
- Dell Pediatric Research Institute, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Institute for Cell and Molecular Biology, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Jonathan Busquets
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Dell Pediatric Research Institute, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Atul Singh Rathore
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Qing Deng
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael R Green
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven Grant
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Susan Demo
- Calithera Biosciences, San Francisco, CA, USA
| | - Gaurav S Choudhary
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Srabani Sahu
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Mason Spodek
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Victor Thiruthuvanathan
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Britta Will
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ulrich Steidl
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - George D Tippett
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jan Burger
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven Kornblau
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval G Daver
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kiran Naqvi
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA.
- Dell Pediatric Research Institute, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
- Institute for Cell and Molecular Biology, College of Natural Sciences, 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.
| | - Amit Verma
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Marina Konopleva
- Departament of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
13
|
Zhou J, Zhang N, Zuo Y, Xu F, Cheng L, Fu Y, Yang F, Shu M, Zhou M, Zou W, Zhang S. Glutamine metabolism-related genes predict the prognostic risk of acute myeloid leukemia and stratify patients by subtype analysis. Hereditas 2024; 161:35. [PMID: 39300580 DOI: 10.1186/s41065-024-00338-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a genetically heterogeneous disease in which glutamine (Gln) contributes to AML progression. Therefore, this study aimed to identify potential prognostic biomarkers for AML based on Gln metabolism-related genes. METHODS Gln-related genes that were differentially expressed between Cancer Genome Atlas-based AML and normal samples were analyzed using the limma package. Univariate, least absolute shrinkage, selection operators, and stepwise Cox regression analyses were used to identify prognostic signatures. Risk score-based prognostic and nomogram models were constructed to predict the prognostic risk of AML. Subsequently, consistent cluster analysis was performed to stratify patients into different subtypes, and subtype-related module genes were screened using weighted gene co-expression network analysis. RESULTS Through a series of regression analyses, HGF, ANGPTL3, MB, F2, CALR, EIF4EBP1, EPHX1, and PDHA1 were identified as potential prognostic biomarkers of AML. Prognostic and nomogram models constructed based on these genes could significantly differentiate between high- and low-risk AML with high predictive accuracy. The eight-signature also stratified patients with AML into two subtypes, among which Cluster 2 was prone to a high risk of AML prognosis. These two clusters exhibited different immune profiles. Of the subtype-related module genes, the HOXA and HOXB family genes may be genetic features of AML subtypes. CONCLUSION Eight Gln metabolism-related genes were identified as potential biomarkers of AML to predict prognostic risk. The molecular subtypes clustered by these genes enabled prognostic risk stratification.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China.
| | - Na Zhang
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Yan Zuo
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Feng Xu
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Lihua Cheng
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Yuanyuan Fu
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Fudong Yang
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Min Shu
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Mi Zhou
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Wenting Zou
- Department of Hematology, Deyang People's Hospital, No. 173 Taishan North Road, Section 1, Jingyang District, Deyang, 618000, Sichuan, China
| | - Shengming Zhang
- Department of health management, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China.
| |
Collapse
|
14
|
Wang Z, Liu M, Yang Q. Glutamine and leukemia research: progress and clinical prospects. Discov Oncol 2024; 15:391. [PMID: 39215845 PMCID: PMC11365919 DOI: 10.1007/s12672-024-01245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Leukemia is an abnormal proliferation of white blood cells that occurs in bone marrow and expands through the blood. It arises from dysregulated differentiation, uncontrolled growth, and inhibition of apoptosis. Glutamine (GLN) is a "conditionally essential" amino acid that promotes growth and proliferation of leukemic cells. Recently, details about the role of GLN and its metabolism in the diagnosis and treatment of acute myeloid, chronic lymphocytic, and acute lymphoblastic leukemia have emerged. The uptake of GLN by leukemia cells and the dynamic changes of glutamine-related indexes in leukemia patients may be able to assist in determining whether the condition of leukemia is in a state of progression, remission or relapse. Utilizing the possible differences in GLN metabolism in different subtypes of leukemia may help to differentiate between different subtypes of leukemia, thus providing a basis for accurate diagnosis. Targeting GLN metabolism in leukemia requires simultaneous blockade of multiple metabolic pathways without interfering with the normal cellular and immune functions of the body to achieve effective leukemia therapy. The present review summarizes recent advances, possible applications, and clinical perspectives of GLN metabolism in leukemia. In particular, it focuses on the prospects of GLN metabolism in the diagnosis and treatment of acute myeloid leukemia. The review provides new directions and hints at potential roles for future clinical treatments and studies.
Collapse
Affiliation(s)
- Zexin Wang
- Mianyang Central Hospital, Fucheng District, Mianyang, 621000, Sichuan, China.
| | - Miao Liu
- Mianyang Central Hospital, Fucheng District, Mianyang, 621000, Sichuan, China
| | - Qiang Yang
- Mianyang Central Hospital, Fucheng District, Mianyang, 621000, Sichuan, China
| |
Collapse
|
15
|
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
|
16
|
Liu TY, Fu X, Yang Y, Gu J, Xiao M, Li DJ. Synergistic Effects of Glutamine Deprivation and Metformin in Acute Myeloid Leukemia. Curr Med Sci 2024; 44:799-808. [PMID: 39096478 DOI: 10.1007/s11596-024-2913-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/19/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVE The metabolic reprogramming of acute myeloid leukemia (AML) cells is a compensatory adaptation to meet energy requirements for rapid proliferation. This study aimed to examine the synergistic effects of glutamine deprivation and metformin exposure on AML cells. METHODS SKM-1 cells (an AML cell line) were subjected to glutamine deprivation and/or treatment with metformin or bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl) ethyl sulfide (BPTES, a glutaminase inhibitor) or cytarabine. Cell viability was detected by Cell Counting Kit-8 (CCK-8) assay, and cell apoptosis and reactive oxygen species (ROS) by flow cytometry. Western blotting was conducted to examine the levels of apoptotic proteins, including cleaved caspase-3 and poly(ADP-ribose) polymerase (PARP). Moreover, the human long noncoding RNA (lncRNA) microarray was used to analyze gene expression after glutamine deprivation, and results were confirmed with quantitative RT-PCR (qRT-PCR). The expression of metallothionein 2A (MT2A) was suppressed using siRNA. Cell growth and apoptosis were further detected by CCK-8 assay and flow cytometry, respectively, in cells with MT2A knockdown. RESULTS Glutamine deprivation or treatment with BPTES inhibited cell growth and induced apoptosis in SKM-1 cells. The lncRNA microarray result showed that the expression of MT family genes was significantly upregulated after glutamine deprivation. MT2A knockdown increased apoptosis, while proliferation was not affected in SKM-1 cells. In addition, metformin inhibited cell growth and induced apoptosis in SKM-1 cells. Both glutamine deprivation and metformin enhanced the sensitivity of SKM-1 cells to cytarabine. Furthermore, the combination of glutamine deprivation with metformin exhibited synergistic antileukemia effects on SKM-1 cells. CONCLUSION Targeting glutamine metabolism in combination with metformin is a promising new therapeutic strategy for AML.
Collapse
Affiliation(s)
- Tong-Yuan Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Fu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Gu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Deng-Ju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
17
|
Zhang Z, Huang J, Zhang Z, Shen H, Tang X, Wu D, Bao X, Xu G, Chen S. Application of omics in the diagnosis, prognosis, and treatment of acute myeloid leukemia. Biomark Res 2024; 12:60. [PMID: 38858750 PMCID: PMC11165883 DOI: 10.1186/s40364-024-00600-1] [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: 03/20/2024] [Accepted: 05/17/2024] [Indexed: 06/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is the most frequent leukemia in adults with a high mortality rate. Current diagnostic criteria and selections of therapeutic strategies are generally based on gene mutations and cytogenetic abnormalities. Chemotherapy, targeted therapies, and hematopoietic stem cell transplantation (HSCT) are the major therapeutic strategies for AML. Two dilemmas in the clinical management of AML are related to its poor prognosis. One is the inaccurate risk stratification at diagnosis, leading to incorrect treatment selections. The other is the frequent resistance to chemotherapy and/or targeted therapies. Genomic features have been the focus of AML studies. However, the DNA-level aberrations do not always predict the expression levels of genes and proteins and the latter is more closely linked to disease phenotypes. With the development of high-throughput sequencing and mass spectrometry technologies, studying downstream effectors including RNA, proteins, and metabolites becomes possible. Transcriptomics can reveal gene expression and regulatory networks, proteomics can discover protein expression and signaling pathways intimately associated with the disease, and metabolomics can reflect precise changes in metabolites during disease progression. Moreover, omics profiling at the single-cell level enables studying cellular components and hierarchies of the AML microenvironment. The abundance of data from different omics layers enables the better risk stratification of AML by identifying prognosis-related biomarkers, and has the prospective application in identifying drug targets, therefore potentially discovering solutions to the two dilemmas. In this review, we summarize the existing AML studies using omics methods, both separately and combined, covering research fields of disease diagnosis, risk stratification, prognosis prediction, chemotherapy, as well as targeted therapy. Finally, we discuss the directions and challenges in the application of multi-omics in precision medicine of AML. Our review may inspire both omics researchers and clinical physicians to study AML from a different angle.
