1
|
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
|
2
|
Collins M, Gorgoglione R, Impedovo V, Pan X, Chakkarai S, Yi SS, Lodi A, Tiziani S. Exploration of the intracellular chiral metabolome in pediatric BCP-ALL: a pilot study investigating the metabolic phenotype of IgH locus aberrations. Front Oncol 2024; 14:1413264. [PMID: 39161381 PMCID: PMC11332069 DOI: 10.3389/fonc.2024.1413264] [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: 04/07/2024] [Accepted: 05/13/2024] [Indexed: 08/21/2024] Open
Abstract
Background and aims Aberrations in the immunoglobulin heavy chain (IgH) locus are associated with poor prognosis in pediatric precursor B-cell acute lymphoblastic leukemia (BCP-ALL) patients. The primary objective of this pilot study is to enhance our understanding of the IgH phenotype by exploring the intracellular chiral metabolome. Materials and methods Leukemia cells were isolated from the bone marrow of BCP-ALL pediatric patients at diagnosis. The samples' metabolome and transcriptome were characterized using untargeted chiral metabolomic and next-generation sequencing transcriptomic analyses. Results For the first time D- amino acids were identified in the leukemic cells' intracellular metabolome from the bone marrow niche. Chiral metabolic signatures at diagnosis was indicative of a resistant phenotype. Through integrated network analysis and Pearson correlation, confirmation was obtained regarding the association of the IgH phenotype with several genes linked to poor prognosis. Conclusion The findings of this study have contributed to the understanding that the chiral metabolome plays a role in the poor prognosis observed in an exceptionally rare patient cohort. The findings include elevated D-amino acid incorporation in the IgH group, the emergence of several unknown, potentially enantiomeric, metabolites, and insights into metabolic pathways that all warrant further exploration.
Collapse
Affiliation(s)
- Meghan Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Ruggiero Gorgoglione
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Valeria Impedovo
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Sathyaseelan Chakkarai
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - S. Stephen Yi
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Interdisciplinary Life Sciences Graduate Programs, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
3
|
Stanford SM, Nguyen TP, Chang J, Zhao Z, Hackman GL, Santelli E, Sanders CM, Katiki M, Dondossola E, Brauer BL, Diaz MA, Zhan Y, Ramsey SH, Watson PA, Sankaran B, Paindelli C, Parietti V, Mikos AG, Lodi A, Bagrodia A, Elliott A, McKay RR, Murali R, Tiziani S, Kettenbach AN, Bottini N. Targeting prostate tumor low-molecular weight tyrosine phosphatase for oxidation-sensitizing therapy. SCIENCE ADVANCES 2024; 10:eadg7887. [PMID: 38295166 PMCID: PMC10830117 DOI: 10.1126/sciadv.adg7887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024]
Abstract
Protein tyrosine phosphatases (PTPs) play major roles in cancer and are emerging as therapeutic targets. Recent reports suggest low-molecular weight PTP (LMPTP)-encoded by the ACP1 gene-is overexpressed in prostate tumors. We found ACP1 up-regulated in human prostate tumors and ACP1 expression inversely correlated with overall survival. Using CRISPR-Cas9-generated LMPTP knockout C4-2B and MyC-CaP cells, we identified LMPTP as a critical promoter of prostate cancer (PCa) growth and bone metastasis. Through metabolomics, we found that LMPTP promotes PCa cell glutathione synthesis by dephosphorylating glutathione synthetase on inhibitory Tyr270. PCa cells lacking LMPTP showed reduced glutathione, enhanced activation of eukaryotic initiation factor 2-mediated stress response, and enhanced reactive oxygen species after exposure to taxane drugs. LMPTP inhibition slowed primary and bone metastatic prostate tumor growth in mice. These findings reveal a role for LMPTP as a critical promoter of PCa growth and metastasis and validate LMPTP inhibition as a therapeutic strategy for treating PCa through sensitization to oxidative stress.
