1
|
Chatzilygeroudi T, Karantanos T, Pappa V. Unraveling Venetoclax Resistance: Navigating the Future of HMA/Venetoclax-Refractory AML in the Molecular Era. Cancers (Basel) 2025; 17:1586. [PMID: 40361510 PMCID: PMC12071220 DOI: 10.3390/cancers17091586] [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: 03/28/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Acute myeloid leukemia (AML) has traditionally been linked to a poor prognosis, particularly in older patients who are ineligible for intensive chemotherapy. The advent of Venetoclax, a powerful oral BH3 mimetic targeting anti-apoptotic protein BCL2, has significantly advanced AML treatment. Its combination with the hypomethylating agent azacitidine (AZA/VEN) has become a standard treatment for this group of AML patients, demonstrating a 65% overall response rate and a median overall survival of 14.7 months, compared to 22% and 8 months with azacitidine monotherapy, respectively. However, resistance and relapses remain common, representing a significant clinical challenge. Recent studies have identified molecular alterations, such as mutations in FLT3-ITD, NRAS/KRAS, TP53, and BAX, as major drivers of resistance. Additionally, other factors, including metabolic changes, anti-apoptotic protein expression, and monocytic or erythroid/megakaryocytic differentiation status, contribute to treatment failure. Clinical trials are exploring strategies to overcome venetoclax resistance, including doublet or triplet therapies targeting IDH and FLT3 mutations; novel epigenetic approaches; menin, XPO1, and MDM2 inhibitors; along with immunotherapies like monoclonal antibodies and antibody-drug conjugates. A deeper understanding of the molecular mechanisms of resistance through single-cell analysis will be crucial for developing future therapeutic strategies.
Collapse
Affiliation(s)
- Theodora Chatzilygeroudi
- Second Department of Internal Medicine and Research Unit, Hematology Unit, National and Kapodistrian University of Athens School of Medicine, Attikon University Hospital, 12462 Athens, Greece;
| | - Theodoros Karantanos
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Medical Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Unit, Hematology Unit, National and Kapodistrian University of Athens School of Medicine, Attikon University Hospital, 12462 Athens, Greece;
| |
Collapse
|
2
|
Luo Y, Fu Y, Kuang M, Wang J, Zhao R, Luo S, Wang L, Chen J, Xu S, Zhou C. Ciclosporin A potentiates venetoclax efficacy in FLT3-ITD AML by targeting the NFATC1-AKT-mTOR-BCL-2/MCL-1 signaling axis. Br J Haematol 2025. [PMID: 40328636 DOI: 10.1111/bjh.20137] [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: 01/05/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025]
Abstract
The Fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutation in acute myeloid leukaemia (AML) is associated with adverse clinical outcomes, including poor prognosis, high relapse rates and reduced responses to conventional treatment regimens. While venetoclax (VEN) monotherapy has shown limited efficacy in FLT3-ITD AML due to intrinsic resistance mechanisms, this study demonstrates that ciclosporin A (CsA) synergistically enhances VEN's anti-leukaemic activity. CsA significantly suppresses cell proliferation, induces mitochondrial apoptosis and impairs mitochondrial bioenergetics in FLT3-ITD AML cells. Mechanistically, CsA enhances the effects of VEN through the downregulation of NFATC1, a critical regulator of the PI3K/AKT/mTOR signalling pathway. This suppression of NFATC1 leads to the coordinated downregulation of the anti-apoptotic proteins BCL-2 and MCL-1, thereby overcoming resistance and reinstating therapeutic susceptibility in FLT3-ITD AML.
Collapse
Affiliation(s)
- Yu Luo
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yinghao Fu
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Mei Kuang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jianming Wang
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Runlong Zhao
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Siqi Luo
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuangnian Xu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chengfang Zhou
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
3
|
Wang Z, Lai R, Wang X, Chen X, Zhou Y, Li S, Qiu X, Zeng Z, Yuan J, Mao J, Chen Z, Wang J. Targeted Penetrating Motif Engineering of BH3 Mimetic: Harnessing Non-Canonical Amino Acids for Coinhibition of MCL-1 and BCL-xL in Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503682. [PMID: 40305693 DOI: 10.1002/advs.202503682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/15/2025] [Indexed: 05/02/2025]
Abstract
Acute Myeloid Leukemia (AML) remains a formidable clinical challenge, predominantly due to the emergence of resistance to existing therapeutic regimens, including BCL-2 inhibitors like Venetoclax. Here, a novel approach is introduced by engineering BH3 mimetics utilizing non-canonical amino acids (ncAAs) to achieve dual inhibition of MCL-1 and BCL-xL. Through site saturation mutagenesis scanning, the I58(Chg) mutation is identified, significantly enhancing binding affinity with IC50 values of 2.77 nm for MCL-1 and 10.69 nm for BCL-xL, reflecting an increase of fourfold or more. The developed vMIP-II-TAT-I peptide, incorporating a CXCR4-targeted penetrating motif, demonstrated superior cellular uptake, with mean fluorescence intensity (MFI) 7.2-fold higher in CXCR4-positive AML cells and exhibited a high selectivity index (SI) for AML cells, with minimal impact on normal human hematopoietic stem cells (HSCs). When combined with Venetoclax, this peptide induced synergistic apoptosis, reducing tumor burden and prolonging survival in an AML mouse model, with median survival extended to 53 days from 37 days with Venetoclax alone. These findings reveal the therapeutic potential of dual inhibition in overcoming Venetoclax resistance and selectively targeting leukemic cells with reduced off-target effects, while laying the foundation for developing advanced BH3 mimetics with enhanced targeting, binding affinity, and efficacy for AML treatment.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Ruizhi Lai
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Youjian Zhou
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shengbin Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaohui Qiu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zekai Zeng
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Jianye Yuan
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Jinghuan Mao
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhidong Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
4
|
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
|
5
|
Li F, Chen Y, Zhuang H, Pei R, Lu Y, Chen D, Li S, Ye P, Lian J, Lu Y. A combination of Dihydroartemisinin and Venetoclax enhances antitumor effect in AML via C-MYC/BCL-XL/MCL-1 triple targeting. Discov Oncol 2025; 16:496. [PMID: 40202582 PMCID: PMC11982003 DOI: 10.1007/s12672-025-02242-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is associated with high rates of resistance to standard therapies, necessitating the exploration of novel treatment strategies. Venetoclax (VEN) has shown efficacy in AML, yet drug resistance remains a significant challenge. This study aims to explore the synergistic effects of combining dihydroartemisinin (DHA) with VEN to improve therapeutic outcomes in AML. METHODS AML cell lines and primary cells from AML patients were treated with various concentrations of DHA, VEN and their combined regimen. The cytotoxic effects were evaluated using MTS assays, flow cytometry for apoptosis analysis, and cell cycle assessments. Protein levels of caspase-3, PARP, MCL-1, BCL-XL and C-MYC were analyzed to elucidate the underlying mechanisms of the observed synergy. RESULTS The combination of VEN and DHA demonstrated a significant synergistic cytotoxic effect on AML cells, characterized by reduced cell proliferation, induced apoptosis, and cell cycle arrest in the G0/G1 phase. Mechanistically, the synergy was associated with increased levels of cleaved caspase-3 and PARP, along with the downregulation of anti-apoptotic proteins MCL-1 and BCL-XL. Additionally, the combined treatment led to a significant decrease in C-MYC expression. This synergistic effect was consistently observed across all primary AML patient samples analyzed. CONCLUSION The findings suggest that the combination of VEN and DHA exerts synergistic anti-leukemic effects by targeting BCL-XL, MCL-1 and C-MYC, offering a promising therapeutic strategy for AML.
Collapse
Affiliation(s)
- Fenglin Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Yao Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Shaoxing Central Hospital, Shaoxing, China
| | - Haihui Zhuang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Yuyu Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Dong Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Shuangyue Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Peipei Ye
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Jiaying Lian
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China.
- Institute of Hematology, Ningbo University, Ningbo, China.
| |
Collapse
|
6
|
Goulart H, Kantarjian H, Pemmaraju N, Daver N, DiNardo CD, Rausch CR, Ravandi F, Kadia TM. Venetoclax-Based Combination Regimens in Acute Myeloid Leukemia. Blood Cancer Discov 2025; 6:23-37. [PMID: 39565177 PMCID: PMC11707511 DOI: 10.1158/2643-3230.bcd-24-0171] [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: 07/05/2024] [Revised: 08/22/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
SIGNIFICANCE In recent years, there has been tremendous interest surrounding the integration of venetoclax into both non-intensive and intensive chemotherapy regimens for AML. However, with this increasing utilization of venetoclax, considerable questions surrounding key issues such as dosing strategies and the practicality of venetoclax administration have arisen. This review highlights the evolution of venetoclax-based regimens in AML and provides a commentary on notable practical considerations when utilizing this agent.
Collapse
Affiliation(s)
- Hannah Goulart
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naveen Pemmaraju
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney D. DiNardo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caitlin R. Rausch
- Division of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan M. Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
7
|
Safa-Tahar-Henni S, Páez Martinez K, Gress V, Esparza N, Roques É, Bonnet-Magnaval F, Bilodeau M, Gagné V, Bresson E, Cardin S, El-Hachem N, Iasenza I, Alzial G, Boivin I, Nakamichi N, Soufflet AC, Mirela Pascariu C, Duchaine J, Mathien S, Bonneil É, Eppert K, Marinier A, Sauvageau G, Deblois G, Thibault P, Hébert J, Eaves CJ, Cellot S, Barabé F, Wilhelm BT. Comparative small molecule screening of primary human acute leukemias, engineered human leukemia and leukemia cell lines. Leukemia 2025; 39:29-41. [PMID: 39472547 PMCID: PMC11717705 DOI: 10.1038/s41375-024-02400-w] [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: 12/12/2023] [Revised: 07/14/2024] [Accepted: 08/28/2024] [Indexed: 01/11/2025]
Abstract
Targeted therapeutics for high-risk cancers remain an unmet medical need. Here we report the results of a large-scale screen of over 11,000 molecules for their ability to inhibit the survival and growth in vitro of human leukemic cells from multiple sources including patient samples, de novo generated human leukemia models, and established human leukemic cell lines. The responses of cells from de novo models were most similar to those of patient samples, both of which showed striking differences from the cell-line responses. Analysis of differences in subtype-specific therapeutic vulnerabilities made possible by the scale of this screen enabled the identification of new specific modulators of apoptosis, while also highlighting the complex polypharmacology of anti-leukemic small molecules such as shikonin. These findings introduce a new platform for uncovering new therapeutic options for high-risk human leukemia, in addition to reinforcing the importance of the test sample choice for effective drug discovery.
Collapse
Affiliation(s)
- Safia Safa-Tahar-Henni
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Karla Páez Martinez
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Verena Gress
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Nayeli Esparza
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Élodie Roques
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Florence Bonnet-Magnaval
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Mélanie Bilodeau
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Valérie Gagné
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Eva Bresson
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Sophie Cardin
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Nehme El-Hachem
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Isabella Iasenza
- Centre for Translational Biology, McGill University Heath Centre Research Institute, Montréal, QC, Canada
| | - Gabriel Alzial
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Metabolic and Epigenetic Alterations in Cancer Research unit, Montréal, QC, Canada
| | - Isabel Boivin
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Molecular Genetics of Stem Cells Research Unit, Montréal, QC, Canada
| | - Naoto Nakamichi
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Anne-Cécile Soufflet
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Cristina Mirela Pascariu
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Proteomics and Bioanalytical Mass Spectrometry Research Unit, Montréal, QC, Canada
| | - Jean Duchaine
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- High throughput screening platform, Montréal, QC, Canada
| | - Simon Mathien
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- High throughput screening platform, Montréal, QC, Canada
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Proteomics and Bioanalytical Mass Spectrometry Research Unit, Montréal, QC, Canada
| | - Kolja Eppert
- Centre for Translational Biology, McGill University Heath Centre Research Institute, Montréal, QC, Canada
- Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Anne Marinier
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Medicinal Chemistry/Drug Discovery Unit, Montréal, QC, Canada
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Molecular Genetics of Stem Cells Research Unit, Montréal, QC, Canada
- Institut universitaire d'hémato-oncologie et de thérapie cellulaire, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Geneviève Deblois
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Faculty of Medicine, Faculty of Pharmacy, University of Montréal, Montréal, QC, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Proteomics and Bioanalytical Mass Spectrometry Research Unit, Montréal, QC, Canada
- Department of Chemistry, Faculty of Arts and Sciences, Université de Montréal, Montréal, QC, Canada
| | - Josée Hébert
- Institut universitaire d'hémato-oncologie et de thérapie cellulaire, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Sonia Cellot
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| | - Frédéric Barabé
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada.
- Department of Medicine, Université Laval, Quebec City, QC, Canada.
| | - Brian T Wilhelm
- Laboratory for High Throughput Biology, Montréal, QC, Canada.