Collapse
Affiliation(s)
- Zhiyu Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Jiayi Huang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhibo Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongjie Shen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaowen Tang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiebing Bao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, Jiangsu, China.
- Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu Province, China.
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
18
|
Hasan Bou Issa L, Fléchon L, Laine W, Ouelkdite A, Gaggero S, Cozzani A, Tilmont R, Chauvet P, Gower N, Sklavenitis-Pistofidis R, Brinster C, Thuru X, Touil Y, Quesnel B, Mitra S, Ghobrial IM, Kluza J, Manier S. MYC dependency in GLS1 and NAMPT is a therapeutic vulnerability in multiple myeloma. iScience 2024; 27:109417. [PMID: 38510131 PMCID: PMC10952034 DOI: 10.1016/j.isci.2024.109417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/26/2023] [Accepted: 03/01/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy in which MYC alterations contribute to the malignant phenotype. Nevertheless, MYC lacks therapeutic druggability. Here, we leveraged large-scale loss-of-function screens and conducted a small molecule screen to identify genes and pathways with enhanced essentiality correlated with MYC expression. We reported a specific gene dependency in glutaminase (GLS1), essential for the viability and proliferation of MYC overexpressing cells. Conversely, the analysis of isogenic models, as well as cell lines dataset (CCLE) and patient datasets, revealed GLS1 as a non-oncogenic dependency in MYC-driven cells. We functionally delineated the differential modulation of glutamine to maintain mitochondrial function and cellular biosynthesis in MYC overexpressing cells. Furthermore, we observed that pharmaceutical inhibition of NAMPT selectively affects MYC upregulated cells. We demonstrate the effectiveness of combining GLS1 and NAMPT inhibitors, suggesting that targeting glutaminolysis and NAD synthesis may be a promising strategy to target MYC-driven MM.
Collapse
Affiliation(s)
- Lama Hasan Bou Issa
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Léa Fléchon
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - William Laine
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Aicha Ouelkdite
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Silvia Gaggero
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Adeline Cozzani
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Remi Tilmont
- Department of Hematology, CHU Lille, 59000 Lille, France
| | - Paul Chauvet
- Department of Hematology, CHU Lille, 59000 Lille, France
| | - Nicolas Gower
- Department of Hematology, CHU Lille, 59000 Lille, France
| | | | - Carine Brinster
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Xavier Thuru
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Yasmine Touil
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Bruno Quesnel
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
- Department of Hematology, CHU Lille, 59000 Lille, France
| | - Suman Mitra
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Irene M. Ghobrial
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jérôme Kluza
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
| | - Salomon Manier
- Canther, INSERM UMR-S1277 and CNRS UMR9020, Lille University, 59000 Lille, France
- Department of Hematology, CHU Lille, 59000 Lille, France
| |
Collapse
|
19
|
Lu MJ, Busquets J, Impedovo V, Wilson CN, Chan HR, Chang YT, Matsui W, Tiziani S, Cambronne XA. SLC25A51 decouples the mitochondrial NAD +/NADH ratio to control proliferation of AML cells. Cell Metab 2024; 36:808-821.e6. [PMID: 38354740 PMCID: PMC10990793 DOI: 10.1016/j.cmet.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/30/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024]
Abstract
SLC25A51 selectively imports oxidized NAD+ into the mitochondrial matrix and is required for sustaining cell respiration. We observed elevated expression of SLC25A51 that correlated with poorer outcomes in patients with acute myeloid leukemia (AML), and we sought to determine the role SLC25A51 may serve in this disease. We found that lowering SLC25A51 levels led to increased apoptosis and prolonged survival in orthotopic xenograft models. Metabolic flux analyses indicated that depletion of SLC25A51 shunted flux away from mitochondrial oxidative pathways, notably without increased glycolytic flux. Depletion of SLC25A51 combined with 5-azacytidine treatment limits expansion of AML cells in vivo. Together, the data indicate that AML cells upregulate SLC25A51 to decouple mitochondrial NAD+/NADH for a proliferative advantage by supporting oxidative reactions from a variety of fuels. Thus, SLC25A51 represents a critical regulator that can be exploited by cancer cells and may be a vulnerability for refractory AML.
Collapse
Affiliation(s)
- Mu-Jie Lu
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Jonathan Busquets
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, Austin, TX, USA
| | - Valeria Impedovo
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, Austin, TX, USA
| | - Crystal N Wilson
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Hsin-Ru Chan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Yu-Tai Chang
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA; LIVESTRONG Cancer Institutes, University of Texas at Austin, Austin, TX, USA
| | - William Matsui
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA; LIVESTRONG Cancer Institutes, University of Texas at Austin, Austin, TX, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, Austin, TX, USA; Department of Pediatrics, Dell Medical School, University of Texas at Austin, Austin, TX, USA; Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA; LIVESTRONG Cancer Institutes, University of Texas at Austin, Austin, TX, USA
| | - Xiaolu A Cambronne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA; LIVESTRONG Cancer Institutes, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
20
|
Chen Y, Chen J, Zou Z, Xu L, Li J. Crosstalk between autophagy and metabolism: implications for cell survival in acute myeloid leukemia. Cell Death Discov 2024; 10:46. [PMID: 38267416 PMCID: PMC10808206 DOI: 10.1038/s41420-024-01823-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024] Open
Abstract
Acute myeloid leukemia (AML), a prevalent form of leukemia in adults, is often characterized by low response rates to chemotherapy, high recurrence rates, and unfavorable prognosis. A critical barrier in managing refractory or recurrent AML is the resistance to chemotherapy. Increasing evidence indicates that tumor cell metabolism plays a crucial role in AML progression, survival, metastasis, and treatment resistance. Autophagy, an essential regulator of cellular energy metabolism, is increasingly recognized for its role in the metabolic reprogramming of AML. Autophagy sustains leukemia cells during chemotherapy by not only providing energy but also facilitating rapid proliferation through the supply of essential components such as amino acids and nucleotides. Conversely, the metabolic state of AML cells can influence the activity of autophagy. Their mutual coordination helps maintain intrinsic cellular homeostasis, which is a significant contributor to chemotherapy resistance in leukemia cells. This review explores the recent advancements in understanding the interaction between autophagy and metabolism in AML cells, emphasizing their roles in cell survival and drug resistance. A comprehensive understanding of the interplay between autophagy and leukemia cell metabolism can shed light on leukemia cell survival strategies, particularly under adverse conditions such as chemotherapy. This insight may also pave the way for innovative targeted treatment strategies.
Collapse
Affiliation(s)
- Yongfeng Chen
- Department of Basic Medical Sciences, Medical College of Taizhou University, 318000, Taizhou, Zhejiang, China.
| | - Jia Chen
- School of Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Zhenyou Zou
- Brain Hospital of Guangxi Zhuang Autonomous Region, 542005, Liuzhou, Guangxi, China.
| | - Linglong Xu
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, Zhejiang, China
| | - Jing Li
- Department of Histology and Embryology, North Sichuan Medical College, 637000, Nanchong, Sichuan, China
| |
Collapse
|
21
|
Tsuchimochi S, Wada-Hiraike O, Urano Y, Kukita A, Yamaguchi K, Honjo H, Taguchi A, Tanikawa M, Sone K, Mori-Uchino M, Tsuruga T, Oda K, Osuga Y. Characterization of a fluorescence imaging probe that exploits metabolic dependency of ovarian clear cell carcinoma. Sci Rep 2023; 13:20292. [PMID: 37985723 PMCID: PMC10662153 DOI: 10.1038/s41598-023-47637-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The purpose of this study is to clarify the metabolic dependence of ovarian clear cell carcinoma (CCC) by comparing normal tissues and to examine the applicability of fluorescence imaging probe to exploit these metabolic differences. Enhanced glutathione synthesis was supported by the increased uptake of related metabolites and elevated expression levels of genes. Accumulation of intracellular iron and lipid peroxide, induction of cell death by inhibition of the glutathione synthesis pathway indicated that ferroptosis was induced. The activation of γ-glutamyl hydroxymethyl rhodamine green (gGlu-HMRG), a fluorescent imaging probe that recognizes γ-glutamyl transferase, which is essential for the synthesis of glutathione, was investigated in fresh-frozen surgical specimens. gGlu-HMRG detected extremely strong fluorescent signals in the tumor lesions of CCC patients, compared to normal ovaries or endometrium. These results revealed that CCC occurs in the stressful and unique environment of free radical-rich endometrioma, and that glutathione metabolism is enhanced as an adaptation to oxidative stress. Furthermore, a modality that exploits these metabolic differences would be useful for distinguishing between CCC and normal tissues.