Collapse
Affiliation(s)
| | - Tiffany P. Nguyen
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Joseph Chang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Zixuan Zhao
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - G. Lavender Hackman
- Department of Nutritional Sciences, College of Natural Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Eugenio Santelli
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Kao Autoimmunity Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Colton M. Sanders
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brooke L. Brauer
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Michael A. Diaz
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yuan Zhan
- Department of Pediatrics and Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, College of Natural Sciences, The University of Texas at Austin, Austin, TX USA
| | - Sterling H. Ramsey
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Philip A. Watson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Banumathi Sankaran
- Department of Molecular Biophysics and Integrated Bioimaging, Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Claudia Paindelli
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vanessa Parietti
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Aditya Bagrodia
- Department of Urology, University of California, San Diego, La Jolla, CA, USA
| | - Andrew Elliott
- Department of Clinical and Translational Research, Caris Life Sciences, Phoenix, AZ, USA
| | - Rana R. McKay
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics and Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, College of Natural Sciences, The University of Texas at Austin, Austin, TX USA
| | - Arminja N. Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Nunzio Bottini
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Kao Autoimmunity Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
4
|
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
|
5
|
Feng B, Dong X, Liu Z, Zhang J, Liu H, Xu Y. Virtual Screening and Biological Evaluation of Novel Low Molecular Weight Protein Tyrosine Phosphatase Inhibitor for the Treatment of Insulin Resistance. Drug Des Devel Ther 2023; 17:1191-1201. [PMID: 37113468 PMCID: PMC10128076 DOI: 10.2147/dddt.s406956] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Purpose Protein tyrosine phosphatases (PTPs) play an essential way in diseases including cancer, obesity, diabetes and autoimmune disorders. As a member of PTPs, low molecular weight PTP (LMPTP) has been a well-recognized anti-insulin resistance target in obesity. However, the number of reported LMPTP inhibitors is limited. Our research aims to discover a novel LMPTP inhibitor and evaluate its biological activity against insulin resistance. Methods A virtual screening pipeline based on the X-ray co-crystal complex of LMPTP was constructed. Enzyme inhibition assay and cellular bioassay were used to evaluate the activity of screened compounds. Results The screening pipeline rendered 15 potential hits from Specs chemical library. Enzyme inhibition assay identified compound F9 (AN-465/41163730) as a potential LMPTP inhibitor with a K i value of 21.5 ± 7.3 μM. Cellular bioassay showed F9 could effectively increase the glucose consumption of HepG2 cells as a result of releasing insulin resistance by regulating PI3K-Akt pathway. Conclusion In summary, this study presents a versatile virtual screening pipeline for potential LMPTP inhibitor discovery and provides a novel-scaffold lead compound that is worthy of further modification to get more potent LMPTP inhibitors.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Xu Dong
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Zhen Liu
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Jie Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Hongyu Liu
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Correspondence: Hongyu Liu; Yuan Xu, Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China, Email ;
| | - Yuan Xu
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
6
|
Yap TA, Daver N, Mahendra M, Zhang J, Kamiya-Matsuoka C, Meric-Bernstam F, Kantarjian HM, Ravandi F, Collins ME, Francesco MED, Dumbrava EE, Fu S, Gao S, Gay JP, Gera S, Han J, Hong DS, Jabbour EJ, Ju Z, Karp DD, Lodi A, Molina JR, Baran N, Naing A, Ohanian M, Pant S, Pemmaraju N, Bose P, Piha-Paul SA, Rodon J, Salguero C, Sasaki K, Singh AK, Subbiah V, Tsimberidou AM, Xu QA, Yilmaz M, Zhang Q, Li Y, Bristow CA, Bhattacharjee MB, Tiziani S, Heffernan TP, Vellano CP, Jones P, Heijnen CJ, Kavelaars A, Marszalek JR, Konopleva M. Complex I inhibitor of oxidative phosphorylation in advanced solid tumors and acute myeloid leukemia: phase I trials. Nat Med 2023; 29:115-126. [PMID: 36658425 DOI: 10.1038/s41591-022-02103-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/21/2022] [Indexed: 01/21/2023]
Abstract