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
8
|
Tatarata QZ, Wang Z, Konopleva M. BCL-2 inhibition in acute myeloid leukemia: resistance and combinations. Expert Rev Hematol 2024; 17:935-946. [PMID: 39552410 DOI: 10.1080/17474086.2024.2429604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The introduction of venetoclax has revolutionized the treatment landscape of acute myeloid leukemia, offering new therapeutic opportunities. However, the clinical response to venetoclax varies significantly between patients, with many experiencing limited duration of response. AREAS COVERED Identified resistance mechanisms include both intrinsic and acquired resistance to VEN. The former is associated with cell lineage and differentiation state. The latter includes dependency on alternative BCL-2 family anti-apoptotic protein(s) mediated by genetic, epigenetic, or post-translational mechanisms, mitochondrial and metabolic involvement, as well as microenvironment. Understanding these mechanisms is crucial for optimizing venetoclax-based therapies and enhancing treatment outcomes for patients with acute myeloid leukemia. This review aims to elucidate the primary mechanisms underlying resistance to venetoclax and explore current therapeutic strategies to overcome this challenge. EXPERT OPINION In patients with venetoclax resistance, alternative options include targeted combination therapies tailored to individual cases based on cytogenetics and prior treatments. Many of these therapies require further clinical investigation to validate their safety and efficacy.
Collapse
Affiliation(s)
- Qi Zhang Tatarata
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- The Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhe Wang
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
9
|
Wu D, Li M, Hong Y, Jin L, Liu Q, Sun C, Li L, Han X, Deng S, Feng Y, Shen Y, Kai G. Integrated stress response activation induced by usnic acid alleviates BCL-2 inhibitor ABT-199 resistance in acute myeloid leukemia. J Adv Res 2024:S2090-1232(24)00436-3. [PMID: 39384125 DOI: 10.1016/j.jare.2024.10.003] [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: 06/16/2024] [Revised: 10/06/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024] Open
Abstract
INTRODUCTION ABT-199 (venetoclax) is a BCL-2 suppressor with pronounced effects on acute myeloid leukemia (AML). However, its usefulness as a monotherapy or in combination with hypomethylating medicines like azacitidine is debatable due to acquired resistance. Usnic acid, a dibenzofuran extracted from lichen Usnea diffracta Vain, exhibits anticancer properties and may counteract multidrug resistance in leukemia cells. OBJECTIVE This study investigated whether usnic acid at low-cytotoxicity level could enhance sensitivity of AML cells with acquired resistance to ABT-199 by targeting the integrated stress response pathways. METHODS To investigate the combined effects on AML cells, we used a cell viability test, flow cytometry to quantify apoptosis, cell cycle analysis, and mitochondrial membrane potential measurement. RNA-seq and immunoblot were used to determine the potential mechanisms of ABT-199 + usnic acid combination. RESULTS Usnic acid, at a low cytotoxicity level, successfully restored ABT-199 sensitivity in AML cell lines that had developed ABT-199 resistance and increased ABT-199's antileukemic activity in a xenograft model. Mechanistically, the combination of usnic acid and ABT-199 cooperated to boost the expression of the integrated stress response (ISR)-associated genes ATF4, CHOP, and NOXA through the heme-regulated inhibitor kinase (HRI), while also promoting the degradation of the anti-apoptotic protein MCL-1. ISRIB, a compound that blocks the ISR, was able to reverse the growth suppression and cell death, the increase in expression of genes related with the ISR, and the inhibition of MCL-1 protein caused by combination therapy. Additionally, the downregulation of MCL-1 was linked to an increase in MCL-1 phosphorylation at serine 159 and subsequent destruction by the proteasome. CONCLUSION In summary, usnic acid improves chemosensitivity to ABT-199 by triggering the integrated stress response, leading to decreased levels of MCL-1 protein, suggesting a potential treatment for AML cases resistant to Bcl-2 inhibitors.
Collapse
Affiliation(s)
- Dijiong Wu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China; Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Man Li
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China; Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yaonan Hong
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Li Jin
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qi Liu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Chengtao Sun
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liqin Li
- Key Laboratory of Traditional Chinese Medicine for the Development and Clinical Transformation of Immunomodulatory Traditional Chinese Medicine in Zhejiang Province, Huzhou Central Hospital, the Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| | - Xiaoxiao Han
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengqian Deng
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yue Feng
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yiping Shen
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Guoyin Kai
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China; Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Shahswar R, Ganser A. Relapse and resistance in acute myeloid leukemia post venetoclax: improving second lines therapy and combinations. Expert Rev Hematol 2024; 17:723-739. [PMID: 39246164 DOI: 10.1080/17474086.2024.2402283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION The combined use of the BCL-2 inhibitor venetoclax with azacitidine now is the standard of care for patients with acute myeloid leukemia (AML) unfit for intensive chemotherapy with outcomes exceeding those achieved with hypomethylating agents alone. Venetoclax in combination with intensive chemotherapy is also increasingly used both as frontline as well as salvage therapy. However, resistance to and relapse after venetoclax-based therapies are of major concern and outcomes after treatment failure remain poor. AREAS COVERED A comprehensive search was performed using PubMed database (up to April 2024). Studies evaluating venetoclax-based combination treatments in AML and studies assessing markers of response and resistance to venetoclax were investigated. We summarize the status of venetoclax-based therapies in the frontline and relapsed/refractory setting with focus on the main mechanisms of resistance to BCL-2 inhibition. Further, strategies to overcome resistance including combinatorial regimens of hypomethylating agent (HMA) + venetoclax + inhibitors targeting actionable mutations like IDH1/2 or FLT3-ITD and the introduction of novel agents like menin-inhibitors are addressed. EXPERT OPINION Although venetoclax is reshaping the treatment of unfit and fit AML patients, prognosis of patients after HMA/VEN failure remains dismal, and strategies to abrogate primary and secondary resistance are an unmet clinical need.
Collapse
Affiliation(s)
- Rabia Shahswar
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Lu Y, Jiang X, Li Y, Li F, Zhao M, Lin Y, Jin L, Zhuang H, Li S, Ye P, Pei R, Jin J, Jiang L. NL101 synergizes with the BCL-2 inhibitor venetoclax through PI3K-dependent suppression of c-Myc in acute myeloid leukaemia. J Transl Med 2024; 22:867. [PMID: 39334157 PMCID: PMC11429391 DOI: 10.1186/s12967-024-05647-0] [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: 07/18/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Acute myeloid leukaemia (AML) comprises a group of heterogeneous and aggressive haematological malignancies with unsatisfactory prognoses and limited treatment options. Treatments targeting B-cell lymphoma-2 (BCL-2) with venetoclax have been approved for patients with AML, and venetoclax-based drug combinations are becoming the standard of care for older patients unfit for intensive chemotherapy. However, the therapeutic duration of either single or combination strategies is limited, and the development of resistance seems inevitable. Therefore, more effective combination regimens are urgently needed. METHODS The efficacy of combination therapy with NL101, a SAHA-bendamustine hybrid, and venetoclax was evaluated in preclinical models of AML including established cell lines, primary blasts from patients, and animal models. RNA-sequencing and immunoblotting were used to explore the underlying mechanism. RESULTS NL101 significantly potentiated the activity of venetoclax in AML cell lines, as evidenced by the enhanced decrease in viability and induction of apoptosis. Mechanistically, the addition of NL101 to venetoclax decreased the stability of the antiapoptotic protein myeloid cell leukaemia-1 (MCL-1) by inhibiting ERK, thereby facilitating the release of BIM and triggering mitochondrial apoptosis. Moreover, the strong synergy between NL101 and venetoclax also relied on the downregulation of c-Myc via PI3K/Akt/GSK3β signalling. The combination of NL101 and venetoclax synergistically eliminated primary blasts from 10 AML patients and reduced the leukaemia burden in an MV4-11 cell-derived xenograft model. CONCLUSIONS Our results encourage the pursuit of clinical trials of combined treatment with NL101 and venetoclax and provide a novel venetoclax-incorporating therapeutic strategy for AML.
Collapse
Affiliation(s)
- Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Xia Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Department of Pathology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Youhong Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Department of Pathology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Fenglin Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Mengting Zhao
- Department of Pathology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Ye Lin
- Department of Pathology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lili Jin
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Department of Pathology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Haihui Zhuang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Shuangyue Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Peipei Ye
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lei Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China.
- Department of Pathology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
12
|
Marvin-Peek J, Gilbert JS, Pollyea DA, DiNardo CD. Frontline therapy of acute myeloid leukemia with lower intensity regimens: Where are we now and where can we go? Am J Hematol 2024; 99:1790-1801. [PMID: 39016070 DOI: 10.1002/ajh.27434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/22/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
The advent of molecularly targeted therapeutics has transformed the management of patients with acute myeloid leukemia (AML). Particularly for individuals unfit for intensive chemotherapy, lower intensity therapies (LIT) incorporating small molecules have significantly improved patient outcomes. With BCL2, IDH1, IDH2, and FLT3 inhibitors widely used for relapsed AML, combination regimens are now utilized in the frontline. Expansion of these targeted LIT combinations, along with development of novel agents including menin inhibitors, exemplifies the promise of precision medicine. Further understanding of molecular drivers of leukemic transformation and mechanisms of relapse will continue to advance frontline treatment options for patients with AML.
Collapse
Affiliation(s)
- Jennifer Marvin-Peek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason S Gilbert
- Department of Internal Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
13
|
Peng X, Tang F, Li Y, Bai J, Li L, Zhang L. Combination of BCL-2 inhibitors and immunotherapy: a promising therapeutic strategy for hematological malignancies. Discov Oncol 2024; 15:311. [PMID: 39060763 PMCID: PMC11282050 DOI: 10.1007/s12672-024-01161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The rapid development of high-throughput sequencing in recent years has facilitated great progress in the molecular-targeted therapy of hematological malignancies, including leukemia, lymphoma, and multiple myeloma. BCL-2 inhibitors are among the most important molecular-targeted agents. Immunotherapy for hematologic malignancy has rapidly increased in popularity in recent years and has been proven to improve the overall survival rate. However, few clinical studies have investigated combination therapy with BCL-2 inhibitors and immunotherapies, such as immune molecule-targeted drugs or immune cell adoptive therapy. In this review, we discuss the drug discovery process, current clinical application status, and resistance and tolerance issues associated with BCL-2 inhibitors. We emphasize their important role in regulating the immune system and propose that the combination of BCL-2 inhibitors with immunotherapy may be one of the most promising treatment methods for hematologic malignancies.
Collapse
Affiliation(s)
- Xiaohuan Peng
- Department of Hematology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Futian Tang
- Key Laboratory of the Digestive Tumor of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yanhong Li
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jun Bai
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Liansheng Zhang
- Department of Hematology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
14
|
Enzenmüller S, Niedermayer A, Seyfried F, Muench V, Tews D, Rupp U, Tausch E, Groß A, Fischer-Posovszky P, Walther P, Stilgenbauer S, Kestler HA, Debatin KM, Meyer LH. Venetoclax resistance in acute lymphoblastic leukemia is characterized by increased mitochondrial activity and can be overcome by co-targeting oxidative phosphorylation. Cell Death Dis 2024; 15:475. [PMID: 38961053 PMCID: PMC11222427 DOI: 10.1038/s41419-024-06864-7] [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: 11/17/2023] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Deregulated apoptosis signaling is characteristic for many cancers and contributes to leukemogenesis and treatment failure in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Apoptosis is controlled by different pro- and anti-apoptotic molecules. Inhibition of anti-apoptotic molecules like B-cell lymphoma 2 (BCL-2) has been developed as therapeutic strategy. Venetoclax (VEN), a selective BCL-2 inhibitor has shown clinical activity in different lymphoid malignancies and is currently evaluated in first clinical trials in BCP-ALL. However, insensitivity to VEN has been described constituting a major clinical concern. Here, we addressed and modeled VEN-resistance in BCP-ALL, investigated the underlying mechanisms in cell lines and patient-derived xenograft (PDX) samples and identified potential strategies to overcome VEN-insensitivity. Leukemia lines with VEN-specific resistance were generated in vitro and further characterized using RNA-seq analysis. Interestingly, gene sets annotated to the citric/tricarboxylic acid cycle and the respiratory electron transport chain were significantly enriched and upregulated, indicating increased mitochondrial metabolism in VEN-resistant ALL. Metabolic profiling showed sustained high mitochondrial metabolism in VEN-resistant lines as compared to control lines. Accordingly, primary PDX-ALL samples with intrinsic VEN-insensitivity showed higher oxygen consumption and ATP production rates, further highlighting that increased mitochondrial activity is a characteristic feature of VEN-resistant ALL. VEN-resistant PDX-ALL showed significant higher mitochondrial DNA content and differed in mitochondria morphology with significantly larger and elongated structures, further corroborating our finding of augmented mitochondrial metabolism upon VEN-resistance. Using Oligomycin, an inhibitor of the complex V/ATPase subunit, we found synergistic activity and apoptosis induction in VEN-resistant BCP-ALL cell lines and PDX samples, demonstrating that acquired and intrinsic VEN-insensitivity can be overcome by co-targeting BCL-2 and the OxPhos pathway. These findings of reprogrammed, high mitochondrial metabolism in VEN-resistance and synergistic activity upon co-targeting BCL-2 and oxidative phosphorylation strongly suggest further preclinical and potential clinical evaluation in VEN-resistant BCP-ALL.