Collapse
Affiliation(s)
- Saki Tsuchimochi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan.
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
- CREST, Japan Agency for Medical Research and Development, Chiyoda, Tokyo, 100-0004, Japan
| | - Asako Kukita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Kohei Yamaguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Harunori Honjo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Michihiro Tanikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Mayuyo Mori-Uchino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Tetsushi Tsuruga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Katsutoshi Oda
- Department of Integrated Genomics, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| |
Collapse
|
22
|
Alqahtani A, Wang M, Lou M, Alachkar H. Genomics and transcriptomic alterations of the glutamate receptors in acute myeloid leukemia. Clin Transl Sci 2023; 16:1828-1841. [PMID: 37670476 PMCID: PMC10582680 DOI: 10.1111/cts.13588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 09/07/2023] Open
Abstract
Glutamine and glutamate have been widely explored as potential therapeutic targets in acute myeloid leukemia (AML). In addition to its bioenergetic role in leukemia cell proliferation, L-glutamate is a neurotransmitter that acts on glutamate receptors. However, the role of glutamate receptors in AML is largely understudied. Here, we comprehensively analyze the genomic and transcriptomic alterations of glutamate receptor genes in AML using publicly available data. We investigated the frequency of mutations in the glutamate receptor genes and whether an association exist between the presence of these mutations and clinical and molecular characteristics or patient's clinical outcome. We also assessed the dysregulation of glutamate receptor gene expression in AML with and without mutations and whether gene dysregulation is associated with clinical outcomes. We found that 29 (14.5%) of 200 patients with AML had a mutation in at least one glutamate receptor gene. The DNMT3A mutations were significantly more frequent in patients with mutations in at least one glutamate receptor gene compared with patients without mutations (13 of 29 [44.8%] vs. 41 of 171 [23.9%], p value: 0.02). Notably, patients with mutations in at least one glutamate receptor gene survived shorter than patients without mutations; however, the results did not reach statistical significance (overall survival: 15.5 vs. 19.0 months; p value: 0.10). Mutations in the glutamate receptor genes were not associated with changes in gene expression and the transcriptomic levels of glutamate receptor genes were not associated with clinical outcome.
Collapse
Affiliation(s)
- Amani Alqahtani
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Clinical Pharmacy, School of PharmacyNajran UniversityNajranSaudi Arabia
| | - Mengxi Wang
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mimi Lou
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Houda Alachkar
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- USC Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
23
|
Pendleton KE, Wang K, Echeverria GV. Rewiring of mitochondrial metabolism in therapy-resistant cancers: permanent and plastic adaptations. Front Cell Dev Biol 2023; 11:1254313. [PMID: 37779896 PMCID: PMC10534013 DOI: 10.3389/fcell.2023.1254313] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Deregulation of tumor cell metabolism is widely recognized as a "hallmark of cancer." Many of the selective pressures encountered by tumor cells, such as exposure to anticancer therapies, navigation of the metastatic cascade, and communication with the tumor microenvironment, can elicit further rewiring of tumor cell metabolism. Furthermore, phenotypic plasticity has been recently appreciated as an emerging "hallmark of cancer." Mitochondria are dynamic organelles and central hubs of metabolism whose roles in cancers have been a major focus of numerous studies. Importantly, therapeutic approaches targeting mitochondria are being developed. Interestingly, both plastic (i.e., reversible) and permanent (i.e., stable) metabolic adaptations have been observed following exposure to anticancer therapeutics. Understanding the plastic or permanent nature of these mechanisms is of crucial importance for devising the initiation, duration, and sequential nature of metabolism-targeting therapies. In this review, we compare permanent and plastic mitochondrial mechanisms driving therapy resistance. We also discuss experimental models of therapy-induced metabolic adaptation, therapeutic implications for targeting permanent and plastic metabolic states, and clinical implications of metabolic adaptations. While the plasticity of metabolic adaptations can make effective therapeutic treatment challenging, understanding the mechanisms behind these plastic phenotypes may lead to promising clinical interventions that will ultimately lead to better overall care for cancer patients.
Collapse
Affiliation(s)
- Katherine E. Pendleton
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Karen Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Gloria V. Echeverria
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
24
|
Huang S, Ren L, Beck JA, Phelps TE, Olkowski C, Ton A, Roy J, White ME, Adler S, Wong K, Cherukuri A, Zhang X, Basuli F, Choyke PL, Jagoda EM, LeBlanc AK. Exploration of Imaging Biomarkers for Metabolically-Targeted Osteosarcoma Therapy in a Murine Xenograft Model. Cancer Biother Radiopharm 2023; 38:475-485. [PMID: 37253167 PMCID: PMC10623067 DOI: 10.1089/cbr.2022.0090] [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] [Indexed: 06/01/2023] Open
Abstract
Background: Osteosarcoma (OS) is an aggressive pediatric cancer with unmet therapeutic needs. Glutaminase 1 (GLS1) inhibition, alone and in combination with metformin, disrupts the bioenergetic demands of tumor progression and metastasis, showing promise for clinical translation. Materials and Methods: Three positron emission tomography (PET) clinical imaging agents, [18F]fluoro-2-deoxy-2-D-glucose ([18F]FDG), 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT), and (2S, 4R)-4-[18F]fluoroglutamine ([18F]GLN), were evaluated in the MG63.3 human OS xenograft mouse model, as companion imaging biomarkers after treatment for 7 d with a selective GLS1 inhibitor (CB-839, telaglenastat) and metformin, alone and in combination. Imaging and biodistribution data were collected from tumors and reference tissues before and after treatment. Results: Drug treatment altered tumor uptake of all three PET agents. Relative [18F]FDG uptake decreased significantly after telaglenastat treatment, but not within control and metformin-only groups. [18F]FLT tumor uptake appears to be negatively affected by tumor size. Evidence of a flare effect was seen with [18F]FLT imaging after treatment. Telaglenastat had a broad influence on [18F]GLN uptake in tumor and normal tissues. Conclusions: Image-based tumor volume quantification is recommended for this paratibial tumor model. The performance of [18F]FLT and [18F]GLN was affected by tumor size. [18F]FDG may be useful in detecting telaglenastat's impact on glycolysis. Exploration of kinetic tracer uptake protocols is needed to define clinically relevant patterns of [18F]GLN uptake in patients receiving telaglenastat.
Collapse
Affiliation(s)
- Shan Huang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica A. Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tim E. Phelps
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Colleen Olkowski
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anita Ton
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jyoti Roy
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Margaret E. White
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Bethesda, Maryland, USA
| | - Karen Wong
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Aswini Cherukuri
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiang Zhang
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine M. Jagoda
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Patnana PK, Liu L, Frank D, Nimmagadda SC, Behrens M, Ahmed H, Xie X, Liebmann M, Wei L, Gerdemann A, Thivakaran A, Humpf HU, Klotz L, Dugas M, Varghese J, Trajkovic-Arsic M, Siveke JT, Hanenberg H, Opalka B, Dührsen U, Reinhardt HC, Guenther U, von Bubnoff N, Khandanpour C. Dose-dependent expression of GFI1 alters metabolism in the haematopoietic progenitors and MLL::AF9-induced leukaemic cells. Br J Haematol 2023; 202:1033-1048. [PMID: 37423893 DOI: 10.1111/bjh.18939] [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: 08/30/2022] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
Growth factor independence 1 (GFI1) is a transcriptional repressor protein that plays an essential role in the differentiation of myeloid and lymphoid progenitors. We and other groups have shown that GFI1 has a dose-dependent role in the initiation, progression, and prognosis of acute myeloid leukaemia (AML) patients by inducing epigenetic changes. We now demonstrate a novel role for dose-dependent GFI1 expression in regulating metabolism in haematopoietic progenitor and leukaemic cells. Using in-vitro and ex-vivo murine models of MLL::AF9-induced human AML and extra-cellular flux assays, we now demonstrate that a lower GFI1 expression enhances oxidative phosphorylation rate via upregulation of the FOXO1- MYC axis. Our findings underscore the significance of therapeutic exploitation in GFI1-low-expressing leukaemia cells by targeting oxidative phosphorylation and glutamine metabolism.
