1
|
Vagaggini C, D'Ursi P, Poggialini F, Fossa P, Francesconi V, Trombetti G, Orro A, Dreassi E, Schenone S, Tonelli M, Carbone A. Deciphering the landscape of allosteric glutaminase 1 inhibitors as anticancer agents. Bioorg Chem 2025; 161:108523. [PMID: 40311238 DOI: 10.1016/j.bioorg.2025.108523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/02/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Glutamine is the second most utilised energy source after glucose for cancer cells to support their proliferation and survival. Glutaminase 1 (GLS1) is the rate-limiting enzyme during the glutaminolysis pathway and thus represents a promising therapeutic target for the development of innovative antitumor agents. Two main classes of GLS1 inhibitors, based on their different binding mode, are reported: the substrate active site and the allosteric site inhibitors. Despite the intense efforts made to date, only two GLS1 inhibitors (i.e.,CB-839 and IPN60090) have entered clinical trials. Therefore, this research field remains to be explored to improve the effectiveness of anticancer therapy. Hence, we describe the discovery and development of reversible allosteric GLS1 inhibitors disclosed in the last six years, dividing them based on their structural similarity with bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) and CB-839. Furthermore, macrocyclic and thiadiazole derivatives, and other structurally different compounds are discussed to present a wider picture of the chemical space under investigation. The study of the binding interactions governing GLS1 inhibition is also analyzed, to help prospectively refine the structural features for greater efficacy. Interestingly, an overview of a new class of irreversible allosteric inhibitors targeting GLS1 Lys320 key residue is provided for the first time. We also summarize the most important biological studies conducted on CB-839 and IPN60090 and their significance for further assessment. The insights garnered from this paper are expected to guide future drug design endeavours toward the identification of novel therapeutics targeting GLS1 to complement and potentially enhance the arsenal of anticancer medications.
Collapse
Affiliation(s)
- Chiara Vagaggini
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Pasqualina D'Ursi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy
| | - Federica Poggialini
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Paola Fossa
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy; Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy
| | - Valeria Francesconi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy
| | - Gabriele Trombetti
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy
| | - Alessandro Orro
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Michele Tonelli
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy.
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy.
| |
Collapse
|
2
|
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
|
3
|
Cooper AJL, Denton TT. ω-Amidase and Its Substrate α-Ketoglutaramate (the α-Keto Acid Analogue of Glutamine) as Biomarkers in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1660-1680. [PMID: 39523108 DOI: 10.1134/s000629792410002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 11/16/2024]
Abstract
A large literature exists on the biochemistry, chemistry, metabolism, and clinical importance of the α-keto acid analogues of many amino acids. However, although glutamine is the most abundant amino acid in human tissues, and transamination of glutamine to its α-keto acid analogue (α-ketoglutaramate; KGM) was described more than seventy years ago, little information is available on the biological importance of KGM. Herein, we summarize the metabolic importance of KGM as an intermediate in the glutamine transaminase - ω-amidase (GTωA) pathway for the conversion of glutamine to anaplerotic α-ketoglutarate. We describe some properties of KGM, notably its occurrence as a lactam (2-hydroxy-5-oxoproline; 99.7% at pH 7.2), and its presence in normal tissues and body fluids. We note that the concentration of KGM is elevated in the cerebrospinal fluid of liver disease patients and that the urinary KGM/creatinine ratio is elevated in patients with an inborn error of the urea cycle and in patients with citrin deficiency. Recently, of the 607 urinary metabolites measured in a kidney disease study, KGM was noted to be one of five metabolites that was most significantly associated with uromodulin (a potential biomarker for tubular functional mass). Finally, we note that KGM is an intermediate in the breakdown of nicotine in certain organisms and is an important factor in nitrogen homeostasis in some microorganisms and plants. In conclusion, we suggest that biochemists and clinicians should consider KGM as (i) a key intermediate in nitrogen metabolism in all branches of life, and (ii) a biomarker, along with ω-amidase, in several diseases.