Collapse
Affiliation(s)
- Stefanie Enzenmüller
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Alexandra Niedermayer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
- International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Felix Seyfried
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Vera Muench
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Daniel Tews
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ulrich Rupp
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Eugen Tausch
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Alexander Groß
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | | | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Stephan Stilgenbauer
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Lüder Hinrich Meyer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
15
|
Derippe T, Fouliard S, Decleves X, Mager DE. Quantitative systems pharmacology modeling of tumor heterogeneity in response to BH3-mimetics using virtual tumors calibrated with cell viability assays. CPT Pharmacometrics Syst Pharmacol 2024; 13:1252-1263. [PMID: 38747730 PMCID: PMC11247121 DOI: 10.1002/psp4.13158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/17/2024] [Accepted: 04/24/2024] [Indexed: 07/16/2024] Open
Abstract
Both primary and acquired resistance mechanisms that involve intra-tumoral cell heterogeneity limit the use of BH3-mimetics to trigger tumor cell apoptosis. This article proposes a new quantitative systems pharmacology (QSP)-based methodology in which cell viability assays are used to calibrate virtual tumors (VTs) made of virtual cells whose fate is determined by simulations from an apoptosis QSP model. VTs representing SU-DHL-4 and KARPAS-422 cell lines were calibrated using in vitro data involving venetoclax (anti-BCL2), A-1155463 (anti-BCLXL), and/or A-1210477 (anti-MCL1). The calibrated VTs provide insights into the combination of several BH3-mimetics, such as the distinction between cells eliminated by at least one of the drugs (monotherapies) from the cells eliminated by a pharmacological combination only. Calibrated VTs can also be used as initial conditions in an agent-based model (ABM) framework, and a minimal ABM was developed to bridge in vitro SU-DHL-4 cell viability results to tumor growth inhibition experiments in mice.
Collapse
Affiliation(s)
- Thibaud Derippe
- Institut de Recherches Internationales Servier, Suresnes, France
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, New York, USA
| | - Sylvain Fouliard
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Xavier Decleves
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, New York, USA
- Enhanced Pharmacodynamics, LLC, Buffalo, New York, USA
| |
Collapse
|
16
|
Kelly LM, Rutter JC, Lin KH, Ling F, Duchmann M, Latour E, Arang N, Pasquer H, Ho Nhat D, Charles J, Killarney ST, Ang HX, Namor F, Culeux C, Lombard B, Loew D, Swaney DL, Krogan NJ, Brunel L, Carretero É, Verdié P, Amblard M, Fodil S, Huynh T, Sebert M, Adès L, Raffoux E, Fenouille N, Itzykson R, Lobry C, Benajiba L, Forget A, Martin AR, Wood KC, Puissant A. Targeting a lineage-specific PI3Kɣ-Akt signaling module in acute myeloid leukemia using a heterobifunctional degrader molecule. NATURE CANCER 2024; 5:1082-1101. [PMID: 38816660 PMCID: PMC11778622 DOI: 10.1038/s43018-024-00782-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
Dose-limiting toxicity poses a major limitation to the clinical utility of targeted cancer therapies, often arising from target engagement in nonmalignant tissues. This obstacle can be minimized by targeting cancer dependencies driven by proteins with tissue-restricted and/or tumor-restricted expression. In line with another recent report, we show here that, in acute myeloid leukemia (AML), suppression of the myeloid-restricted PIK3CG/p110γ-PIK3R5/p101 axis inhibits protein kinase B/Akt signaling and compromises AML cell fitness. Furthermore, silencing the genes encoding PIK3CG/p110γ or PIK3R5/p101 sensitizes AML cells to established AML therapies. Importantly, we find that existing small-molecule inhibitors against PIK3CG are insufficient to achieve a sustained long-term antileukemic effect. To address this concern, we developed a proteolysis-targeting chimera (PROTAC) heterobifunctional molecule that specifically degrades PIK3CG and potently suppresses AML progression alone and in combination with venetoclax in human AML cell lines, primary samples from patients with AML and syngeneic mouse models.
Collapse
Affiliation(s)
- Lois M Kelly
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Justine C Rutter
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Kevin H Lin
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Frank Ling
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Matthieu Duchmann
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Emmanuelle Latour
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Nadia Arang
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Hélène Pasquer
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Duong Ho Nhat
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Juliette Charles
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Shane T Killarney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Hazel X Ang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Federica Namor
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Cécile Culeux
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Bérangère Lombard
- Curie Institute, Mass Spectrometry and Proteomics Facility, PSL Research University, Paris, France
| | - Damarys Loew
- Curie Institute, Mass Spectrometry and Proteomics Facility, PSL Research University, Paris, France
| | - Danielle L Swaney
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Gladstone Institutes, San Francisco, California, USA
| | - Nevan J Krogan
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Gladstone Institutes, San Francisco, California, USA
| | - Luc Brunel
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Élodie Carretero
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Pascal Verdié
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Muriel Amblard
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Sofiane Fodil
- Department of Hematology and Immunology, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Tony Huynh
- Department of Hematology and Immunology, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Marie Sebert
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
- Department of Hematology and Immunology, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Lionel Adès
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
- Department of Hematology and Immunology, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Emmanuel Raffoux
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
- Department of Hematology and Immunology, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Nina Fenouille
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Raphaël Itzykson
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
- Department of Hematology and Immunology, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Camille Lobry
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
| | - Lina Benajiba
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France
- Clinical Investigation Center, Saint-Louis Hospital, AP-HP, Paris Cité University, Paris, France
| | - Antoine Forget
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Anthony R Martin
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| | - Alexandre Puissant
- INSERM UMR 944, IRSL, Saint-Louis Hospital, Paris Cité University, Paris, France.
| |
Collapse
|
17
|
Nwosu GO, Ross DM, Powell JA, Pitson SM. Venetoclax therapy and emerging resistance mechanisms in acute myeloid leukaemia. Cell Death Dis 2024; 15:413. [PMID: 38866760 PMCID: PMC11169396 DOI: 10.1038/s41419-024-06810-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Acute myeloid leukaemia (AML) is a highly aggressive and devastating malignancy of the bone marrow and blood. For decades, intensive chemotherapy has been the frontline treatment for AML but has yielded only poor patient outcomes as exemplified by a 5-year survival rate of < 30%, even in younger adults. As knowledge of the molecular underpinnings of AML has advanced, so too has the development new strategies with potential to improve the treatment of AML patients. To date the most promising of these targeted agents is the BH3-mimetic venetoclax which in combination with standard of care therapies, has manageable non-haematological toxicity and exhibits impressive efficacy. However, approximately 30% of AML patients fail to respond to venetoclax-based regimens and almost all treatment responders eventually relapse. Here, we review the emerging mechanisms of intrinsic and acquired venetoclax resistance in AML and highlight recent efforts to identify novel strategies to overcome resistance to venetoclax.
Collapse
Affiliation(s)
- Gus O Nwosu
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - David M Ross
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Department of Haematology, Flinders University and Medical Centre, Adelaide, SA, Australia
| | - Jason A Powell
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
- Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
- Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia.
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
18
|
Sun Y, Zhu G, Zhong H. Minimal residual disease monitoring in acute myeloid leukemia: Focus on MFC-MRD and treatment guidance for elderly patients. Eur J Haematol 2024; 112:870-878. [PMID: 38342613 DOI: 10.1111/ejh.14187] [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: 11/24/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/13/2024]
Abstract
Acute myeloid leukemia (AML) is distinguished by clonal growth of myeloid precursor cells, which impairs normal hematopoiesis. Minimal residual disease (MRD) refers to the residual leukemia cells that persist after chemotherapy. Patients who test positive for MRD have a higher likelihood of experiencing a recurrence, regardless of the specific chemotherapy approach used. Multi-parameter flow cytometry (MFC), polymerase chain reaction (PCR), and next-generation sequencing (NGS) are commonly employed techniques for identifying MRD. In the context of AML, patients are frequently monitored for measurable residual disease via multi-parameter flow cytometry (MFC-MRD). In order to explore recent advancements in AML and MRD diagnosis, an extensive search of the PubMed database was conducted, focusing on relevant research in the past 20 years. This review aims to examine various MRD monitoring methods, the optimal time points for assessment, as well as different specimen types used. Additionally, it underscores the significance of MFC-MRD assessment in guiding the treatment of elderly AML.
Collapse
Affiliation(s)
- Yue Sun
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Gelan Zhu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
Fowler-Shorten DJ, Hellmich C, Markham M, Bowles KM, Rushworth SA. BCL-2 inhibition in haematological malignancies: Clinical application and complications. Blood Rev 2024; 65:101195. [PMID: 38523032 DOI: 10.1016/j.blre.2024.101195] [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: 01/10/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
B-cell lymphoma-2 (BCL-2) family proteins are fundamental regulators of the intrinsic apoptotic pathway which modulate cellular fate. In many haematological malignancies, overexpression of anti-apoptotic factors (BCL-2, BCL-XL and MCL-1) circumvent apoptosis. To address this cancer hallmark, a concerted effort has been made to induce apoptosis by inhibiting BCL-2 family proteins. A series of highly selective BCL-2 homology 3 (BH3) domain mimetics are in clinical use and in ongoing clinical trials for acute myeloid leukaemia (AML), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), and multiple myeloma (MM). These inhibitors serve as promising candidates, both as single agents or in combination therapy to improve patient outcomes. In other diseases such as follicular lymphoma, efficacy has been notably limited. There are also clinical problems with BCL-2 family inhibition, including drug resistance, disease relapse, tumour lysis syndrome, and clinically relevant cytopenias. Here, we provide a balanced view on both the clinical benefits of BCL-2 inhibition as well as the associated challenges.
Collapse
Affiliation(s)
- Dominic J Fowler-Shorten
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Charlotte Hellmich
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK; Department of Haematology, Norfolk and Norwich University Hospital NHS Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Matthew Markham
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Kristian M Bowles
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK; Department of Haematology, Norfolk and Norwich University Hospital NHS Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Stuart A Rushworth
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK.
| |
Collapse
|
20
|
Short NJ, Daver N, Dinardo CD, Kadia T, Nasr LF, Macaron W, Yilmaz M, Borthakur G, Montalban-Bravo G, Garcia-Manero G, Issa GC, Chien KS, Jabbour E, Nasnas C, Huang X, Qiao W, Matthews J, Stojanik CJ, Patel KP, Abramova R, Thankachan J, Konopleva M, Kantarjian H, Ravandi F. Azacitidine, Venetoclax, and Gilteritinib in Newly Diagnosed and Relapsed or Refractory FLT3-Mutated AML. J Clin Oncol 2024; 42:1499-1508. [PMID: 38277619 PMCID: PMC11095865 DOI: 10.1200/jco.23.01911] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/10/2023] [Accepted: 11/28/2023] [Indexed: 01/28/2024] Open
Abstract
PURPOSE Azacitidine plus venetoclax is a standard of care for patients with newly diagnosed AML who are unfit for intensive chemotherapy. However, FLT3 mutations are a common mechanism of resistance to this regimen. The addition of gilteritinib, an oral FLT3 inhibitor, to azacitidine and venetoclax may improve outcomes in patients with FLT3-mutated AML. METHODS This phase I/II study evaluated azacitidine, venetoclax, and gilteritinib in two cohorts: patients with (1) newly diagnosed FLT3-mutated AML who were unfit for intensive chemotherapy or (2) relapsed/refractory FLT3-mutated AML (ClinicalTrials.gov identifier: NCT04140487). The primary end points were the maximum tolerated dose of gilteritinib (phase I) and the combined complete remission (CR)/CR with incomplete hematologic recovery (CRi) rate (phase II). RESULTS Fifty-two patients were enrolled (frontline [n = 30]; relapsed/refractory [n = 22]). The recommended phase II dose was gilteritinib 80 mg once daily in combination with azacitidine and venetoclax. In the frontline cohort, the median age was 71 years and 73% of patients had an FLT3-internal tandem duplication (ITD) mutation. The CR/CRi rate was 96% (CR, 90%; CRi, 6%). Sixty-five percent of evaluable patients achieved FLT3-ITD measurable residual disease <5 × 10-5 within four cycles. With a median follow-up of 19.3 months, the median relapse-free survival (RFS) and overall survival (OS) have not been reached and the 18-month RFS and OS rates are 71% and 72%, respectively. In the relapsed/refractory cohort, the CR/CRi rate was 27%; nine additional patients (41%) achieved a morphologic leukemia-free state. The most common grade 3 or higher nonhematologic adverse events were infection (62%) and febrile neutropenia (38%), which were more frequent in the relapsed/refractory cohort. CONCLUSION The combination of azacitidine, venetoclax, and gilteritinib resulted in high rates of CR/CRi, deep FLT3 molecular responses, and encouraging survival in newly diagnosed FLT3-mutated AML. Myelosuppression was manageable with mitigative dosing strategies.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D. Dinardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lewis F. Nasr
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Walid Macaron
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Ghayas C. Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kelly S. Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cedric Nasnas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jairo Matthews
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Keyur P. Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Regina Abramova
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jennifer Thankachan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
21
|
Nachmias B, Aumann S, Haran A, Schimmer AD. Venetoclax resistance in acute myeloid leukaemia-Clinical and biological insights. Br J Haematol 2024; 204:1146-1158. [PMID: 38296617 DOI: 10.1111/bjh.19314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 04/11/2024]
Abstract
Venetoclax, an oral BCL-2 inhibitor, has been widely incorporated in the treatment of acute myeloid leukaemia. The combination of hypomethylating agents and venetoclax is the current standard of care for elderly and patient's ineligible for aggressive therapies. However, venetoclax is being increasingly used with aggressive chemotherapy regimens both in the front line and in the relapse setting. Our growing experience and intensive research demonstrate that certain genetic abnormalities are associated with venetoclax sensitivity, while others with resistance, and that resistance can emerge during treatment leading to disease relapse. In the current review, we provide a summary of the known mechanisms of venetoclax cytotoxicity, both regarding the inhibition of BCL-2-mediated apoptosis and its effect on cell metabolism. We describe how these pathways are linked to venetoclax resistance and are associated with specific mutations. Finally, we provide the rationale for novel drug combinations in current and future clinical trials.