Collapse
Affiliation(s)
- Pradeep Kumar Patnana
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Longlong Liu
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Daria Frank
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Subbaiah Chary Nimmagadda
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Matthias Behrens
- Institute of Food Chemistry, University of Muenster, Muenster, Germany
| | - Helal Ahmed
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Xiaoqing Xie
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Marie Liebmann
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany
| | - Lanying Wei
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Institute of Medical Informatics, University of Muenster, Muenster, Germany
| | - Andrea Gerdemann
- Institute of Food Chemistry, University of Muenster, Muenster, Germany
| | | | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Muenster, Muenster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Muenster, Muenster, Germany
| | - Marija Trajkovic-Arsic
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Helmut Hanenberg
- Clinic for Pediatrics III, University Hospital Essen, Essen, Germany
- Pediatric Oncology, Hematology & Immunology, Heinrich Heine University, University Hospital Düsseldorf, Dusseldorf, Germany
| | - Bertram Opalka
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Guenther
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| |
Collapse
|
26
|
Vanhooren J, Dobbelaere R, Derpoorter C, Deneweth L, Van Camp L, Uyttebroeck A, De Moerloose B, Lammens T. CAR-T in the Treatment of Acute Myeloid Leukemia: Barriers and How to Overcome Them. Hemasphere 2023; 7:e937. [PMID: 37674860 PMCID: PMC10479376 DOI: 10.1097/hs9.0000000000000937] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/26/2023] [Indexed: 09/08/2023] Open
Abstract
Conventional therapies for acute myeloid leukemia (AML) are characterized by high rates of relapse, severe toxicities, and poor overall survival rates. Thus, the development of new therapeutic strategies is crucial for improving the survival and quality of life of AML patients. CD19-directed chimeric antigen receptor (CAR) T-cell immunotherapy has been extremely successful in the treatment of B-cell acute lymphoid leukemia and several mature B-cell lymphomas. However, the use of CAR T-cell therapy for AML is currently prevented due to the lack of a myeloid equivalent to CD19, as currently known cell surface targets on leukemic blasts are also expressed on healthy hematopoietic stem and progenitor cells as well as their progeny. In addition, the immunosuppressive tumor microenvironment has a dampening effect on the antitumor activity of CAR-T cells. Here, we review the therapeutic challenges limiting the use of CAR T-cell therapy for AML and discuss promising novel strategies to overcome them.
Collapse
Affiliation(s)
- Jolien Vanhooren
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
- Cancer Research Institute Ghent, Belgium
| | - Rani Dobbelaere
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
| | - Charlotte Derpoorter
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
- Cancer Research Institute Ghent, Belgium
| | - Larissa Deneweth
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
- Cancer Research Institute Ghent, Belgium
| | - Laurens Van Camp
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
- Cancer Research Institute Ghent, Belgium
| | - Anne Uyttebroeck
- Department of Pediatric Hematology and Oncology, University Hospitals Leuven, Department of Oncology, KU Leuven, Belgium
| | - Barbara De Moerloose
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
- Cancer Research Institute Ghent, Belgium
| | - Tim Lammens
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium
- Cancer Research Institute Ghent, Belgium
| |
Collapse
|
27
|
Maffeo B, Panuzzo C, Moraca A, Cilloni D. A Leukemic Target with a Thousand Faces: The Mitochondria. Int J Mol Sci 2023; 24:13069. [PMID: 37685874 PMCID: PMC10487524 DOI: 10.3390/ijms241713069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
In the era of personalized medicine greatly improved by molecular diagnosis and tailor-made therapies, the survival rate of acute myeloid leukemia (AML) at 5 years remains unfortunately low. Indeed, the high heterogeneity of AML clones with distinct metabolic and molecular profiles allows them to survive the chemotherapy-induced changes, thus leading to resistance, clonal evolution, and relapse. Moreover, leukemic stem cells (LSCs), the quiescent reservoir of residual disease, can persist for a long time and activate the recurrence of disease, supported by significant metabolic differences compared to AML blasts. All these points highlight the relevance to develop combination therapies, including metabolism inhibitors to improve treatment efficacy. In this review, we summarized the metabolic differences in AML blasts and LSCs, the molecular pathways related to mitochondria and metabolism are druggable and targeted in leukemia therapies, with a distinct interest for Venetoclax, which has revolutionized the therapeutic paradigms of several leukemia subtype, unfit for intensive treatment regimens.
Collapse
Affiliation(s)
| | - Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (B.M.); (A.M.); (D.C.)
| | | | | |
Collapse
|
28
|
Xiao Y, Hu B, Guo Y, Zhang D, Zhao Y, Chen Y, Li N, Yu L. Targeting Glutamine Metabolism as an Attractive Therapeutic Strategy for Acute Myeloid Leukemia. Curr Treat Options Oncol 2023:10.1007/s11864-023-01104-0. [PMID: 37249801 PMCID: PMC10356674 DOI: 10.1007/s11864-023-01104-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2023] [Indexed: 05/31/2023]
Abstract
OPINION STATEMENT Relapse after chemotherapy and hematopoietic stem cell transplantation leads to adverse prognosis for acute myeloid leukemia (AML) patients. As a "conditionally essential amino acid," glutamine contributes to the growth and proliferation of AML cells. Glutamine-target strategies as new treatment approaches have been widely explored in AML treatment to improve outcome. Glutamine-target strategies including depletion of systemic glutamine and application of glutamine uptake inhibitors, glutamine antagonists/analogues, and glutaminase inhibitors. Because glutamine metabolism involved in multiple pathways in cells and each pathway of glutamine metabolism has many regulatory factors, therefore, AML therapy targeting glutamine metabolism should focus on how to inhibit multiple metabolic pathways without affecting normal cells and host immune to achieve effective treatment for AML.
Collapse
Affiliation(s)
- Yan Xiao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Bingbing Hu
- Reproductive Medicine Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yuming Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Na Li
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Nephrology, Center of Kidney and Urology, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Liuting Yu
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
29
|
Alberti G, Arsuffi C, Pievani A, Salerno D, Mantegazza F, Dazzi F, Biondi A, Tettamanti S, Serafini M. Engineering tandem CD33xCD146 CAR CIK (cytokine-induced killer) cells to target the acute myeloid leukemia niche. Front Immunol 2023; 14:1192333. [PMID: 37304257 PMCID: PMC10247966 DOI: 10.3389/fimmu.2023.1192333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
In acute myeloid leukemia (AML), malignant stem cells hijack the normal bone marrow niche where they are largely protected from the current therapeutic approaches. Thus, eradicating these progenitors is the ultimate challenge in the treatment of this disease. Specifically, the development of chimeric antigen receptors (CARs) against distinct mesenchymal stromal cell subpopulations involved in the maintenance of leukemic stem cells within the malignant bone marrow microenvironment could represent a new strategy to improve CAR T-cell therapy efficacy, which is still unsuccessful in AML. As a proof of concept, we generated a novel prototype of Tandem CAR, with one specificity directed against the leukemic cell marker CD33 and the other against the mesenchymal stromal cell marker CD146, demonstrating its capability of simultaneously targeting two different cell types in a 2D co-culture system. Interestingly, we could also observe an in vitro inhibition of CAR T cell functionality mediated by stromal cells, particularly in later effector functions, such as reduction of interferon-gamma and interleukin-2 release and impaired proliferation of the CAR+ effector Cytokine-Induced Killer (CIK) cells. Taken together, these data demonstrate the feasibility of a dual targeting model against two molecules, which are expressed on two different target cells, but also highlight the immunomodulatory effect on CAR CIK cells exerted by stromal cells, confirming that the niche could be an obstacle to the efficacy of CAR T cells. This aspect should be considered in the development of novel CAR T cell approaches directed against the AML bone marrow niche.