Although targeting oxidative phosphorylation (OXPHOS) is a rational anticancer strategy, clinical benefit with OXPHOS inhibitors has yet to be achieved. Here we advanced IACS-010759, a highly potent and selective small-molecule complex I inhibitor, into two dose-escalation phase I trials in patients with relapsed/refractory acute myeloid leukemia (NCT02882321, n = 17) and advanced solid tumors (NCT03291938, n = 23). The primary endpoints were safety, tolerability, maximum tolerated dose and recommended phase 2 dose (RP2D) of IACS-010759. The PK, PD, and preliminary antitumor activities of IACS-010759 in patients were also evaluated as secondary endpoints in both clinical trials. IACS-010759 had a narrow therapeutic index with emergent dose-limiting toxicities, including elevated blood lactate and neurotoxicity, which obstructed efforts to maintain target exposure. Consequently no RP2D was established, only modest target inhibition and limited antitumor activity were observed at tolerated doses, and both trials were discontinued. Reverse translational studies in mice demonstrated that IACS-010759 induced behavioral and physiological changes indicative of peripheral neuropathy, which were minimized with the coadministration of a histone deacetylase 6 inhibitor. Additional studies are needed to elucidate the association between OXPHOS inhibition and neurotoxicity, and caution is warranted in the continued development of complex I inhibitors as antitumor agents.
Collapse
Affiliation(s)
- Timothy A Yap
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Naval Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mikhila Mahendra
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jixiang Zhang
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meghan E Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Maria Emilia Di Francesco
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sisi Gao
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason P Gay
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonal Gera
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Han
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias J Jabbour
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Jennifer R Molina
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalia Baran
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maro Ohanian
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carolina Salguero
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anand K Singh
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Quanyun A Xu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Zhang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christopher A Bristow
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meenakshi B Bhattacharjee
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA
| | - Timothy P Heffernan
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher P Vellano
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip Jones
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cobi J Heijnen
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Psychological Sciences, Rice University, Houston, TX, USA
| | - Annemieke Kavelaars
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph R Marszalek
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Marina Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
Faria AVS, Fonseca EMB, Fernandes-Oliveira PDS, de Lima TI, Clerici SP, Justo GZ, Silveira LR, Durán N, Ferreira-Halder CV. Violacein switches off low molecular weight tyrosine phosphatase and rewires mitochondria in colorectal cancer cells. Bioorg Chem 2022; 127:106000. [PMID: 35853296 DOI: 10.1016/j.bioorg.2022.106000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 11/26/2022]
Abstract
In the last decade, emerging evidence has shown that low molecular weight protein tyrosine phosphatase (LMWPTP) not only contributes to the progression of cancer but is associated with prostate low survival rate and colorectal cancer metastasis. We report that LMWPTP favors the glycolytic profile in some tumors. Therefore, the focus of the present study was to identify metabolic enzymes that correlate with LMWPTP expression in patient samples. Exploratory data analysis from RNA-seq, proteomics, and histology staining, confirmed the higher expression of LMWPTP in CRC. Our descriptive statistical analyses indicate a positive expression correlation between LMWPTP and energy metabolism enzymes such as acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN). In addition, we examine the potential of violacein to reprogram energetic metabolism and LMWPTP activity. Violacein treatment induced a shift of glycolytic to oxidative metabolism associated with alteration in mitochondrial efficiency, as indicated by higher oxygen consumption rate. Particularly, violacein treated cells displayed higher proton leak and ATP-linked oxygen consumption rate (OCR) as an indicator of the OXPHOS preference. Notably, violacein is able to bind and inhibit LMWPTP. Since the LMWPTP acts as a hub of signaling pathways that offer tumor cells invasive advantages, such as survival and the ability to migrate, our findings highlight an unexplored potential of violacein in circumventing the metabolic plasticity of tumor cells.