Collapse
Affiliation(s)
- Boaz Nachmias
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shlomzion Aumann
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arnon Haran
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Nguyen TM, Joyce P, Ross DM, Bremmell K, Jambhrunkar M, Wong SS, Prestidge CA. Combating Acute Myeloid Leukemia via Sphingosine Kinase 1 Inhibitor-Nanomedicine Combination Therapy with Cytarabine or Venetoclax. Pharmaceutics 2024; 16:209. [PMID: 38399263 PMCID: PMC10893145 DOI: 10.3390/pharmaceutics16020209] [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: 12/25/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
MP-A08 is a novel sphingosine kinase 1 (SPHK1) inhibitor with activity against acute myeloid leukemia (AML). A rationally designed liposome-based encapsulation and delivery system has been shown to overcome the physicochemical challenges of MP-A08 and enable its effective delivery for improved efficacy and survival of mice engrafted with human AML in preclinical models. To establish therapies that overcome AML's heterogeneous nature, here we explored the combination of MP-A08-loaded liposomes with both the standard chemotherapy, cytarabine, and the targeted therapy, venetoclax, against human AML cell lines. Cytarabine (over the dose range of 0.1-0.5 µM) in combination with MP-A08 liposomes showed significant synergistic effects (as confirmed by the Chou-Talalay Combination Index) against the chemosensitised human AML cell lines MV4-11 and OCI-AML3. Venetoclax (over the dose range of 0.5-250 nM) in combination with MP-A08 liposomes showed significant synergistic effects against the chemosensitised human AML cell lines, particularly in venetoclax-resistant human AML cells. This strong synergistic effect is due to multiple mechanisms of action, i.e., inhibiting MCL-1 through SPHK1 inhibition, leading to ceramide accumulation, activation of protein kinase R, ATF4 upregulation, and NOXA activation, ultimately resulting in MCL-1 degradation. These combination therapies warrant further consideration and investigation in the search for a more comprehensive treatment strategy for AML.
Collapse
Affiliation(s)
- Thao M. Nguyen
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia;
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - David M. Ross
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia;
- Department of Haematology, Flinders University and Medical Centre, Adelaide, SA 5001, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA 5001, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Kristen Bremmell
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - Manasi Jambhrunkar
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - Sook S. Wong
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - Clive A. Prestidge
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| |
Collapse
|
23
|
Forsberg M, Konopleva M. SOHO State of the Art Updates and Next Questions: Understanding and Overcoming Venetoclax Resistance in Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:1-14. [PMID: 38007372 DOI: 10.1016/j.clml.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/27/2023]
Abstract
The discovery of Venetoclax (VEN) has transformed the therapeutic landscape of acute myeloid leukemia (AML) and chronic lymphocytic leukemia (CLL). However, the response is heterogeneous with 10% to 50% of newly diagnosed AML patients not responding to hypomethylating agent (HMA) and VEN. Furthermore, up to 40% of responding patients relapse shortly. This review discusses the mechanism of action of Venetoclax and the major mechanisms of inherent and acquired resistance to VEN. VEN is highly specific to BCL-2 binding, as such other antiapoptotic proteins in BCL-2 family induce resistance. These antiapoptotic proteins can also be upregulated via a number of compensatory cell signaling pathways including PI3K/AKT/mTOR, the MAPK/ERK pathway, and mutant FLT3-ITD. Mutations can occur in BCL-2 and BAX proteins, or they can be silenced by TP53 mutations and other epigenetic changes. Changes to mitochondrial structure and metabolism can induce resistance. Key metabolic regulators include OXPHOS and alternative amino acid metabolism. Finally microenvironmental factors can influence VEN responses. This paper evaluates subsets of AML by differentiation, histology, cytogenetics and molecular markers and their different responses to VEN; with spliceosome mutations, ASXL1, NPM1 and IDH1/2 being favorable while others such as FLT3, TP53 and BCL-2 mutations being less responsive. Currently intensive multiagent chemotherapy and Venetoclax combinations such as 7+3+VEN are favored in fit younger AML patients. However, with resistant patients' subsets targeted combination therapies are becoming an increasingly attractive option. We explore the incorporation of non-BCL-2 inhibitors, next-generation BCL-2 and multi-protein agents, other inhibitors most prominently FLT-3 inhibitors in addition to Venetoclax, and other novel approaches for resolving Venetoclax resistance.
Collapse
Affiliation(s)
- Mark Forsberg
- Department of Oncology, Montefiore Einstein Cancer Center, Bronx, NY
| | - Marina Konopleva
- Department of Oncology, Montefiore Einstein Cancer Center, Bronx, NY.
| |
Collapse
|
24
|
Mishra R, Zokaei Nikoo M, Veeraballi S, Singh A. Venetoclax and Hypomethylating Agent Combination in Myeloid Malignancies: Mechanisms of Synergy and Challenges of Resistance. Int J Mol Sci 2023; 25:484. [PMID: 38203655 PMCID: PMC10778677 DOI: 10.3390/ijms25010484] [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: 11/29/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
There has been a widespread adoption of hypomethylating agents (HMA: 5-Azacytidine (5-Aza)/decitabine) and venetoclax (Ven) for the treatment of acute myeloid leukemia (AML); however, the mechanisms behind the combination's synergy are poorly understood. Monotherapy often encounters resistance, leading to suboptimal outcomes; however, the combination of HMA and Ven has demonstrated substantial improvements in treatment responses. This study elucidates multiple synergistic pathways contributing to this enhanced therapeutic effect. Key mechanisms include HMA-mediated downregulation of anti-apoptotic proteins, notably MCL-1, and the priming of cells for Ven through the induction of genes encoding pro-apoptotic proteins such as Noxa. Moreover, Ven induces sensitization to HMA, induces overcoming resistance by inhibiting the DHODH enzyme, and disrupts antioxidant pathways (Nrf2) induced by HMA. The combination further disrupts oxidative phosphorylation in leukemia stem cells, amplifying the therapeutic impact. Remarkably, clinical studies have revealed a favorable response, particularly in patients harboring specific mutations, such as IDH1/2, NPM1, CEBPA, or ASXL1. This prompts future studies to explore the nuanced underpinnings of these synergistic mechanisms in AML patients with these molecular signatures.
Collapse
Affiliation(s)
- Rahul Mishra
- Department of Internal Medicine, Anne Arundel Medical Center, Annapolis, MD 21401, USA;
| | - Maedeh Zokaei Nikoo
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (M.Z.N.); (S.V.)
| | - Sindhusha Veeraballi
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (M.Z.N.); (S.V.)
| | - Abhay Singh
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (M.Z.N.); (S.V.)
| |
Collapse
|
25
|
Chan O, Walker AR. Novel therapies upon failure of HMA plus venetoclax. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:702-708. [PMID: 38066883 PMCID: PMC10727075 DOI: 10.1182/hematology.2023000456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The efficacy and tolerability of the combination of hypomethylating agents with venetoclax (HMA-VEN) in patients with newly diagnosed acute myeloid leukemia has been a practice-changing milestone in the field. However, treatment failure and relapse remain major barriers to prolonged survival. TP53 mutation is a predictor of primary induction failure and portends especially poor outcomes. Prelinical data suggest that VEN resistance stems from these genetic changes, which lead to increases in antiapoptotic proteins such as MCL-1 and BCLXL. For patients who discontinue HMA-VEN for reasons other than disease progression, such as post allotransplantation, infection, and personal preference, rechallenge with HMA-VEN at the time of relapse may be considered. For those who progress on HMA-VEN, clinical trials with novel agents or rational drug combinations are preferred if available. If no trial option is available, fit patients may benefit from intensive chemotherapy. Emerging therapies aim to overcome venetoclax resistance, target interactions that promote leukemogenesis, and harness the immune system to irradicate leukemic blasts and stem cells.
Collapse
Affiliation(s)
- Onyee Chan
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Alison R Walker
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
26
|
Leśniak M, Lipniarska J, Majka P, Lejman M, Zawitkowska J. Recent Updates in Venetoclax Combination Therapies in Pediatric Hematological Malignancies. Int J Mol Sci 2023; 24:16708. [PMID: 38069030 PMCID: PMC10706781 DOI: 10.3390/ijms242316708] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Venetoclax is a strongly effective B-cell lymphoma-2 inhibitor (BCL-2) with an ability to selectively restore the apoptotic potential of cancerous cells. It has been proven that in combination with immunotherapy, targeted therapies, and lower-intensity therapies such as hypomethylating agents (HMAs) or low-dose cytarabine (LDAC), the drug can improve overall outcomes for adult patients with acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and multiple myeloma (MM), amongst other hematological malignancies, but its benefit in pediatric hematology remains unclear. With a number of preclinical and clinical trials emerging, the newest findings suggest that in many cases of younger patients, venetoclax combination treatment can be well-tolerated, with a safety profile similar to that in adults, despite often leading to severe infections. Studies aim to determine the activity of BCL-2 inhibitor in the treatment of both primary and refractory acute leukemias in combination with standard and high-dose chemotherapy. Although more research is required to identify the optimal venetoclax-based regimen for the pediatric population and its long-term effects on patients' outcomes, it can become a potential therapeutic agent for pediatric oncology.
Collapse
Affiliation(s)
- Maria Leśniak
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (M.L.); (J.L.); (P.M.)
| | - Justyna Lipniarska
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (M.L.); (J.L.); (P.M.)
| | - Patrycja Majka
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (M.L.); (J.L.); (P.M.)
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
27
|
Chen L, Lowe B, Fletcher S. Tetrazole and acylsulfonamide bioisosteric replacements of the carboxylic acid in a dual MCL-1/BCL-x L inhibitor are tolerated. RSC Adv 2023; 13:34322-34334. [PMID: 38024975 PMCID: PMC10664828 DOI: 10.1039/d3ra05711a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Overexpression of the anti-apoptotic protein MCL-1 is associated with a plethora of human cancers, and it reduces the sensitivity of cancer cells to approved chemotherapies. Accordingly, the discovery of MCL-1 inhibitors is an active area of interest. Many inhibitors of the anti-apoptotic MCL-1 protein bear a crucial carboxylic acid that may engage Arg263 in the BH3-binding groove. We previously described the salicylic acid-based dual MCL-1/BCL-xL inhibitor 17cd, which is currently undergoing lead optimization. As part of that process, we wished to investigate bioisosteric replacement of 17cd's key carboxylic acid. Herein we describe the synthesis of a variety of analogues of a simpler analogue of 17cd presenting carboxylic acid surrogates. The acylsulfonamide and tetrazole motifs, which exhibit comparable pKas to the carboxylic acid function, displayed similar, or better, binding affinities to MCL-1 and BCL-xL as the corresponding carboxylic acid-containing lead. Our best compound was acylsulfonamide 7d with a Ki of 800 nM against MCL-1 and 1.82 mM against BCL-xL, and demonstrated an improved effect on the viability of the HL60 acute myeloid leukemia cell line relative to the parent carboxylic acid-containing dual inhibitor from which it was derived.