Collapse
Affiliation(s)
- Gaia Alberti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Corinne Arsuffi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alice Pievani
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Domenico Salerno
- School of Medicine and Surgery, BioNanoMedicine Center NANOMIB, Universita di Milano-Bicocca, Vedano al Lambro (MB), Italy
| | - Francesco Mantegazza
- School of Medicine and Surgery, BioNanoMedicine Center NANOMIB, Universita di Milano-Bicocca, Vedano al Lambro (MB), Italy
| | - Francesco Dazzi
- School of Cardiovascular Sciences, King’s College London, London, United Kingdom
| | - Andrea Biondi
- Dipartimento di Medicina e Chirurgia, Università degli Studi Milano-Bicocca, Monza (MB), Italy
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Marta Serafini
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
30
|
Solomou G, Finch A, Asghar A, Bardella C. Mutant IDH in Gliomas: Role in Cancer and Treatment Options. Cancers (Basel) 2023; 15:cancers15112883. [PMID: 37296846 DOI: 10.3390/cancers15112883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Altered metabolism is a common feature of many cancers and, in some cases, is a consequence of mutation in metabolic genes, such as the ones involved in the TCA cycle. Isocitrate dehydrogenase (IDH) is mutated in many gliomas and other cancers. Physiologically, IDH converts isocitrate to α-ketoglutarate (α-KG), but when mutated, IDH reduces α-KG to D2-hydroxyglutarate (D2-HG). D2-HG accumulates at elevated levels in IDH mutant tumours, and in the last decade, a massive effort has been made to develop small inhibitors targeting mutant IDH. In this review, we summarise the current knowledge about the cellular and molecular consequences of IDH mutations and the therapeutic approaches developed to target IDH mutant tumours, focusing on gliomas.
Collapse
Affiliation(s)
- Georgios Solomou
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Division of Academic Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Alina Finch
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Asim Asghar
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Chiara Bardella
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
31
|
Timofeeva N, Ayres ML, Baran N, Santiago-O’Farrill JM, Bildik G, Lu Z, Konopleva M, Gandhi V. Preclinical investigations of the efficacy of the glutaminase inhibitor CB-839 alone and in combinations in chronic lymphocytic leukemia. Front Oncol 2023; 13:1161254. [PMID: 37228498 PMCID: PMC10203524 DOI: 10.3389/fonc.2023.1161254] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Chronic lymphocytic leukemia (CLL) cells are metabolically flexible and adapt to modern anticancer treatments. Bruton tyrosine kinase (BTK) and B-cell lymphoma-2 (BCL-2) inhibitors have been widely used to treat CLL, but CLL cells become resistant to these treatments over time. CB-839 is a small-molecule glutaminase-1 (GLS-1) inhibitor that impairs glutamine use, disrupts downstream energy metabolism, and impedes the elimination of reactive oxygen species. Methods To investigate the in vitro effects of CB-839 on CLL cells, we tested CB-839 alone and in combination with ibrutinib, venetoclax, or AZD-5991 on the HG-3 and MEC-1 CLL cell lines and on primary CLL lymphocytes. Results We found that CB-839 caused dose-dependent decreases in GLS-1 activity and glutathione synthesis. CB-839-treated cells also showed increased mitochondrial superoxide metabolism and impaired energy metabolism, which were reflected in decreases in the oxygen consumption rate and depletion of the adenosine triphosphate pool and led to the inhibition of cell proliferation. In the cell lines, CB-839 combined with venetoclax or AZD-5991, but not with ibrutinib, demonstrated synergism with an increased apoptosis rate and cell proliferation inhibition. In the primary lymphocytes, no significant effects of CB-839 alone or in combination with venetoclax, ibrutinib, or AZD-5991 were observed. Discussion Our findings suggest that CB-839 has limited efficacy in CLL treatment and shows limited synergy in combination with widely used CLL drugs.
Collapse
Affiliation(s)
- Natalia Timofeeva
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mary L. Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Janice M. Santiago-O’Farrill
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
32
|
Konopleva M, DiNardo C, Bhagat T, Baran N, Lodi A, Saxena K, Cai T, Su X, Skwarska A, Guerra V, Kuruvilla V, Konoplev S, Gordon-Mitchell S, Pradhan K, Aluri S, Collins M, Sweeney S, Busquet J, Rathore A, Deng Q, Green M, Grant S, Demo S, Choudhary G, Sahu S, Agarwal B, Spodek M, Thiruthuvanathan V, Will B, Steidl U, Tippett G, Burger J, Borthakur G, Jabbour E, Pemmaraju N, Kadia T, Komblau S, Daver N, Naqvi K, Short N, Garcia-Manero G, Tiziani S, Verma A. Glutaminase inhibition in combination with azacytidine in myelodysplastic syndromes: Clinical efficacy and correlative analyses. RESEARCH SQUARE 2023:rs.3.rs-2518774. [PMID: 36865338 PMCID: PMC9980221 DOI: 10.21203/rs.3.rs-2518774/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Malignancies can become reliant on glutamine as an alternative energy source and as a facilitator of aberrant DNA methylation, thus implicating glutaminase (GLS) as a potential therapeutic target. We demonstrate preclinical synergy of telaglenastat (CB-839), a selective GLS inhibitor, when combined with azacytidine (AZA), in vitro and in vivo, followed by a phase Ib/II study of the combination in patients with advanced MDS. Treatment with telaglenastat/AZA led to an ORR of 70% with CR/mCRs in 53% patients and a median overall survival of 11.6 months. scRNAseq and flow cytometry demonstrated a myeloid differentiation program at the stem cell level in clinical responders. Expression of non-canonical glutamine transporter, SLC38A1, was found to be overexpressed in MDS stem cells; was associated with clinical responses to telaglenastat/AZA and predictive of worse prognosis in a large MDS cohort. These data demonstrate the safety and efficacy of a combined metabolic and epigenetic approach in MDS.
Collapse
Affiliation(s)
| | | | | | | | - Alessia Lodi
- College of Natural Sciences, The University of Texas at Austin
| | - Kapil Saxena
- The University of Texas, MD Anderson Cancer Center
| | - Tianyu Cai
- The University of Texas, MD Anderson Cancer Center
| | - Xiaoping Su
- Dan L. Duncan Cancer Center and , Baylor College of Medicine
| | - Anna Skwarska
- Albert Einstein College of Medicine-Montefiore Medical Center
| | | | | | | | | | | | | | - Meghan Collins
- College of Natural Sciences, The University of Texas at Austin
| | - Shannon Sweeney
- Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | | | - Atul Rathore
- Dell Medical School, The University of Texas at Austin
| | - Qing Deng
- The University of Texas MD Anderson Cancer Cent
| | | | - Steven Grant
- Department of Medicine, Virginia Commonwealth University
| | | | | | | | | | - Mason Spodek
- Albert Einstein College of Medicine-Montefiore Medical Center
| | | | | | | | | | | | | | | | | | - Tapan Kadia
- The University of Texas MD Anderson Cancer Center
| | | | - Naval Daver
- The University of Texas MD Anderson Cancer Center
| | - Kiran Naqvi
- The University of Texas, MD Anderson Cancer Center
| | | | | | - Stefano Tiziani
- Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | | |
Collapse
|
33
|
The "Superoncogene" Myc at the Crossroad between Metabolism and Gene Expression in Glioblastoma Multiforme. Int J Mol Sci 2023; 24:ijms24044217. [PMID: 36835628 PMCID: PMC9966483 DOI: 10.3390/ijms24044217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The concept of the Myc (c-myc, n-myc, l-myc) oncogene as a canonical, DNA-bound transcription factor has consistently changed over the past few years. Indeed, Myc controls gene expression programs at multiple levels: directly binding chromatin and recruiting transcriptional coregulators; modulating the activity of RNA polymerases (RNAPs); and drawing chromatin topology. Therefore, it is evident that Myc deregulation in cancer is a dramatic event. Glioblastoma multiforme (GBM) is the most lethal, still incurable, brain cancer in adults, and it is characterized in most cases by Myc deregulation. Metabolic rewiring typically occurs in cancer cells, and GBM undergoes profound metabolic changes to supply increased energy demand. In nontransformed cells, Myc tightly controls metabolic pathways to maintain cellular homeostasis. Consistently, in Myc-overexpressing cancer cells, including GBM cells, these highly controlled metabolic routes are affected by enhanced Myc activity and show substantial alterations. On the other hand, deregulated cancer metabolism impacts Myc expression and function, placing Myc at the intersection between metabolic pathway activation and gene expression. In this review paper, we summarize the available information on GBM metabolism with a specific focus on the control of the Myc oncogene that, in turn, rules the activation of metabolic signals, ensuring GBM growth.