Collapse
Affiliation(s)
- Arthur J L Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Travis T Denton
- LiT Biosciences, Spokane, WA, 99202-5029, USA. ARRAY(0x5d17383a0090)
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA, USA
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University Health Sciences Spokane, Spokane, WA, USA
- Steve Gleason Institute for Neuroscience, Washington State University Health Sciences Spokane, Spokane, WA, USA
| |
Collapse
|
4
|
Feng S, Aplin C, Nguyen TTT, Milano SK, Cerione RA. Filament formation drives catalysis by glutaminase enzymes important in cancer progression. Nat Commun 2024; 15:1971. [PMID: 38438397 PMCID: PMC10912226 DOI: 10.1038/s41467-024-46351-3] [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: 07/24/2023] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
The glutaminase enzymes GAC and GLS2 catalyze the hydrolysis of glutamine to glutamate, satisfying the 'glutamine addiction' of cancer cells. They are the targets of anti-cancer drugs; however, their mechanisms of activation and catalytic activity have been unclear. Here we demonstrate that the ability of GAC and GLS2 to form filaments is directly coupled to their catalytic activity and present their cryo-EM structures which provide a view of the conformational states essential for catalysis. Filament formation guides an 'activation loop' to assume a specific conformation that works together with a 'lid' to close over the active site and position glutamine for nucleophilic attack by an essential serine. Our findings highlight how ankyrin repeats on GLS2 regulate enzymatic activity, while allosteric activators stabilize, and clinically relevant inhibitors block, filament formation that enables glutaminases to catalyze glutaminolysis and support cancer progression.
Collapse
Affiliation(s)
- Shi Feng
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Cody Aplin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Thuy-Tien T Nguyen
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Shawn K Milano
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Richard A Cerione
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA.
- Department of Molecular Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
5
|
Saad H, Nour El-Dien FA, El-Gamel NEA, Abo Dena AS. Removal of bromophenol blue from polluted water using a novel azo-functionalized magnetic nano-adsorbent. RSC Adv 2024; 14:1316-1329. [PMID: 38174277 PMCID: PMC10763660 DOI: 10.1039/d3ra04222g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Water pollution from organic dyes poses a serious danger to the environment. In the present work, we report a novel adsorbent (ADFS) based on azo-dye-functionalized superparamagnetic iron oxide nanoparticles (SPIONs) for the removal of the anionic dye bromophenol blue (BPB) from contaminated water. The fabricated SPIONs, azo dye, and ADFS adsorbent were characterized with FTIR and UV-vis absorption spectroscopy, 1HNMR spectroscopy, mass spectrometry, SEM imaging, dynamic light scattering (DLS), zeta potential measurements, vibrating sample magnetometry, thermogravimetric analysis, differential thermal analysis, and X-ray diffraction analysis. DLS measurements showed a particle size of 46.1 and 176.5 nm for the SPIONs and the ADFS, respectively. The adsorbent exhibited an adsorption capacity of 7.43 mg g-1 and followed the pseudo-second-order kinetics model (r2 = 0.9981). The ADFS could efficiently remove BPB from water after stirring for 120 minutes at room temperature and pH 2. The adsorption process was proved to occur via physisorption, as revealed by the Freundlich isotherm (n = 1.82 and KF = 11.5). Thermodynamic studies implied that the adsorption is spontaneous (-8.03 ≤ ΔG ≤ -0.58 kJ mol-1) and enthalpy-driven might take place via van der Waals interactions and/or hydrogen bonding (ΔH = -82.19 kJ mol-1 and ΔS = -0.24 kJ mol-1 K-1).