Collapse
Affiliation(s)
- Lijia Chen
- University of Maryland School of Pharmacy 20N Pine St Baltimore MD 21201 USA
| | - Brandon Lowe
- University of Maryland School of Pharmacy 20N Pine St Baltimore MD 21201 USA
| | - Steven Fletcher
- University of Maryland School of Pharmacy 20N Pine St Baltimore MD 21201 USA
- University of Maryland Greenebaum Cancer Center 20 S. Greene St. Baltimore MD 21201 USA
| |
Collapse
|
28
|
Brady PB, Sorensen BK, Risi RM, Curtin ML, Mantei RA, Florjancic AS, Mastracchio A, Ji C, Kunzer AR, Lai C, Storer GE, Chan VS, Henry RF, Souers AJ, Michaelides MR, Judd AS, Hansen TM. Enabling, Decagram-Scale Synthesis of Macrocyclic MCL-1 Inhibitor ABBV-467. J Org Chem 2023; 88:15562-15568. [PMID: 37909857 DOI: 10.1021/acs.joc.3c00939] [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: 11/03/2023]
Abstract
ABBV-467 is a highly potent and selective MCL-1 inhibitor that was advanced to a phase I clinical trial for the treatment of multiple myeloma. Due to its large size and structural complexity, ABBV-467 is a challenging synthetic target. Herein, we describe the synthesis of ABBV-467 on a decagram scale, which enabled preclinical characterization. The strategy is convergent and stereoselective, featuring a hindered biaryl cross coupling, enantioselective hydrogenation, and conformationally preorganized macrocyclization by C-O bond formation as key steps.
Collapse
Affiliation(s)
- Patrick B Brady
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Bryan K Sorensen
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Roberto M Risi
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Michael L Curtin
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Robert A Mantei
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Alan S Florjancic
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Anthony Mastracchio
- Global Medicinal Chemistry, Small Molecule Therapeutic and Platform Technologies, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | - Cheng Ji
- Global Medicinal Chemistry, Small Molecule Therapeutic and Platform Technologies, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | - Aaron R Kunzer
- Global Medicinal Chemistry, Small Molecule Therapeutic and Platform Technologies, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | - Chunqiu Lai
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Gregory E Storer
- Center of Catalysis, Process Research and Development, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | - Vincent S Chan
- Center of Catalysis, Process Research and Development, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | - Rodger F Henry
- Analytical Sciences, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Andrew J Souers
- Oncology Discovery, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | | | - Andrew S Judd
- Global Medicinal Chemistry, Small Molecule Therapeutic and Platform Technologies, AbbVie, Inc., North Chicago, Illinois 60064, United States
| | - T Matthew Hansen
- Centralized Organic Synthesis Group, Small Molecule Therapeutic and Platform Technologies, AbbVie Inc., North Chicago, Illinois 60064, United States
| |
Collapse
|
29
|
Santinelli E, Pascale MR, Xie Z, Badar T, Stahl MF, Bewersdorf JP, Gurnari C, Zeidan AM. Targeting apoptosis dysregulation in myeloid malignancies - The promise of a therapeutic revolution. Blood Rev 2023; 62:101130. [PMID: 37679263 DOI: 10.1016/j.blre.2023.101130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
In recent years, the therapeutic landscape of myeloid malignancies has been completely revolutionized by the introduction of several new drugs, targeting molecular alterations or pathways crucial for leukemia cells survival. Particularly, many agents targeting apoptosis have been investigated in both pre-clinical and clinical studies. For instance, venetoclax, a pro-apoptotic agent active on BCL-2 signaling, has been successfully used in the treatment of acute myeloid leukemia (AML). The impressive results achieved in this context have made the apoptotic pathway an attractive target also in other myeloid neoplasms, translating the experience of AML. Therefore, several drugs are now under investigation either as single or in combination strategies, due to their synergistic efficacy and capacity to overcome resistance. In this paper, we will review the mechanisms of apoptosis and the specific drugs currently used and under investigation for the treatment of myeloid neoplasia, identifying critical research necessities for the upcoming years.
Collapse
Affiliation(s)
- Enrico Santinelli
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, 00133 Rome, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Maria Rosaria Pascale
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Talha Badar
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Maximilian F Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jan P Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carmelo Gurnari
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
30
|
Milnerowicz S, Maszewska J, Skowera P, Stelmach M, Lejman M. AML under the Scope: Current Strategies and Treatment Involving FLT3 Inhibitors and Venetoclax-Based Regimens. Int J Mol Sci 2023; 24:15849. [PMID: 37958832 PMCID: PMC10647248 DOI: 10.3390/ijms242115849] [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: 09/23/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Acute myeloid leukemia (AML) is a disease that mainly affects elderly patients who are more often unfit for intensive chemotherapy (median age of diagnosis is 68). The regimens, including venetoclax, a highly specific BCL-2 (B-cell lymphoma-2) inhibitor, are a common alternative because of their safer profile and fewer side effects. However, the resistance phenomenon of leukemic cells necessitates the search for drugs that would help to overcome the resistance and improve treatment outcomes. One of the resistance mechanisms takes place through the upregulation of MCL-1 and BCL-XL, preventing BAX/BAK-driven MOMP (mitochondrial outer membrane permeabilization), thus stopping the apoptosis process. Possible partners for BCL-2 inhibitors may include inhibitors from the FLT3i (FMS-like tyrosine kinase-3 inhibitor) group. They resensitize cancer cells through the downregulation of MCL-1 expression in the FLT3 mutated cells, resulting in the stronger efficacy of BCL-2 inhibitors. Also, they provide an additional pathway for targeting the clonal cell. Both preclinical and clinical data suggest that the combination might show a synergistic effect and improve patients' outcomes. The aim of this review is to determine whether the combination of venetoclax and FLT3 inhibitors can impact the therapeutic approaches and what other agents they can be combined with.
Collapse
Affiliation(s)
- Szymon Milnerowicz
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (J.M.)
| | - Julia Maszewska
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (J.M.)
| | - Paulina Skowera
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (P.S.); (M.S.)
| | - Magdalena Stelmach
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (P.S.); (M.S.)
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (P.S.); (M.S.)
| |
Collapse
|
31
|
Jia Y, Han L, Ramage CL, Wang Z, Weng CC, Yang L, Colla S, Ma H, Zhang W, Andreeff M, Daver N, Jain N, Pemmaraju N, Bhalla K, Mustjoki S, Zhang P, Zheng G, Zhou D, Zhang Q, Konopleva M. Co-targeting BCL-XL and BCL-2 by PROTAC 753B eliminates leukemia cells and enhances efficacy of chemotherapy by targeting senescent cells. Haematologica 2023; 108:2626-2638. [PMID: 37078252 PMCID: PMC10542840 DOI: 10.3324/haematol.2022.281915] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 04/07/2023] [Indexed: 04/21/2023] Open
Abstract
BCL-XL and BCL-2 are key anti-apoptotic proteins and validated cancer targets. 753B is a novel BCL-XL/BCL-2 proteolysis targeting chimera (PROTAC) that targets both BCL-XL and BCL-2 to the von Hippel-Lindau (VHL) E3 ligase, leading to BCLX L/BCL-2 ubiquitination and degradation selectively in cells expressing VHL. Because platelets lack VHL expression, 753B spares on-target platelet toxicity caused by the first-generation dual BCL-XL/BCL-2 inhibitor navitoclax (ABT-263). Here, we report pre-clinical single-agent activity of 753B against different leukemia subsets. 753B effectively reduced cell viability and induced dose-dependent degradation of BCL-XL and BCL-2 in a subset of hematopoietic cell lines, acute myeloid leukemia (AML) primary samples, and in vivo patient-derived xenograft AML models. We further demonstrated the senolytic activity of 753B, which enhanced the efficacy of chemotherapy by targeting chemotherapy-induced cellular senescence. These results provide a pre-clinical rationale for the utility of 753B in AML therapy, and suggest that 753B could produce an added therapeutic benefit by overcoming cellular senescence-induced chemoresistance when combined with chemotherapy.
Collapse
Affiliation(s)
- Yannan Jia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai
| | - Lina Han
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cassandra L Ramage
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zhe Wang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Connie C Weng
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lei Yang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Helen Ma
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Weiguo Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kapil Bhalla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer center, Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology and Center for Innovative Drug Discovery, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
32
|
Short NJ, Nguyen D, Ravandi F. Treatment of older adults with FLT3-mutated AML: Emerging paradigms and the role of frontline FLT3 inhibitors. Blood Cancer J 2023; 13:142. [PMID: 37696819 PMCID: PMC10495326 DOI: 10.1038/s41408-023-00911-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/13/2023] Open
Abstract
FLT3 is the most frequently mutated gene in acute myeloid leukemia (AML), with FLT3 internal tandem duplication (ITD) mutations being associated with a more aggressive clinical course. While two large, randomized clinical trials have shown a survival benefit with the frontline use of an oral FLT3 inhibitor (midostaurin or quizartinib) in patients with FLT3-mutated AML, the role of FLT3 inhibitors in older adults with newly diagnosed FLT3-mutated AML remains unclear. A definitive improvement in survival has not been observed in intensively treated patients over 60 years of age receiving frontline FLT3 inhibitors. Furthermore, many patients with FLT3-mutated AML are unsuitable for intensive chemotherapy due to age and/or comorbidities, and this population represents a particular unmet need. For these older patients who are unfit for intensive approaches, azacitidine + venetoclax is a new standard of care and is used by many clinicians irrespective of FLT3 mutation status. However, FLT3-ITD mutations confer resistance to venetoclax and are a well-established mechanism of relapse to lower-intensity venetoclax-based regimens, leading to short durations of remission and poor survival. Preclinical and clinical data suggest synergy between FLT3 inhibitors and venetoclax, providing rationale for their combination. Novel strategies to safely incorporate FLT3 inhibitors into the standard hypomethylating agent + venetoclax backbone are now being explored in this older, less fit population with newly diagnosed FLT3-mutated AML, with encouraging early results. Herein, we discuss the frontline use of FLT3 inhibitors in older adults with FLT3-mutated AML, including the potential role of FLT3 inhibitors in combination with intensive chemotherapy and as part of novel, lower-intensity doublet and triplet regimens in this older population.
Collapse
Affiliation(s)
- Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Daniel Nguyen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
33
|
Hua L, Yang N, Li Y, Huang K, Jiang X, Liu F, Yu Z, Chen J, Lai J, Du J, Zeng H. Metformin sensitizes AML cells to venetoclax through endoplasmic reticulum stress-CHOP pathway. Br J Haematol 2023; 202:971-984. [PMID: 37409755 DOI: 10.1111/bjh.18968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/26/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Venetoclax inhibits acute myeloid leukaemia by inhibiting BCL-2 targeting, and a combination regimen with venetoclax has been explored. Although these regimens produce better clinical results, the vast majority of patients still suffer from disease recurrence or primary drug resistance. Metformin has been demonstrated to induce apoptosis in cancer cells. However, whether it can synergize with venetoclax and the underlying mechanisms of metformin-induced apoptosis are not fully understood. In this study, we investigated the effect of metformin and venetoclax on the growth of AML cells in vitro and in vivo. In both Molm13 and THP-1 cell lines, metformin and venetoclax synergistically inhibited the proliferation and induced apoptosis of leukaemia cells. Most importantly, the combination of metformin and venetoclax treatment significantly increased the expression levels of the endoplasmic reticulum (ER) stress-related marker CHOP, for example, in AML cell lines. Knockdown of CHOP markedly attenuated the metformin- and venetoclax-induced cell apoptosis. Moreover, the combination of metformin and venetoclax demonstrated prominent anti-leukaemia effects in xenograft models and bone marrow samples from AML patients. In summary, the combination of metformin and venetoclax showed enhanced anti-leukaemia activity with acceptable safety in AML patients, representing a new combinatorial strategy worth further clinical investigation to treat AML.