Collapse
|
34
|
Chianese U, Papulino C, Ali A, Ciardiello F, Cappabianca S, Altucci L, Carafa V, Benedetti R. FASN multi-omic characterization reveals metabolic heterogeneity in pancreatic and prostate adenocarcinoma. J Transl Med 2023; 21:32. [PMID: 36650542 PMCID: PMC9847120 DOI: 10.1186/s12967-023-03874-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) and prostate cancer (PCa) are among the most prevalent malignant tumors worldwide. There is now a comprehensive understanding of metabolic reprogramming as a hallmark of cancer. Fatty acid synthase (FASN) is a key regulator of the lipid metabolic network, providing energy to favor tumor proliferation and development. Whereas the biological role of FASN is known, its response and sensitivity to inhibition have not yet been fully established in these two cancer settings. METHODS To evaluate the association between FASN expression, methylation, prognosis, and mutational profile in PDAC and PCa, we interrogated public databases and surveyed online platforms using TCGA data. The STRING database was used to investigate FASN interactors, and the Gene Set Enrichment Analysis platform Reactome database was used to perform an enrichment analysis using data from RNA sequencing public databases of PDAC and PCa. In vitro models using PDAC and PCa cell lines were used to corroborate the expression of FASN, as shown by Western blot, and the effects of FASN inhibition on cell proliferation/cell cycle progression and mitochondrial respiration were investigated with MTT, colony formation assay, cell cycle analysis and MitoStress Test. RESULTS The expression of FASN was not modulated in PDAC compared to normal pancreatic tissues, while it was overexpressed in PCa, which also displayed a different level of promoter methylation. Based on tumor grade, FASN expression decreased in advanced stages of PDAC, but increased in PCa. A low incidence of FASN mutations was found for both tumors. FASN was overexpressed in PCa, despite not reaching statistical significance, and was associated with a worse prognosis than in PDAC. The biological role of FASN interactors correlated with lipid metabolism, and GSEA indicated that lipid-mediated mitochondrial respiration was enriched in PCa. Following validation of FASN overexpression in PCa compared to PDAC in vitro, we tested TVB-2640 as a FASN inhibitor. PCa proliferation arrest was modulated by FASN inhibition in a dose- and time-dependent manner, whereas PDAC proliferation was not altered. In line with this finding, mitochondrial respiration was found to be more affected in PCa than in PDAC. FASN inhibition interfered with metabolic signaling causing lipid accumulation and affecting cell viability with an impact on the replicative processes. CONCLUSIONS FASN exhibited differential expression patterns in PDAC and PCa, suggesting a different evolution during cancer progression. This was corroborated by the fact that both tumors responded differently to FASN inhibition in terms of proliferative potential and mitochondrial respiration, indicating that its use should reflect context specificity.
Collapse
Affiliation(s)
- Ugo Chianese
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy
| | - Chiara Papulino
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy
| | - Ahmad Ali
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy
| | - Fortunato Ciardiello
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy
| | - Salvatore Cappabianca
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy
| | - Lucia Altucci
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy ,grid.428067.f0000 0004 4674 1402Biogem Institute of Molecular and Genetic Biology, 83031 Ariano Irpino, Italy ,grid.429047.c0000 0004 6477 0469IEOS, Institute for Endocrinology and Oncology “Gaetano Salvatore”, 80131 Naples, Italy
| | - Vincenzo Carafa
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy ,grid.428067.f0000 0004 4674 1402Biogem Institute of Molecular and Genetic Biology, 83031 Ariano Irpino, Italy
| | - Rosaria Benedetti
- grid.9841.40000 0001 2200 8888Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, L. De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
35
|
Zhang W, Liu B, Wu S, Zhao L. TMT-based comprehensive proteomic profiling identifies serum prognostic signatures of acute myeloid leukemia. Open Med (Wars) 2023; 18:20220602. [PMID: 37016705 PMCID: PMC10066874 DOI: 10.1515/med-2022-0602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/19/2022] [Accepted: 10/16/2022] [Indexed: 04/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is classified into favorable-risk, intermediate-risk, and poor-risk subtypes. This study aimed to compare the serum proteomic signatures of the three AML subtypes and identify prognostic biomarkers for AML. Serum samples from patients with favorable-risk (n = 14), intermediate-risk (n = 19), and poor-risk AMLs (n = 18) were used for the analysis of tandem mass tag (TMT) labeling-based quantitative proteomics. Comparative analysis was performed to identify differentially expressed proteins (DEPs) between groups. Prognostic proteins were screened using binary logistics regression analysis. TMT-MS/MS proteomics analysis identified 138 DEPs. Fumarate hydratase (FH), isocitrate dehydrogenase 2 (IDH2), and enolase 1 (ENO1) were significantly upregulated in poor-risk patients compared with favorable-risk patients. ELISA assay confirmed that patients with poor-risk AMLs had higher levels of IDH2, ENO1, and FH compared with intermediate-risk AML patients. Logistics analysis identified that proteins 3-hydroxyacyl-CoA dehydrogenase type-2 (HADH, odds ratio (OR) = 1.035, p = 0.010), glutamine synthetase (GLUL, OR = 1.022, p = 0.039), and lactotransferrin (LTF, OR = 1.1224, p = 0.016) were associated with poor prognosis, and proteins ENO1 (OR = 1.154, p = 0.053), FH (OR = 1.043, p = 0.059), and IDH2 (OR = 3.350, p = 0.055) were associated with AML prognosis. This study showed that AML patients had elevated levels of FH, IDH2, ENO1, LTF, and GLUL proteins and might be at high risk of poor prognosis.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Central Laboratory, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Bei Liu
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Shiwen Wu
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Li Zhao
- Department of Central Laboratory, The First Hospital of Lanzhou University, #1 Donggang West Road, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
36
|
Wang K, Yang T, Zhang Y, Gao X, Tao L. The opportunities and challenges for nutritional intervention in childhood cancers. Front Nutr 2023; 10:1091067. [PMID: 36925958 PMCID: PMC10012036 DOI: 10.3389/fnut.2023.1091067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Diet dictates nutrient availability in the tumor microenvironment, thus affecting tumor metabolic activity and growth. Intrinsically, tumors develop unique metabolic features and are sensitive to environmental nutrient concentrations. Tumor-driven nutrient dependencies provide opportunities to control tumor growth by nutritional restriction or supplementation. This review summarized the existing data on nutrition and pediatric cancers after systematically searching articles up to 2023 from four databases (PubMed, Web of Science, Scopus, and Ovid MEDLINE). Epidemiological studies linked malnutrition with advanced disease stages and poor clinical outcomes in pediatric cancer patients. Experimental studies identified several nutrient dependencies (i.e., amino acids, lipids, vitamins, etc.) in major pediatric cancer types. Dietary modifications such as calorie restriction, ketogenic diet, and nutrient restriction/supplementation supported pediatric cancer treatment, but studies remain limited. Future research should expand epidemiological studies through data sharing and multi-institutional collaborations and continue to discover critical and novel nutrient dependencies to find optimal nutritional approaches for pediatric cancer patients.
Collapse
Affiliation(s)
- Kaiyue Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yubin Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Xiang Gao
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Ling Tao
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Gong T, Zheng C, Ou X, Zheng J, Yu J, Chen S, Duan Y, Liu W. Glutamine metabolism in cancers: Targeting the oxidative homeostasis. Front Oncol 2022; 12:994672. [PMID: 36324588 PMCID: PMC9621616 DOI: 10.3389/fonc.2022.994672] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Glutamine is the most abundant amino acid in blood and tissues, and the most important nutrient except for glucose in cancer cells. Over the past years, most studies have focused on the role of Gln metabolism in supporting energy metabolism rather than maintaining oxidative homeostasis. In fact, Gln is an important factor in maintaining oxidative homeostasis of cancer cells, especially in “Glutamine addicted” cancer cells. Here, this paper will review the recent scientific literature about the link between Gln metabolism and oxidative homeostasis, with an emphasis on the potential role of Gln metabolism in different cancers. Given that oxidative homeostasis is of critical importance in cancer, understanding the impacts of a Gln metabolism on oxidative homeostasis, gaining great insights into underlying molecular mechanisms, and developing effective therapeutic strategies are of great importance.
Collapse
Affiliation(s)
- Tengfang Gong
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Changbing Zheng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Xidan Ou
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuyu Chen
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Yehui Duan, ; Wei Liu,
| | - Wei Liu
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- *Correspondence: Yehui Duan, ; Wei Liu,
| |
Collapse
|
38
|
Patel SB, Nemkov T, D'Alessandro A, Welner RS. Deciphering Metabolic Adaptability of Leukemic Stem Cells. Front Oncol 2022; 12:846149. [PMID: 35756656 PMCID: PMC9213881 DOI: 10.3389/fonc.2022.846149] [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: 12/30/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Therapeutic targeting of leukemic stem cells is widely studied to control leukemia. An emerging approach gaining popularity is altering metabolism as a potential therapeutic opportunity. Studies have been carried out on hematopoietic and leukemic stem cells to identify vulnerable pathways without impacting the non-transformed, healthy counterparts. While many metabolic studies have been conducted using stem cells, most have been carried out in vitro or on a larger population of progenitor cells due to challenges imposed by the low frequency of stem cells found in vivo. This creates artifacts in the studies carried out, making it difficult to interpret and correlate the findings to stem cells directly. This review discusses the metabolic difference seen between hematopoietic stem cells and leukemic stem cells across different leukemic models. Moreover, we also shed light on the advancements of metabolic techniques and current limitations and areas for additional research of the field to study stem cell metabolism.