Collapse
Affiliation(s)
- Alessandra V S Faria
- Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Emanuella M B Fonseca
- Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Federal Institute of Education, Science and Technology of São Paulo (IFSP), São Roque, São Paulo, Brazil
| | | | - Tanes I de Lima
- Department of Structural and Functional Biology, University of Campinas, (UNICAMP), Campinas, SP, Brazil
| | - Stefano P Clerici
- Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Giselle Z Justo
- Department of Pharmaceutical Sciences and Department of Biochemistry, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Leonardo R Silveira
- Department of Structural and Functional Biology, University of Campinas, (UNICAMP), Campinas, SP, Brazil
| | - Nelson Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Nanomedicine Research Unit (Nanomed), Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, Brazil
| | - Carmen V Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
8
|
Jensen-Cody S, Coyne ES, Ding X, Sebin A, Vogel J, Goldstein J, Rosahl TW, Zhou HH, Jacobs H, Champy MF, About GB, Talukdar S, Zhou Y. Loss of low-molecular-weight protein tyrosine phosphatase shows limited improvement in glucose tolerance but causes mild cardiac hypertrophy in mice. Am J Physiol Endocrinol Metab 2022; 322:E517-E527. [PMID: 35403438 DOI: 10.1152/ajpendo.00161.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin resistance is a major public health burden that often results in other comorbidities including type 2 diabetes, nonalcoholic fatty liver disease (NAFLD), and cardiovascular disease. An insulin sensitizer has the potential to become a disease-modifying therapy. It remains an unmet medical need to identify therapeutics that target the insulin signaling pathway to treat insulin resistance. Low-molecular-weight protein tyrosine phosphatase (LMPTP) negatively regulates insulin signaling and has emerged as a potential therapeutic target for insulin sensitization. Genetic studies have demonstrated that LMPTP is positively associated with obesity in humans and promotes insulin resistance in rodents. A recent study showed that pharmacological inhibition or genetic deletion of LMPTP protects mice from high-fat diet-induced insulin resistance and diabetes. Here, we show that loss of LMPTP by genetic deletion has no significant effects on improving glucose tolerance in lean or diet-induced obese mice. Furthermore, our data demonstrate that LMPTP deficiency potentiates cardiac hypertrophy that leads to mild cardiac dysfunction. Our findings suggest that the development of LMPTP inhibitors for the treatment of insulin resistance and type 2 diabetes should be reevaluated, and further studies are needed to characterize the molecular and pathophysiological role of LMPTP.NEW & NOTEWORTHY Inhibition of LMPTP with a small-molecule inhibitor, Cmpd23, improves glucose tolerance in mice as reported earlier. However, genetic deficiency of the LMPTP-encoding gene, Acp1, has limited effects on glucose metabolism but leads to mild cardiac hypertrophy in mice. The findings suggest the potential off-target effects of Cmpd23 and call for reevaluation of LMPTP as a therapeutic target for the treatment of insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
| | - Erin S Coyne
- Merck & Co., Inc., South San Francisco, California
| | - Xunshan Ding
- Merck & Co., Inc., South San Francisco, California
| | - Anu Sebin
- Merck & Co., Inc., South San Francisco, California
| | - Jen Vogel
- Merck & Co., Inc., South San Francisco, California
| | | | | | | | - Hugues Jacobs
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris, Université de Strasbourg, CNRS, INSERM, Illkirch, France
| | - Marie-France Champy
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris, Université de Strasbourg, CNRS, INSERM, Illkirch, France
| | - Ghina Bou About
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris, Université de Strasbourg, CNRS, INSERM, Illkirch, France
| | | | | |
Collapse
|