Collapse
Affiliation(s)
- Lei Hua
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Nianhui Yang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yue Li
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Kexiu Huang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xinya Jiang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Fangshu Liu
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhi Yu
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jie Chen
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jing Lai
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Juan Du
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Weidenauer K, Schmidt C, Rohde C, Pauli C, Blank MF, Heid D, Waclawiczek A, Corbacioglu A, Göllner S, Lotze M, Vierbaum L, Renders S, Krijgsveld J, Raffel S, Sauer T, Trumpp A, Pabst C, Müller-Tidow C, Janssen M. The ribosomal protein S6 kinase alpha-1 (RPS6KA1) induces resistance to venetoclax/azacitidine in acute myeloid leukemia. Leukemia 2023; 37:1611-1625. [PMID: 37414921 PMCID: PMC10400424 DOI: 10.1038/s41375-023-01951-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
Venetoclax/azacitidine combination therapy is effective in acute myeloid leukemia (AML) and tolerable for older, multimorbid patients. Despite promising response rates, many patients do not achieve sustained remission or are upfront refractory. Identification of resistance mechanisms and additional therapeutic targets represent unmet clinical needs. By using a genome-wide CRISPR/Cas9 library screen targeting 18,053 protein- coding genes in a human AML cell line, various genes conferring resistance to combined venetoclax/azacitidine treatment were identified. The ribosomal protein S6 kinase A1 (RPS6KA1) was among the most significantly depleted sgRNA-genes in venetoclax/azacitidine- treated AML cells. Addition of the RPS6KA1 inhibitor BI-D1870 to venetoclax/azacitidine decreased proliferation and colony forming potential compared to venetoclax/azacitidine alone. Furthermore, BI-D1870 was able to completely restore the sensitivity of OCI-AML2 cells with acquired resistance to venetoclax/azacitidine. Analysis of cell surface markers revealed that RPS6KA1 inhibition efficiently targeted monocytic blast subclones as a potential source of relapse upon venetoclax/azacitidine treatment. Taken together, our results suggest RPS6KA1 as mediator of resistance towards venetoclax/azacitidine and additional RPS6KA1 inhibition as strategy to prevent or overcome resistance.
Collapse
Affiliation(s)
- Katharina Weidenauer
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
| | - Christina Schmidt
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
| | - Christian Rohde
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Cornelius Pauli
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Division of Mechanisms Regulating Gene Expression, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maximilian F Blank
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Heid
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Alexander Waclawiczek
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Anika Corbacioglu
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
| | - Stefanie Göllner
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michelle Lotze
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lisa Vierbaum
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Simon Renders
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Jeroen Krijgsveld
- University of Heidelberg Medical Faculty, Heidelberg, Germany
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Raffel
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Caroline Pabst
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Maike Janssen
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany.
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
35
|
Oyogoa E, Traer E, Tyner J, Lachowiez C. Building on Foundations: Venetoclax-Based Combinations in the Treatment of Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:3589. [PMID: 37509251 PMCID: PMC10377106 DOI: 10.3390/cancers15143589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Frontline acute myeloid leukemia (AML) treatment is determined by a combination of patient and genetic factors. This includes patient fitness (i.e., comorbidities that increase the risk of treatment-related mortality) and genetic characteristics, including cytogenetic events and gene mutations. In older unfit patients, the standard of care treatment is typically venetoclax (VEN) combined with hypomethylating agents (HMA). Recently, several drugs have been developed targeting specific genomic subgroups of AML patients, enabling individualized therapy. This has resulted in investigations of doublet and triplet combinations incorporating VEN aimed at overcoming known resistance mechanisms and improving outcomes in older patients with AML. These combinations include isocitrate dehydrogenase-1/2 (IDH1/2) inhibitors (i.e., ivosidenib and enasidenib), fms-like tyrosine kinase 3 (FLT3) inhibitors (i.e., gilteritinib), anti-CD47 antibodies (i.e., magrolimab), mouse double minute-2 (MDM2) inhibitors, and p53 reactivators (i.e., eprenetapopt). This review summarizes ongoing trials aimed at overcoming known VEN resistance mechanisms and improving outcomes beyond that observed with HMA + VEN combinations in the treatment of AML.
Collapse
Affiliation(s)
- Emmanuella Oyogoa
- Department of Internal Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elie Traer
- Knight Cancer Institute, Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jeffrey Tyner
- Knight Cancer Institute, Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Curtis Lachowiez
- Knight Cancer Institute, Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
36
|
Mi RH, Chen L, Wang L, Ai H, Yin QS, Wei XD. [Retrospective analysis of the efficacy and safety of Venetoclax-based regimen in the treatment of 12 cases of acute myeloid leukemia with t (8; 21)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:501-504. [PMID: 37550208 PMCID: PMC10450551 DOI: 10.3760/cma.j.issn.0253-2727.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Indexed: 08/09/2023]
Affiliation(s)
- R H Mi
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou 450008, China
| | - L Chen
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou 450008, China
| | - L Wang
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou 450008, China
| | - H Ai
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou 450008, China
| | - Q S Yin
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou 450008, China
| | - X D Wei
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
37
|
Wang H, Zhou J, Ma X, Jiao C, Chen E, Wu Z, Zhang Y, Pan M, Cui J, Luan C, Ge J. Dexamethasone enhances venetoclax-induced apoptosis in acute myeloid leukemia cells. Med Oncol 2023; 40:193. [PMID: 37261571 DOI: 10.1007/s12032-023-02056-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/17/2023] [Indexed: 06/02/2023]
Abstract
Acute myeloid leukemia (AML) therapies have been significantly improved by the development of medicines that can target BCL-2. On the other hand, non-recurrent alterations in oncogenic pathways and gene expression patterns have already been linked to therapeutic resistance to venetoclax therapy. Bone marrow mesenchymal stromal cells (BM-MSCs) support leukemic cells in preventing chemotherapy-induced apoptosis by mitochondrial transfer in leukemic microenvironment. In this study, we investigated the enhancement of the antitumor effect of BCL-2 inhibitor venetoclax by dexamethasone. In particular, dexamethasone had no significant effect on the viability of AML cells, but dexamethasone combined with venetoclax could significantly increase the apoptosis of AML cells induced by venetoclax. When AML cells were co-cultured with BM-MSCs, dexamethasone combined with venetoclax showed additional anti-tumor effect compared to venetoclax alone. Venetoclax increased reactive oxygen species level in co-cultured AML cells, contributed to transfer more mitochondria from BM-MSCs to AML cells and protect AML cells from apoptosis. Dexamethasone combined with venetoclax induced more apoptosis, but dexamethasone reduced the venetoclax-induced reactive oxygen species level in AML cells and reduced the transfer of mitochondria from BM-MSCs to AML cells. This may lead to a diminished protective effect of BM-MSCs on AML cells. Together, our findings indicated that venetoclax in combination with dexamethasone could be a promising therapy in AML.
Collapse
Affiliation(s)
- Haixia Wang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Junjie Zhou
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaoyu Ma
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Changqing Jiao
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Enbo Chen
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhonghui Wu
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yan Zhang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Mengya Pan
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Jianling Cui
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Chengxin Luan
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Jian Ge
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
38
|
El-Cheikh J, Bidaoui G, Saleh M, Moukalled N, Abou Dalle I, Bazarbachi A. Venetoclax: A New Partner in the Novel Treatment Era for Acute Myeloid Leukemia and Myelodysplastic Syndrome. Clin Hematol Int 2023:10.1007/s44228-023-00041-x. [PMID: 37071328 DOI: 10.1007/s44228-023-00041-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/09/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) are two closely related blood cancers that are more frequent in older adults. AML is the most common type of adult acute leukemia, and MDS is characterized by ineffective blood cell production and abnormalities in the bone marrow and blood. Both can be resistant to treatment, often due to dysfunction in the process of apoptosis, the body's natural mechanism for cell death. Venetoclax, an orally-administered medication that selectively targets the BCL-2 protein, has shown promise in enhancing treatment sensitivity in some hematological malignancies by reducing the apoptotic threshold. This review aims to evaluate the effectiveness of venetoclax in treating AML and MDS, as well as potential mechanisms of resistance to the medication. METHODS A literature search was conducted utilizing PUBMED to capture all relevant research articles on the use of venetoclax as a therapy for both diseases. The MeSH terms "acute myeloid leukemia", "myelodysplastic syndrome" and "venetoclax" were searched. Furthermore, Clinicaltrials.gov was accessed to ensure the inclusion of all ongoing clinical trials. RESULTS Although Venetoclax showed modest results as a single-agent therapy in AML, venetoclax-based combination therapies? mainly with hypomethylating agents or low-dose cytarabine? yielded significantly positive results. Preliminary results oN the use of venetoclax-based combination therapy with HMA, mainly azacitidine, in unfit high-risk MDS also yielded optimistic results. Identification of mutations for which various drugs have been approved has spurred active investigation of venetoclax in combination trials. CONCLUSION Venetoclax-based combination therapies have been shown to induce rapid responses and increase overall survival in AML patients unfit for intensive chemotherapy. These therapies are also yielding positive preliminary results in high-risk MDS patients in phase I trials. Resistance to venetoclax and drug-related toxicity are two main obstacles that need to be overcome to reap the full benefits of this therapy.
Collapse
Affiliation(s)
- Jean El-Cheikh
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
- Department of Internal Medicine, Medical Center, Bone Marrow Transplantation Program, American University of Beirut, P.O. Box 113-6044, Beirut, Lebanon.
| | - Ghassan Bidaoui
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mustafa Saleh
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Nour Moukalled
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Iman Abou Dalle
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Bazarbachi
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
39
|
Opydo M, Mlyczyńska A, Mlyczyńska E, Rak A, Kolaczkowska E. Synergistic Action of MCL-1 Inhibitor with BCL-2/BCL-XL or MAPK Pathway Inhibitors Enhances Acute Myeloid Leukemia Cell Apoptosis and Differentiation. Int J Mol Sci 2023; 24:ijms24087180. [PMID: 37108344 PMCID: PMC10138770 DOI: 10.3390/ijms24087180] [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: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by excessive proliferation of abnormal myeloid precursors accompanied by a differentiation block and inhibition of apoptosis. Increased expression of an anti-apoptotic MCL-1 protein was shown to be critical for the sustained survival and expansion of AML cells. Therefore, herein, we examined the pro-apoptotic and pro-differentiating effects of S63845, a specific inhibitor of MCL-1, in a single-agent treatment and in combination with BCL-2/BCL-XL inhibitor, ABT-737, in two AML cell lines: HL-60 and ML-1. Additionally, we determined whether inhibition of the MAPK pathway had an impact on the sensitivity of AML cells to S63845. To assess AML cells' apoptosis and differentiation, in vitro studies were performed using PrestoBlue assay, Coulter electrical impedance method, flow cytometry, light microscopy and Western blot techniques. S63845 caused a concentration-dependent decrease in the viability of HL-60 and ML-1 cells and increased the percentage of apoptotic cells. Combined treatment with S63845 and ABT-737 or MAPK pathway inhibitor enhanced apoptosis but also induced differentiation of tested cells, as well as altering the expression of the MCL-1 protein. Taken together, our data provide the rationale for further studies regarding the use of MCL-1 inhibitor in combination with other pro-survival protein inhibitors.
Collapse
Affiliation(s)
- Małgorzata Opydo
- Laboratory of Experimental Hematology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Anna Mlyczyńska
- Laboratory of Experimental Hematology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-387 Krakow, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Elzbieta Kolaczkowska
- Laboratory of Experimental Hematology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
40
|
Satta T, Li L, Chalasani SL, Hu X, Nkwocha J, Sharma K, Kmieciak M, Rahmani M, Zhou L, Grant S. Dual mTORC1/2 Inhibition Synergistically Enhances AML Cell Death in Combination with the BCL2 Antagonist Venetoclax. Clin Cancer Res 2023; 29:1332-1343. [PMID: 36652560 PMCID: PMC10073266 DOI: 10.1158/1078-0432.ccr-22-2729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/29/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
PURPOSE Acute myelogenous leukemia (AML) is an aggressive disease with a poor outcome. We investigated mechanisms by which the anti-AML activity of ABT-199 (venetoclax) could be potentiated by dual mTORC1/TORC2 inhibition. EXPERIMENTAL DESIGN Venetoclax/INK128 synergism was assessed in various AML cell lines and primary patient AML samples in vitro. AML cells overexpressing MCL-1, constitutively active AKT, BAK, and/or BAX knockout, and acquired venetoclax resistance were investigated to define mechanisms underlying interactions. The antileukemic efficacy of this regimen was also examined in xenograft and patient-derived xenograft (PDX) models. RESULTS Combination treatment with venetoclax and INK128 (but not the mTORC1 inhibitor rapamycin) dramatically enhanced cell death in AML cell lines. Synergism was associated with p-AKT and p-4EBP1 downregulation and dependent upon MCL-1 downregulation and BAK/BAX upregulation as MCL-1 overexpression and BAX/BAK knockout abrogated cell death. Constitutive AKT activation opposed synergism between venetoclax and PI3K or AKT inhibitors, but not INK128. Combination treatment also synergistically induced cell death in venetoclax-resistant AML cells. Similar events occurred in primary patient-derived leukemia samples but not normal CD34+ cells. Finally, venetoclax and INK128 co-treatment displayed increased antileukemia effects in in vivo xenograft and PDX models. CONCLUSIONS The venetoclax/INK128 regimen exerts significant antileukemic activity in various preclinical models through mechanisms involving MCL-1 downregulation and BAK/BAX activation, and offers potential advantages over PI3K or AKT inhibitors in cells with constitutive AKT activation. This regimen is active against primary and venetoclax-resistant AML cells, and in in vivo AML models. Further investigation of this strategy appears warranted.