Collapse
Affiliation(s)
- Sweta B Patel
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at, Birmingham, AL, United States.,Divison of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Robert S Welner
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at, Birmingham, AL, United States
| |
Collapse
|
39
|
Simonaggio A, Auvray-Kuentz M, Rochand A, Thibault C, Gervais C, Oudard S, Vano YA. Future treatment options in metastatic clear cell renal cell carcinoma. Bull Cancer 2022; 109:2S47-2S58. [DOI: 10.1016/s0007-4551(22)00238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
40
|
Zhang H, Puviindran V, Nadesan P, Ding X, Shen L, Tang YJ, Tsushima H, Yahara Y, Ban GI, Zhang GF, Karner CM, Alman BA. Distinct Roles of Glutamine Metabolism in Benign and Malignant Cartilage Tumors With IDH Mutations. J Bone Miner Res 2022; 37:983-996. [PMID: 35220602 PMCID: PMC9314601 DOI: 10.1002/jbmr.4532] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/02/2022] [Accepted: 02/21/2022] [Indexed: 12/04/2022]
Abstract
Enchondromas and chondrosarcomas are common cartilage neoplasms that are either benign or malignant, respectively. The majority of these tumors harbor mutations in either IDH1 or IDH2. Glutamine metabolism has been implicated as a critical regulator of tumors with IDH mutations. Using genetic and pharmacological approaches, we demonstrated that glutaminase-mediated glutamine metabolism played distinct roles in enchondromas and chondrosarcomas with IDH1 or IDH2 mutations. Glutamine affected cell differentiation and viability in these tumors differently through different downstream metabolites. During murine enchondroma-like lesion development, glutamine-derived α-ketoglutarate promoted hypertrophic chondrocyte differentiation and regulated chondrocyte proliferation. Deletion of glutaminase in chondrocytes with Idh1 mutation increased the number and size of enchondroma-like lesions. In contrast, pharmacological inhibition of glutaminase in chondrosarcoma xenografts reduced overall tumor burden partially because glutamine-derived non-essential amino acids played an important role in preventing cell apoptosis. This study demonstrates that glutamine metabolism plays different roles in tumor initiation and cancer maintenance. Supplementation of α-ketoglutarate and inhibiting GLS may provide a therapeutic approach to suppress enchondroma and chondrosarcoma tumor growth, respectively. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | | | | | - Xiruo Ding
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Leyao Shen
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuning J Tang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Yasuhito Yahara
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Ga I Ban
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC, USA
| | - Courtney M Karner
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin A Alman
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
41
|
Lu X, Hackman GL, Saha A, Rathore AS, Collins M, Friedman C, Yi SS, Matsuda F, DiGiovanni J, Lodi A, Tiziani S. Metabolomics-based phenotypic screens for evaluation of drug synergy via direct-infusion mass spectrometry. iScience 2022; 25:104221. [PMID: 35494234 PMCID: PMC9046262 DOI: 10.1016/j.isci.2022.104221] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Drugs used in combination can synergize to increase efficacy, decrease toxicity, and prevent drug resistance. While conventional high-throughput screens that rely on univariate data are incredibly valuable to identify promising drug candidates, phenotypic screening methodologies could be beneficial to provide deep insight into the molecular response of drug combination with a likelihood of improved clinical outcomes. We developed a high-content metabolomics drug screening platform using stable isotope-tracer direct-infusion mass spectrometry that informs an algorithm to determine synergy from multivariate phenomics data. Using a cancer drug library, we validated the drug screening, integrating isotope-enriched metabolomics data and computational data mining, on a panel of prostate cell lines and verified the synergy between CB-839 and docetaxel both in vitro (three-dimensional model) and in vivo. The proposed unbiased metabolomics screening platform can be used to rapidly generate phenotype-informed datasets and quantify synergy for combinatorial drug discovery.
Collapse
Affiliation(s)
- Xiyuan Lu
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - G. Lavender Hackman
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Achinto Saha
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin,TX 78712, USA
| | - Atul Singh Rathore
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Meghan Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Chelsea Friedman
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin,TX 78712, USA
| | - S. Stephen Yi
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78723, USA,Institute for Cellular and Molecular Biology (ICMB), College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA,Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX 78712, USA
| | - Fumio Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - John DiGiovanni
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin,TX 78712, USA,Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78723, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA,Corresponding author
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA,Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78723, USA,Institute for Cellular and Molecular Biology (ICMB), College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA,Corresponding author
| |
Collapse
|
42
|
Carbone D, Vestuto V, Ferraro MR, Ciaglia T, Pecoraro C, Sommella E, Cascioferro S, Salviati E, Novi S, Tecce MF, Amodio G, Iraci N, Cirrincione G, Campiglia P, Diana P, Bertamino A, Parrino B, Ostacolo C. Metabolomics-assisted discovery of a new anticancer GLS-1 inhibitor chemotype from a nortopsentin-inspired library: From phenotype screening to target identification. Eur J Med Chem 2022; 234:114233. [DOI: 10.1016/j.ejmech.2022.114233] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/21/2022]
|
43
|
Mesbahi Y, Trahair TN, Lock RB, Connerty P. Exploring the Metabolic Landscape of AML: From Haematopoietic Stem Cells to Myeloblasts and Leukaemic Stem Cells. Front Oncol 2022; 12:807266. [PMID: 35223487 PMCID: PMC8867093 DOI: 10.3389/fonc.2022.807266] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Despite intensive chemotherapy regimens, up to 60% of adults with acute myeloid leukaemia (AML) will relapse and eventually succumb to their disease. Recent studies suggest that leukaemic stem cells (LSCs) drive AML relapse by residing in the bone marrow niche and adapting their metabolic profile. Metabolic adaptation and LSC plasticity are novel hallmarks of leukemogenesis that provide important biological processes required for tumour initiation, progression and therapeutic responses. These findings highlight the importance of targeting metabolic pathways in leukaemia biology which might serve as the Achilles' heel for the treatment of AML relapse. In this review, we highlight the metabolic differences between normal haematopoietic cells, bulk AML cells and LSCs. Specifically, we focus on four major metabolic pathways dysregulated in AML; (i) glycolysis; (ii) mitochondrial metabolism; (iii) amino acid metabolism; and (iv) lipid metabolism. We then outline established and emerging drug interventions that exploit metabolic dependencies of leukaemic cells in the treatment of AML. The metabolic signature of AML cells alters during different biological conditions such as chemotherapy and quiescence. Therefore, targeting the metabolic vulnerabilities of these cells might selectively eradicate them and improve the overall survival of patients with AML.
Collapse
Affiliation(s)
- Yashar Mesbahi
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Patrick Connerty
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| |
Collapse
|
44
|
Fan K, Liu Z, Gao M, Tu K, Xu Q, Zhang Y. Targeting Nutrient Dependency in Cancer Treatment. Front Oncol 2022; 12:820173. [PMID: 35178349 PMCID: PMC8846368 DOI: 10.3389/fonc.2022.820173] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumor. Growing evidence suggests metabolic changes that support oncogenic progression may cause selective vulnerabilities that can be exploited for cancer treatment. Increasing demands for certain nutrients under genetic determination or environmental challenge enhance dependency of tumor cells on specific nutrient, which could be therapeutically developed through targeting such nutrient dependency. Various nutrients including several amino acids and glucose have been found to induce dependency in genetic alteration- or context-dependent manners. In this review, we discuss the extensively studied nutrient dependency and the biological mechanisms behind such vulnerabilities. Besides, existing applications and strategies to target nutrient dependency in different cancer types, accompanied with remaining challenges to further exploit these metabolic vulnerabilities to improve cancer therapies, are reviewed.