Collapse
Affiliation(s)
- Hadeel Saad
- Chemistry Department, Faculty of Science, Cairo University Giza 12613 Egypt
- General Organization for Export and Import Control Ramses Street Cairo Egypt
| | - F A Nour El-Dien
- Chemistry Department, Faculty of Science, Cairo University Giza 12613 Egypt
| | - Nadia E A El-Gamel
- Chemistry Department, Faculty of Science, Cairo University Giza 12613 Egypt
| | - Ahmed S Abo Dena
- Pharmaceutical Chemistry Department, National Organization for Drug Control and Research (NODCAR) Giza Egypt
- Faculty of Oral and Dental Medicine, Future University in Egypt (FUE) New Cairo Egypt
| |
Collapse
|
6
|
Barreca F, Aventaggiato M, Vitiello L, Sansone L, Russo MA, Mai A, Valente S, Tafani M. SIRT5 Activation and Inorganic Phosphate Binding Reduce Cancer Cell Vitality by Modulating Autophagy/Mitophagy and ROS. Antioxidants (Basel) 2023; 12:1635. [PMID: 37627630 PMCID: PMC10451763 DOI: 10.3390/antiox12081635] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/24/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer cells show increased glutamine consumption. The glutaminase (GLS) enzyme controls a limiting step in glutamine catabolism. Breast tumors, especially the triple-negative subtype, have a high expression of GLS. Our recent study demonstrated that GLS activity and ammonia production are inhibited by sirtuin 5 (SIRT5). We developed MC3138, a selective SIRT5 activator. Treatment with MC3138 mimicked the deacetylation effect mediated by SIRT5 overexpression. Moreover, GLS activity was regulated by inorganic phosphate (Pi). Considering the interconnected roles of GLS, SIRT5 and Pi in cancer growth, our hypothesis is that activation of SIRT5 and reduction in Pi could represent a valid antitumoral strategy. Treating cells with MC3138 and lanthanum acetate, a Pi chelator, decreased cell viability and clonogenicity. We also observed a modulation of MAP1LC3B and ULK1 with MC3138 and lanthanum acetate. Interestingly, inhibition of the mitophagy marker BNIP3 was observed only in the presence of MC3138. Autophagy and mitophagy modulation were accompanied by an increase in cytosolic and mitochondrial reactive oxygen species (ROS). In conclusion, our results show how SIRT5 activation and/or Pi binding can represent a valid strategy to inhibit cell proliferation by reducing glutamine metabolism and mitophagy, leading to a deleterious accumulation of ROS.
Collapse
Affiliation(s)
- Federica Barreca
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.B.); (M.A.)
| | - Michele Aventaggiato
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.B.); (M.A.)
| | - Laura Vitiello
- Laboratory of Flow Cytometry, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy;
| | - Luigi Sansone
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy; (L.S.); (M.A.R.)
- Cellular and Molecular Pathology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Matteo Antonio Russo
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy; (L.S.); (M.A.R.)
- Cellular and Molecular Pathology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (A.M.); (S.V.)
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (A.M.); (S.V.)
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.B.); (M.A.)
| |
Collapse
|
7
|
Feng S, Aplin C, Nguyen TTT, Milano SK, Cerione RA. Filament formation drives catalysis by glutaminase enzymes important in cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.16.528860. [PMID: 36824706 PMCID: PMC9949068 DOI: 10.1101/2023.02.16.528860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
The glutaminase enzymes GAC and GLS2 catalyze the hydrolysis of glutamine to glutamate, satisfying the 'glutamine addiction' of cancer cells. They are the targets of anti-cancer drugs; however, their mechanisms of activation and catalytic activity have been unclear. Here we demonstrate that the ability of GAC and GLS2 to form filaments is directly coupled to their catalytic activity and present their cryo-EM structures which provide an unprecedented view of the conformational states essential for catalysis. Filament formation guides an 'activation loop' to assume a specific conformation that works together with a 'lid' to close over the active site and position glutamine for nucleophilic attack by an essential serine. Our findings highlight how ankyrin repeats on GLS2 regulate enzymatic activity, while allosteric activators stabilize, and clinically relevant inhibitors block, filament formation that enables glutaminases to catalyze glutaminolysis and support cancer progression.