Collapse
Affiliation(s)
- Toshihisa Satta
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Laboratory Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lin Li
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. USA
| | - Sri Lakshmi Chalasani
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. USA
| | - Xiaoyan Hu
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. USA
| | - Jewel Nkwocha
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. USA
| | - Kanika Sharma
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. USA
| | - Maciej Kmieciak
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohamed Rahmani
- Department of Molecular Biology and Genetics, College of Medicine & Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Liang Zhou
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA. 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
| |
Collapse
|
41
|
Kuusanmäki H, Dufva O, Vähä-Koskela M, Leppä AM, Huuhtanen J, Vänttinen I, Nygren P, Klievink J, Bouhlal J, Pölönen P, Zhang Q, Adnan-Awad S, Mancebo-Pérez C, Saad J, Miettinen J, Javarappa KK, Aakko S, Ruokoranta T, Eldfors S, Heinäniemi M, Theilgaard-Mönch K, Wartiovaara-Kautto U, Keränen M, Porkka K, Konopleva M, Wennerberg K, Kontro M, Heckman CA, Mustjoki S. Erythroid/megakaryocytic differentiation confers BCL-XL dependency and venetoclax resistance in acute myeloid leukemia. Blood 2023; 141:1610-1625. [PMID: 36508699 PMCID: PMC10651789 DOI: 10.1182/blood.2021011094] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 09/20/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Myeloid neoplasms with erythroid or megakaryocytic differentiation include pure erythroid leukemia, myelodysplastic syndrome with erythroid features, and acute megakaryoblastic leukemia (FAB M7) and are characterized by poor prognosis and limited treatment options. Here, we investigate the drug sensitivity landscape of these rare malignancies. We show that acute myeloid leukemia (AML) cells with erythroid or megakaryocytic differentiation depend on the antiapoptotic protein B-cell lymphoma (BCL)-XL, rather than BCL-2, using combined ex vivo drug sensitivity testing, genetic perturbation, and transcriptomic profiling. High-throughput screening of >500 compounds identified the BCL-XL-selective inhibitor A-1331852 and navitoclax as highly effective against erythroid/megakaryoblastic leukemia cell lines. In contrast, these AML subtypes were resistant to the BCL-2 inhibitor venetoclax, which is used clinically in the treatment of AML. Consistently, genome-scale CRISPR-Cas9 and RNAi screening data demonstrated the striking essentiality of BCL-XL-encoding BCL2L1 but not BCL2 or MCL1, for the survival of erythroid/megakaryoblastic leukemia cell lines. Single-cell and bulk transcriptomics of patient samples with erythroid and megakaryoblastic leukemias identified high BCL2L1 expression compared with other subtypes of AML and other hematological malignancies, where BCL2 and MCL1 were more prominent. BCL-XL inhibition effectively killed blasts in samples from patients with AML with erythroid or megakaryocytic differentiation ex vivo and reduced tumor burden in a mouse erythroleukemia xenograft model. Combining the BCL-XL inhibitor with the JAK inhibitor ruxolitinib showed synergistic and durable responses in cell lines. Our results suggest targeting BCL-XL as a potential therapy option in erythroid/megakaryoblastic leukemias and highlight an AML subgroup with potentially reduced sensitivity to venetoclax-based treatments.
Collapse
MESH Headings
- Animals
- Mice
- Humans
- Proto-Oncogene Proteins c-bcl-2/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- bcl-X Protein/genetics
- Leukemia, Megakaryoblastic, Acute/drug therapy
- Leukemia, Megakaryoblastic, Acute/genetics
- Lymphoma, B-Cell
- Cell Differentiation
- Apoptosis
Collapse
Affiliation(s)
- Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Olli Dufva
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Aino-Maija Leppä
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Division of Stem Cells and Cancer, German Cancer Research Center and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Ida Vänttinen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Petra Nygren
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Jonas Bouhlal
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Petri Pölönen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Qi Zhang
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shady Adnan-Awad
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Cristina Mancebo-Pérez
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Joseph Saad
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Juho Miettinen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Komal K. Javarappa
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Sofia Aakko
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tanja Ruokoranta
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Samuli Eldfors
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Kim Theilgaard-Mönch
- Biotech Research & Innovation Centre and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Department of Hematology and Finsen Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulla Wartiovaara-Kautto
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Mikko Keränen
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Kimmo Porkka
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Marina Konopleva
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Mika Kontro
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
42
|
Kelvin JM, Jain J, Thapa A, Qui M, Birnbaum LA, Moore SG, Zecca H, Summers RJ, Costanza E, Uricoli B, Wang X, Jui NT, Fu H, Du Y, DeRyckere D, Graham DK, Dreaden EC. Constitutively synergistic multiagent drug formulations targeting MERTK, FLT3, and BCL-2 for treatment of AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.531236. [PMID: 36993676 PMCID: PMC10054973 DOI: 10.1101/2023.03.13.531236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Although high-dose, multi-agent chemotherapy has improved leukemia survival rates in recent years, treatment outcomes remain poor in high-risk subsets, including acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) in infants. Development of new, more effective therapies for these patients is therefore an urgent, unmet clinical need. To address this challenge, we developed a nanoscale combination drug formulation that exploits ectopic expression of MERTK tyrosine kinase and dependency on BCL-2 family proteins for leukemia cell survival in pediatric AML and MLL- rearranged precursor B-cell ALL (infant ALL). In a novel, high-throughput combination drug screen, the MERTK/FLT3 inhibitor MRX-2843 synergized with venetoclax and other BCL-2 family protein inhibitors to reduce AML cell density in vitro . Neural network models based on drug exposure and target gene expression were used to identify a classifier predictive of drug synergy in AML. To maximize the therapeutic potential of these findings, we developed a combination monovalent liposomal drug formulation that maintains ratiometric drug synergy in cell-free assays and following intracellular delivery. The translational potential of these nanoscale drug formulations was confirmed in a genotypically diverse set of primary AML patient samples and both the magnitude and frequency of synergistic responses were not only maintained but were improved following drug formulation. Together, these findings demonstrate a systematic, generalizable approach to combination drug screening, formulation, and development that maximizes therapeutic potential, was effectively applied to develop a novel nanoscale combination therapy for treatment of AML, and could be extended to other drug combinations or diseases in the future.
Collapse
|
43
|
Past, Present, and a Glance into the Future of Multiple Myeloma Treatment. Pharmaceuticals (Basel) 2023; 16:ph16030415. [PMID: 36986514 PMCID: PMC10056051 DOI: 10.3390/ph16030415] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Multiple myeloma (MM) is a challenging hematological cancer which typically grows in bone marrow. MM accounts for 10% of hematological malignancies and 1.8% of cancers. The recent treatment strategies have significantly improved progression-free survival for MM patients in the last decade; however, a relapse for most MM patients is inevitable. In this review we discuss current treatment, important pathways for proliferation, survival, immune suppression, and resistance that could be targeted for future treatments.
Collapse
|
44
|
Peris I, Romero-Murillo S, Martínez-Balsalobre E, Farrington CC, Arriazu E, Marcotegui N, Jiménez-Muñoz M, Alburquerque-Prieto C, Torres-López A, Fresquet V, Martínez-Climent JA, Mateos MC, Cayuela ML, Narla G, Odero MD, Vicente C. Activation of the PP2A-B56α heterocomplex synergizes with venetoclax therapies in AML through BCL2 and MCL1 modulation. Blood 2023; 141:1047-1059. [PMID: 36455198 PMCID: PMC10023731 DOI: 10.1182/blood.2022016466] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
Venetoclax combination therapies are becoming the standard of care in acute myeloid leukemia (AML). However, the therapeutic benefit of these drugs in older/unfit patients is limited to only a few months, highlighting the need for more effective therapies. Protein phosphatase 2A (PP2A) is a tumor suppressor phosphatase with pleiotropic functions that becomes inactivated in ∼70% of AML cases. PP2A promotes cancer cell death by modulating the phosphorylation state in a variety of proteins along the mitochondrial apoptotic pathway. We therefore hypothesized that pharmacological PP2A reactivation could increase BCL2 dependency in AML cells and, thus, potentiate venetoclax-induced cell death. Here, by using 3 structurally distinct PP2A-activating drugs, we show that PP2A reactivation synergistically enhances venetoclax activity in AML cell lines, primary cells, and xenograft models. Through the use of gene editing tools and pharmacological approaches, we demonstrate that the observed therapeutic synergy relies on PP2A complexes containing the B56α regulatory subunit, of which expression dictates response to the combination therapy. Mechanistically, PP2A reactivation enhances venetoclax-driven apoptosis through simultaneous inhibition of antiapoptotic BCL2 and extracellular signal-regulated kinase signaling, with the latter decreasing MCL1 protein stability. Finally, PP2A targeting increases the efficacy of the clinically approved venetoclax and azacitidine combination in vitro, in primary cells, and in an AML patient-derived xenograft model. These preclinical results provide a scientific rationale for testing PP2A-activating drugs with venetoclax combinations in AML.
Collapse
Affiliation(s)
- Irene Peris
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Silvia Romero-Murillo
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| | - Elena Martínez-Balsalobre
- Cancer and Aging Group, Hospital Universitario Virgen de la Arrixaca, and Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Caroline C. Farrington
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI
| | - Elena Arriazu
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Nerea Marcotegui
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | - Marta Jiménez-Muñoz
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | | | | | - Vicente Fresquet
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose A. Martínez-Climent
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Maria C. Mateos
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Hematology Service, Hospital Universitario de Navarra, Pamplona, Spain
| | - Maria L. Cayuela
- Cancer and Aging Group, Hospital Universitario Virgen de la Arrixaca, and Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI
| | - Maria D. Odero
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Vicente
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
45
|
Aid Z, Robert E, Lopez CK, Bourgoin M, Boudia F, Le Mene M, Riviere J, Baille M, Benbarche S, Renou L, Fagnan A, Thirant C, Federici L, Touchard L, Lecluse Y, Jetten A, Geoerger B, Lapillonne H, Solary E, Gaudry M, Meshinchi S, Pflumio F, Auberger P, Lobry C, Petit A, Jacquel A, Mercher T. High caspase 3 and vulnerability to dual BCL2 family inhibition define ETO2::GLIS2 pediatric leukemia. Leukemia 2023; 37:571-579. [PMID: 36585521 PMCID: PMC10583253 DOI: 10.1038/s41375-022-01800-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022]
Abstract
Pediatric acute myeloid leukemia expressing the ETO2::GLIS2 fusion oncogene is associated with dismal prognosis. Previous studies have shown that ETO2::GLIS2 can efficiently induce leukemia development associated with strong transcriptional changes but those amenable to pharmacological targeting remained to be identified. By studying an inducible ETO2::GLIS2 cellular model, we uncovered that de novo ETO2::GLIS2 expression in human cells led to increased CASP3 transcription, CASP3 activation, and cell death. Patient-derived ETO2::GLIS2+ leukemic cells expressed both high CASP3 and high BCL2. While BCL2 inhibition partly inhibited ETO2::GLIS2+ leukemic cell proliferation, BH3 profiling revealed that it also sensitized these cells to MCL1 inhibition indicating a functional redundancy between BCL2 and MCL1. We further show that combined inhibition of BCL2 and MCL1 is mandatory to abrogate disease progression using in vivo patient-derived xenograft models. These data reveal that a transcriptional consequence of ETO2::GLIS2 expression includes a positive regulation of the pro-apoptotic CASP3 and associates with a vulnerability to combined targeting of two BCL2 family members providing a novel therapeutic perspective for this aggressive pediatric AML subgroup.
Collapse
Affiliation(s)
- Zakia Aid
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Elie Robert
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Cécile K Lopez
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France.
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France.