Collapse
Affiliation(s)
- Kexin Fan
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Second Medical College, Karamay, China
| | - Min Gao
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
| | - Yilei Zhang
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
45
|
PPAR Ligands Induce Antiviral Effects Targeting Perturbed Lipid Metabolism during SARS-CoV-2, HCV, and HCMV Infection. BIOLOGY 2022; 11:biology11010114. [PMID: 35053112 PMCID: PMC8772958 DOI: 10.3390/biology11010114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/24/2022]
Abstract
Simple Summary The current coronavirus disease 2019 pandemic turned the attention of researchers to developing novel strategies to counteract virus infections. Despite several antiviral drugs being commercially available, there is an urgent need to identify novel molecules efficacious against viral infections that act through different mechanisms of action. In this context, our attention is focused on novel compounds acting on nuclear receptors, whose activity could be beneficial in viral infections, including coronavirus, hepatitis C virus, and cytomegalovirus. Abstract The manipulation of host metabolisms by viral infections has been demonstrated by several studies, with a marked influence on the synthesis and utilization of glucose, nucleotides, fatty acids, and amino acids. The ability of virus to perturb the metabolic status of the infected organism is directly linked to the outcome of the viral infection. A great deal of research in recent years has been focusing on these metabolic aspects, pointing at modifications induced by virus, and suggesting novel strategies to counteract the perturbed host metabolism. In this review, our attention is turned on PPARs, nuclear receptors controlling multiple metabolic actions, and on the effects played by PPAR ligands during viral infections. The role of PPAR agonists and antagonists during SARS-CoV-2, HCV, and HCMV infections will be analyzed.
Collapse
|
46
|
Lemberg KM, Gori SS, Tsukamoto T, Rais R, Slusher BS. Clinical development of metabolic inhibitors for oncology. J Clin Invest 2022; 132:e148550. [PMID: 34981784 PMCID: PMC8718137 DOI: 10.1172/jci148550] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Metabolic inhibitors have been used in oncology for decades, dating back to antimetabolites developed in the 1940s. In the past 25 years, there has been increased recognition of metabolic derangements in tumor cells leading to a resurgence of interest in targeting metabolism. More recently there has been recognition that drugs targeting tumor metabolism also affect the often acidic, hypoxic, immunosuppressive tumor microenvironment (TME) and non-tumor cell populations within it, including immune cells. Here we review small-molecule metabolic inhibitors currently in clinical development for oncology applications. For each agent, we evaluate the preclinical studies demonstrating antitumor and TME effects and review ongoing clinical trials. The goal of this Review is to provide an overview of the landscape of metabolic inhibitors in clinical development for oncology.
Collapse
Affiliation(s)
- Kathryn M. Lemberg
- Johns Hopkins Drug Discovery
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center
| | | | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery
- Department of Neurology
- Department of Pharmacology and Molecular Sciences
| | - Rana Rais
- Johns Hopkins Drug Discovery
- Department of Neurology
- Department of Pharmacology and Molecular Sciences
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center
- Department of Neurology
- Department of Pharmacology and Molecular Sciences
- Department of Medicine, and
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Chandran A, Jayasankar V, Spagnuolo P, Subramanian J. Bioactive Compounds from Curcuma amada and Their Effect on Acute Myeloid Leukemia. Crit Rev Oncog 2022; 27:23-31. [PMID: 37183936 DOI: 10.1615/critrevoncog.2023047542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer with limited chemotherapy options and negative patient outcomes. Investigations with bioactive compounds from dietary sources against cancer have increased in the recent years, which highlight the need for novel therapeutic approaches and new anti-leukemic agents possessing higher efficacy and selectivity for AML cells and fewer negative side effects. Bioactive compounds demonstrated the ability to induce cell cycle blockage and apoptosis or autophagy in cancer cells, as well as inhibition of proliferation/migration and tumor progression, etc. Bioactive compounds isolated from dietary sources such as mango ginger show promise for AML treatment. Curcuma amada roots have been used in traditional medicine and showed antioxidant, antimicrobial and anticancer properties. Bioactive molecules isolated from C. amada showed effects on the mitochondrial metabolism and reduced the viability of multiple leukemic cell lines.
Collapse
Affiliation(s)
- Ajila Chandran
- Department of Chemistry, Eastern Kentucky University, Richmond, KY, USA
| | - Varsha Jayasankar
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul Spagnuolo
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
48
|
Sharma ND, Keewan E, Matlawska-Wasowska K. Metabolic Reprogramming and Cell Adhesion in Acute Leukemia Adaptation to the CNS Niche. Front Cell Dev Biol 2021; 9:767510. [PMID: 34957100 PMCID: PMC8703109 DOI: 10.3389/fcell.2021.767510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Involvement of the Central Nervous System (CNS) in acute leukemia confers poor prognosis and lower overall survival. Existing CNS-directed therapies are associated with a significant risk of short- or long-term toxicities. Leukemic cells can metabolically adapt and survive in the microenvironment of the CNS. The supporting role of the CNS microenvironment in leukemia progression and dissemination has not received sufficient attention. Understanding the mechanism by which leukemic cells survive in the nutrient-poor and oxygen-deprived CNS microenvironment will lead to the development of more specific and less toxic therapies. Here, we review the current literature regarding the roles of metabolic reprogramming in leukemic cell adhesion and survival in the CNS.
Collapse
Affiliation(s)
- Nitesh D Sharma
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, United States
| | - Esra'a Keewan
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, United States
| | - Ksenia Matlawska-Wasowska
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
49
|
Basheer HA, Elsalem L, Salem A, Tailor A, Hunter K, Afarinkia K. The Expression of Glutaminases and their Association with Clinicopathological Parameters in the Head and Neck Cancers. Curr Cancer Drug Targets 2021; 22:169-179. [PMID: 34951574 DOI: 10.2174/1568009622666211224111425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/11/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The increased glutamine metabolism is a characteristic feature of cancer cells. The interconversion between glutamine and glutamate is catalyzed by two glutaminase isoforms, GLS1 and GLS2, which appear to have different roles in different types of cancer. We investigated for the first time the protein expression of GLS1 and GLS2, and their correlation with advanced clinicopathological parameters in head and neck cancers. METHOD Consecutive slides from a tissue microarray comprised of 80 samples ranging from normal to metastatic, were stained immunohistochemically for GLS1, GLS2, HIF-1α or CD147. Following analysis by two expert pathologists we carried out statistical analysis of the scores. RESULTS GLS1 and GLS2 are upregulated at protein level in head and neck tumours compared to normal tissues and this increased expression correlated positively (GLS1) and negatively (GLS2) with tumor grade, indicating a shift of expression between GLS enzyme isoforms based on tumor differentiation. Increased expression of GLS1 was associated with high CD147 expression; and elevated GLS2 expression was associated with both high CD147 and high HIF-1α expressions. The correlation of the GLS1 and GLS2 with HIF-1α or CD147 was strongly associated with more advanced clinicopathological parameters. CONCLUSION The increased expression of GLS1 and GLS2 may be explored as a new treatment for head and neck cancers.
Collapse
Affiliation(s)
- Haneen A Basheer
- Faculty of Pharmacy, Zarqa University, PO Box 132222, Zarqa 13132, Jordan
| | - Lina Elsalem
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| | - Anwar Salem
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, BD7 1DP, United Kingdom. 4School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Artysha Tailor
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Keith Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Kamyar Afarinkia
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, BD7 1DP, United Kingdom
| |
Collapse
|
50
|
Johnson RL, Cummings M, Thangavelu A, Theophilou G, de Jong D, Orsi NM. Barriers to Immunotherapy in Ovarian Cancer: Metabolic, Genomic, and Immune Perturbations in the Tumour Microenvironment. Cancers (Basel) 2021; 13:6231. [PMID: 34944851 PMCID: PMC8699358 DOI: 10.3390/cancers13246231] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
A lack of explicit early clinical signs and effective screening measures mean that ovarian cancer (OC) often presents as advanced, incurable disease. While conventional treatment combines maximal cytoreductive surgery and platinum-based chemotherapy, patients frequently develop chemoresistance and disease recurrence. The clinical application of immune checkpoint blockade (ICB) aims to restore anti-cancer T-cell function in the tumour microenvironment (TME). Disappointingly, even though tumour infiltrating lymphocytes are associated with superior survival in OC, ICB has offered limited therapeutic benefits. Herein, we discuss specific TME features that prevent ICB from reaching its full potential, focussing in particular on the challenges created by immune, genomic and metabolic alterations. We explore both recent and current therapeutic strategies aiming to overcome these hurdles, including the synergistic effect of combination treatments with immune-based strategies and review the status quo of current clinical trials aiming to maximise the success of immunotherapy in OC.
Collapse
Affiliation(s)
- Racheal Louise Johnson
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Michele Cummings
- Leeds Institute of Medical Research, St. James’s University Hospital, Leeds LS9 7TF, UK; (M.C.); (N.M.O.)
| | - Amudha Thangavelu
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Georgios Theophilou
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Diederick de Jong
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Nicolas Michel Orsi
- Leeds Institute of Medical Research, St. James’s University Hospital, Leeds LS9 7TF, UK; (M.C.); (N.M.O.)
| |
Collapse
|