Collapse
Affiliation(s)
- Shi Feng
- Department of Chemistry and Chemical Biology, Cornell University,
Ithaca, New York, 14853
| | - Cody Aplin
- Department of Chemistry and Chemical Biology, Cornell University,
Ithaca, New York, 14853
| | - Thuy-Tien T. Nguyen
- Department of Chemistry and Chemical Biology, Cornell University,
Ithaca, New York, 14853
| | - Shawn K. Milano
- Department of Chemistry and Chemical Biology, Cornell University,
Ithaca, New York, 14853
| | - Richard A. Cerione
- Department of Chemistry and Chemical Biology, Cornell University,
Ithaca, New York, 14853
- Department of Molecular Medicine, Cornell University, Ithaca, New
York, 14853
| |
Collapse
|
8
|
Nguyen TTT, Katt WP, Cerione RA. Alone and together: current approaches to targeting glutaminase enzymes as part of anti-cancer therapies. FUTURE DRUG DISCOVERY 2023; 4:FDD79. [PMID: 37009252 PMCID: PMC10051075 DOI: 10.4155/fdd-2022-0011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 02/10/2023] [Indexed: 03/29/2023] Open
Abstract
Metabolic reprogramming is a major hallmark of malignant transformation in cancer, and part of the so-called Warburg effect, in which the upregulation of glutamine catabolism plays a major role. The glutaminase enzymes convert glutamine to glutamate, which initiates this pathway. Inhibition of different forms of glutaminase (KGA, GAC, or LGA) demonstrated potential as an emerging anti-cancer therapeutic strategy. The regulation of these enzymes, and the molecular basis for their inhibition, have been the focus of much recent research. This review will explore the recent progress in understanding the molecular basis for activation and inhibition of different forms of glutaminase, as well as the recent focus on combination therapies of glutaminase inhibitors with other anti-cancer drugs.
Collapse
Affiliation(s)
- Thuy-Tien T Nguyen
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Richard A Cerione
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
9
|
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
|
10
|
Cooper AJL, Dorai T, Pinto JT, Denton TT. α-Ketoglutaramate-A key metabolite contributing to glutamine addiction in cancer cells. Front Med (Lausanne) 2022; 13:1035335. [PMID: 36404951 PMCID: PMC9671947 DOI: 10.3389/fmed.2022.1035335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/10/2022] [Indexed: 08/27/2023] Open
Affiliation(s)
- Arthur J. L. Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Thambi Dorai
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
- Department of Urology, New York Medical College, Valhalla, NY, United States
| | - John T. Pinto
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Travis T. Denton
- Department Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA, United States
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University Health Sciences Spokane, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Washington State University Health Sciences Spokane, Spokane, WA, United States
| |
Collapse
|
11
|
L-Glutamine-, peptidyl- and protein-glutaminases: structural features and applications in the food industry. World J Microbiol Biotechnol 2022; 38:204. [PMID: 36002753 DOI: 10.1007/s11274-022-03391-5] [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: 06/27/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022]
Abstract
L-Glutaminases are enzymes that catalyze the cleavage of the gamma-amido bond of L-glutamine residues, producing ammonia and L-glutamate. These enzymes have several applications in food and pharmaceutical industries. However, the L-glutaminases that hydrolyze free L-glutamine (L-glutamine glutaminases, EC 3.5.1.2) have different structures and properties with respect to the L-glutaminases that hydrolyze the same amino acid covalently bound in peptides (peptidyl glutaminases, EC 3.5.1.43) and proteins (protein-glutamine glutaminase, EC 3.5.1.44). In the food industry, L-glutamine glutaminases are applied to enhance the flavor of foods, whereas protein glutaminases are useful to improve the functional properties of proteins. This review will focus on structural backgrounds and differences between these enzymes, the methodology available to measure the activity as well as strengths and limitations. Production methods, applications, and challenges in the food industry will be also discussed. This review will provide useful information to search and identify the suitable L-glutaminase that best fits to the intended application.
Collapse
|