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| | - Maxence Bourgoin
- Team "Myeloid Malignancies and Multiple Myeloma", Université Côte d'Azur, INSERM U1065/C3M, 06204, Nice, France
| | - Fabien Boudia
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Melchior Le Mene
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Julie Riviere
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Marie Baille
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Salima Benbarche
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
| | - Laurent Renou
- Unité de Recherche (UMR)-E008 Stabilité Génétique, Cellules Souches et Radiations, Team Niche and Cancer in Hematopoiesis, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Université de Paris-Université Paris-Saclay, Fontenay-aux-Roses, 92260, France
| | - Alexandre Fagnan
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Cécile Thirant
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Laetitia Federici
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Laure Touchard
- Unité Mixte de Service - Analyse Moléculaire Modélisation et Imagerie de la maladie Cancéreuse (UMS AMMICA), Gustave Roussy Cancer Campus, 94800, Villejuif, France
| | - Yann Lecluse
- Unité Mixte de Service - Analyse Moléculaire Modélisation et Imagerie de la maladie Cancéreuse (UMS AMMICA), Gustave Roussy Cancer Campus, 94800, Villejuif, France
| | - Anton Jetten
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Birgit Geoerger
- Gustave Roussy Cancer Campus, Pediatric and Adolescent Oncology Department, INSERM U1015, Université Paris Saclay, 94800, Villejuif, France
| | - Hélène Lapillonne
- Pediatric Hematology and Oncology Department, Armand Trousseau Hospital, AP-HP, Sorbonne University, UMRS_938, CONECT-AML, 75012, Paris, France
| | - Eric Solary
- INSERM U1287, Gustave Roussy Cancer Campus, 94800, Villejuif, France
| | - Muriel Gaudry
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Françoise Pflumio
- Unité de Recherche (UMR)-E008 Stabilité Génétique, Cellules Souches et Radiations, Team Niche and Cancer in Hematopoiesis, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Université de Paris-Université Paris-Saclay, Fontenay-aux-Roses, 92260, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, 75010, Paris, France
| | - Patrick Auberger
- Team "Myeloid Malignancies and Multiple Myeloma", Université Côte d'Azur, INSERM U1065/C3M, 06204, Nice, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, 75010, Paris, France
| | - Camille Lobry
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France
- INSERM U944, CNRS UMR7212, Institut de Recherche Saint Louis and Université de Paris, 75010, Paris, France
| | - Arnaud Petit
- Gustave Roussy Cancer Campus, Pediatric and Adolescent Oncology Department, INSERM U1015, Université Paris Saclay, 94800, Villejuif, France
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Arnaud Jacquel
- Team "Myeloid Malignancies and Multiple Myeloma", Université Côte d'Azur, INSERM U1065/C3M, 06204, Nice, France.
| | - Thomas Mercher
- INSERM U1170, Gustave Roussy Cancer Campus, Université Paris Saclay, PEDIAC program, 94800, Villejuif, France.
- Equipe labellisée Ligue Nationale Contre le Cancer, 75013, Paris, France.
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, 75010, Paris, France.
| |
Collapse
|
46
|
BH3 mimetics and TKI combined therapy for Chronic Myeloid Leukemia. Biochem J 2023; 480:161-176. [PMID: 36719792 DOI: 10.1042/bcj20210608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 02/01/2023]
Abstract
Chronic myeloid leukemia (CML) was considered for a long time one of the most hostile leukemia that was incurable for most of the patients, predominantly due to the extreme resistance to chemotherapy. Part of the resistance to cell death (apoptosis) is the result of increased levels of anti-apoptotic and decreased levels of pro-apoptotic member of the BCL-2 family induced by the BCR-ABL1 oncoprotein. BCR-ABL1 is a constitutively active tyrosine kinase responsible for initiating multiple and oncogenic signaling pathways. With the development of specific BCR-ABL1 tyrosine kinase inhibitors (TKIs) CML became a much more tractable disease. Nevertheless, TKIs do not cure CML patients and a substantial number of them develop intolerance or become resistant to the treatment. Therefore, novel anti-cancer strategies must be developed to treat CML patients independently or in combination with TKIs. Here, we will discuss the mechanisms of BCR-ABL1-dependent and -independent resistance to TKIs and the use of BH3-mimetics as a potential tool to fight CML.
Collapse
|
47
|
Tong X, Zhou F. Integrated bioinformatic analysis of mitochondrial metabolism-related genes in acute myeloid leukemia. Front Immunol 2023; 14:1120670. [PMID: 37138869 PMCID: PMC10149950 DOI: 10.3389/fimmu.2023.1120670] [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: 12/10/2022] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Background Acute myeloid leukemia (AML) is a common hematologic malignancy characterized by poor prognoses and high recurrence rates. Mitochondrial metabolism has been increasingly recognized to be crucial in tumor progression and treatment resistance. The purpose of this study was to examined the role of mitochondrial metabolism in the immune regulation and prognosis of AML. Methods In this study, mutation status of 31 mitochondrial metabolism-related genes (MMRGs) in AML were analyzed. Based on the expression of 31 MMRGs, mitochondrial metabolism scores (MMs) were calculated by single sample gene set enrichment analysis. Differential analysis and weighted co-expression network analysis were performed to identify module MMRGs. Next, univariate Cox regression and the least absolute and selection operator regression were used to select prognosis-associated MMRGs. A prognosis model was then constructed using multivariate Cox regression to calculate risk score. We validated the expression of key MMRGs in clinical specimens using immunohistochemistry (IHC). Then differential analysis was performed to identify differentially expressed genes (DEGs) between high- and low-risk groups. Functional enrichment, interaction networks, drug sensitivity, immune microenvironment, and immunotherapy analyses were also performed to explore the characteristic of DEGs. Results Given the association of MMs with prognosis of AML patients, a prognosis model was constructed based on 5 MMRGs, which could accurately distinguish high-risk patients from low-risk patients in both training and validation datasets. IHC results showed that MMRGs were highly expressed in AML samples compared to normal samples. Additionally, the 38 DEGs were mainly related to mitochondrial metabolism, immune signaling, and multiple drug resistance pathways. In addition, high-risk patients with more immune-cell infiltration had higher Tumor Immune Dysfunction and Exclusion scores, indicating poor immunotherapy response. mRNA-drug interactions and drug sensitivity analyses were performed to explore potential druggable hub genes. Furthermore, we combined risk score with age and gender to construct a prognosis model, which could predict the prognosis of AML patients. Conclusion Our study provided a prognostic predictor for AML patients and revealed that mitochondrial metabolism is associated with immune regulation and drug resistant in AML, providing vital clues for immunotherapies.
Collapse
|
48
|
Bouligny IM, Maher KR, Grant S. Augmenting Venetoclax Activity Through Signal Transduction in AML. JOURNAL OF CELLULAR SIGNALING 2023; 4:1-12. [PMID: 36911757 PMCID: PMC9997087 DOI: 10.33696/signaling.4.085] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Venetoclax, a small-molecule B-cell lymphoma 2 (BCL-2) inhibitor, selectively eradicates leukemic stem cells (LSCs). While venetoclax has revolutionized the treatment of acute myeloid leukemia (AML), treatment failure and disease relapse are common. Mechanisms underlying venetoclax resistance are surprisingly heterogeneous. Venetoclax resistance encompasses a spectrum of genetic and epigenetic changes, with numerous pathways contributing to the upregulation of additional anti-apoptotic proteins. In this review, we address the mechanisms of venetoclax resistance in the context of signal transduction. We emphasize how aberrant cell signaling impairs apoptosis and predisposes to venetoclax failure. Commonly activated pathways, such as FLT3, PI3K/AKT/mTOR, and RAS, contribute to upregulated anti-apoptotic mediators and are frequently responsible for refractory disease or disease relapse. We highlight novel combination strategies aimed at disabling constitutively active signal transduction to augment response and overcome venetoclax resistance.
Collapse
Affiliation(s)
- Ian Michael Bouligny
- Virginia Commonwealth University Massey Cancer Center, Division of Hematology and Oncology, Department of Internal Medicine, 1300 E. Marshall St., Richmond, VA, USA
| | - Keri Renee Maher
- Virginia Commonwealth University Massey Cancer Center, Division of Hematology and Oncology, Department of Internal Medicine, 1300 E. Marshall St., Richmond, VA, USA
| | - Steven Grant
- Virginia Commonwealth University Massey Cancer Center, Division of Hematology and Oncology, Department of Internal Medicine, 1300 E. Marshall St., Richmond, VA, USA
| |
Collapse
|
49
|
Janssen M, Schmidt C, Bruch PM, Blank MF, Rohde C, Waclawiczek A, Heid D, Renders S, Göllner S, Vierbaum L, Besenbeck B, Herbst SA, Knoll M, Kolb C, Przybylla A, Weidenauer K, Ludwig AK, Fabre M, Gu M, Schlenk RF, Stölzel F, Bornhäuser M, Röllig C, Platzbecker U, Baldus C, Serve H, Sauer T, Raffel S, Pabst C, Vassiliou G, Vick B, Jeremias I, Trumpp A, Krijgsveld J, Müller-Tidow C, Dietrich S. Venetoclax synergizes with gilteritinib in FLT3 wild-type high-risk acute myeloid leukemia by suppressing MCL-1. Blood 2022; 140:2594-2610. [PMID: 35857899 DOI: 10.1182/blood.2021014241] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 11/20/2022] Open
Abstract
BCL-2 inhibition has been shown to be effective in acute myeloid leukemia (AML) in combination with hypomethylating agents or low-dose cytarabine. However, resistance and relapse represent major clinical challenges. Therefore, there is an unmet need to overcome resistance to current venetoclax-based strategies. We performed high-throughput drug screening to identify effective combination partners for venetoclax in AML. Overall, 64 antileukemic drugs were screened in 31 primary high-risk AML samples with or without venetoclax. Gilteritinib exhibited the highest synergy with venetoclax in FLT3 wild-type AML. The combination of gilteritinib and venetoclax increased apoptosis, reduced viability, and was active in venetoclax-azacitidine-resistant cell lines and primary patient samples. Proteomics revealed increased FLT3 wild-type signaling in specimens with low in vitro response to the currently used venetoclax-azacitidine combination. Mechanistically, venetoclax with gilteritinib decreased phosphorylation of ERK and GSK3B via combined AXL and FLT3 inhibition with subsequent suppression of the antiapoptotic protein MCL-1. MCL-1 downregulation was associated with increased MCL-1 phosphorylation of serine 159, decreased phosphorylation of threonine 161, and proteasomal degradation. Gilteritinib and venetoclax were active in an FLT3 wild-type AML patient-derived xenograft model with TP53 mutation and reduced leukemic burden in 4 patients with FLT3 wild-type AML receiving venetoclax-gilteritinib off label after developing refractory disease under venetoclax-azacitidine. In summary, our results suggest that combined inhibition of FLT3/AXL potentiates venetoclax response in FLT3 wild-type AML by inducing MCL-1 degradation. Therefore, the venetoclax-gilteritinib combination merits testing as a potentially active regimen in patients with high-risk FLT3 wild-type AML.
Collapse
Affiliation(s)
- Maike Janssen
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christina Schmidt
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter-Martin Bruch
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Maximilian F Blank
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Christian Rohde
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Alexander Waclawiczek
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Daniel Heid
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Simon Renders
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Stefanie Göllner
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lisa Vierbaum
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Birgit Besenbeck
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sophie A Herbst
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Mareike Knoll
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carolin Kolb
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Adriana Przybylla
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Katharina Weidenauer
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Anne Kathrin Ludwig
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Margarete Fabre
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
| | - Muxin Gu
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
| | - Richard F Schlenk
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Friedrich Stölzel
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Christoph Röllig
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Uwe Platzbecker
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Claudia Baldus
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Hubert Serve
- Hematology-Oncology, Department of Medicine II, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Simon Raffel
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Caroline Pabst
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - George Vassiliou
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
| | - Binje Vick
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Cancer Consortium, Partner Site Munich, Munich, Germany
- Department of Pediatrics, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Sascha Dietrich
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
50
|
Pelosi E, Castelli G, Testa U. The Growing Role of the BH3 Mimetic Drug Venetoclax in the Therapy of Acute Myeloid Leukemia. Mediterr J Hematol Infect Dis 2022; 14:e2022080. [PMID: 36425147 PMCID: PMC9652018 DOI: 10.4084/mjhid.2022.080] [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: 09/17/2022] [Accepted: 10/17/2022] [Indexed: 01/25/2023] Open
Abstract
Despite recent progress, acute myeloid leukemia (AML) remains a disease associated with poor prognosis, particularly in older AML patients unfit to tolerate intensive chemotherapy treatment. The development and introduction in the therapy of Venetoclax (VEN), a potent BH3 mimetic targeting the antiapoptotic protein BCL-2, inducing apoptosis of leukemic cells, has shown to be a promising treatment for newly diagnosed, relapsed, and refractory AML patients ineligible for induction chemotherapy. Combination treatments using Ventoclax and a hypomethylating agent (azacitidine or decitabine) or low-intensity chemotherapy have shown in newly diagnosed patients variable response rates, with highly responsive patients with NPM1, IDH1-IDH2, TET2, and RUNX1 mutations and with scarcely responsive patients with FLT3, TP53 and ASXL1 mutations, complex karyotypes, and secondary AMLs. Patients with refractory/relapsing disease are less responsive to Venetoclax-based regimens. However, in the majority of patients, the responses have only a limited duration, and the development of resistance is frequently observed. Therefore, understanding the resistance mechanisms is crucial for developing new strategies and identifying rational drug combination regimens. In this context, two strategies seem to be promising: (i) triplet therapies based on the combined administration of Venetoclax, a hypomethylating agent (or low-dose chemotherapy), and an agent targeting a specific genetic alteration of leukemic cells (i.e., FLT3 inhibitors in FLT3-mutated AMLs) or an altered signaling pathway; (ii) combination therapies based on the administration of two BH3 mimetics (i.e., BCL-2 +MCL-1 mimetics) and a hypomethylating agent.
Collapse
Affiliation(s)
- Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanita, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanita, Rome, Italy
| | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanita, Rome, Italy
| |
Collapse
|