1
|
Hao Q, Zhao W, Li Z, Lai Y, Wang Y, Yang Q, Zhang L. Combination therapy and dual-target inhibitors based on cyclin-dependent kinases (CDKs): Emerging strategies for cancer therapy. Eur J Med Chem 2025; 289:117465. [PMID: 40037064 DOI: 10.1016/j.ejmech.2025.117465] [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: 01/04/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Cyclin-dependent kinases (CDKs) are pivotal regulators of the cell cycle and transcriptional machinery, making them attractive targets for cancer therapy. While CDK inhibitors have demonstrated promising clinical outcomes, they also face challenges in enhancing efficacy, particularly in overcoming drug resistance. Combination therapies have emerged as a key strategy to augment the effectiveness of CDK inhibitors when used alongside other kinase inhibitors or non-kinase-targeted agents. Dual-target inhibitors that simultaneously inhibit CDKs and other oncogenic drivers are gaining attention, offering novel avenues to optimize cancer therapy. Based on the structural characterization and biological functions of CDKs, this review comprehensively examines the structure-activity relationship (SAR) of existing dual-target CDK inhibitors from a drug design perspective. We also thoroughly investigate the preclinical studies and clinical translational potential of combination therapies and dual-target inhibitors. Tailoring CDK inhibitors to specific cancer subtypes and therapeutic settings will inspire innovative approaches for the next generation of CDK-related therapies, ultimately improving patient survival.
Collapse
Affiliation(s)
- Qi Hao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wenzhe Zhao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yue Lai
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yan Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qianqian Yang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
2
|
Jin L, Jiang X, Li Y, Xiang S, Pei R, Lu Y, Jiang L. Flavokawain a induces cell cycle arrest through CDT1-dependent p27 regulation and synergizes with venetoclax in acute myeloid leukemia. Toxicol Appl Pharmacol 2025; 498:117305. [PMID: 40113097 DOI: 10.1016/j.taap.2025.117305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
The poor prognosis of patients with acute myeloid leukemia (AML) is largely ascribed to the deficiency of persistently effective therapies. Despite the recent approval of targeted drugs such as the BCL-2 inhibitor venetoclax, the clinical benefit is limited due to the development of resistance. The use of natural products is emerging as a feasible strategy to treat malignant diseases including AML. Flavokawain A (FKA) is a naturally occurring chalcone isolated from the root of kava and possesses extensive antitumor activities. The therapeutic potential of FKA in AML remains unknown. In the present study, we found that treatment with FKA reduced the viability in four AML cell lines in dose- and time-dependent manners. The anti-AML activity of venetoclax was significantly potentiated by FKA. Mechanistically, FKA induced G1 phase arrest in AML cells along with CDT1 downregulation and p27 upregulation. Knockdown of CDT1 increased the expression of p27, leading to the inhibition on cell viability. Both p27 upregulation and viability inhibition caused by FKA was partially rescued by CDT1 overexpression. The therapeutic effect of FKA alone or in combination with venetoclax was verified in primary blasts from AML patients, further strengthening the clinical relevance of the current study. Therefore, our data suggest that FKA can be considered as a potential therapeutic agent in the treatment of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Sulfonamides/pharmacology
- Sulfonamides/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Drug Synergism
- Cell Line, Tumor
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Cyclin-Dependent Kinase Inhibitor p27/genetics
- Cell Cycle Checkpoints/drug effects
- Cell Survival/drug effects
- Antineoplastic Agents/pharmacology
- Dose-Response Relationship, Drug
- Flavonoids/pharmacology
Collapse
Affiliation(s)
- Lili Jin
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, 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, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 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, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Sumeng Xiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, 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
| | - Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China.
| | - Lei Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
3
|
Yang J, Friedman R. Inhibition of FLT3-induced signalling in refractory acute myeloid leukaemia. Bioorg Chem 2025; 160:108424. [PMID: 40209351 DOI: 10.1016/j.bioorg.2025.108424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
Mutations in FLT3 make this receptor tyrosine kinase overactive. Such mutations found in ∼30 % of the patients who suffer from acute myeloid leukaemia (AML). FLT3 mediates signalling networks that lead to cell proliferation and survival. FLT3 inhibitors are used to treat AML but patients who are treated with them typically become resistant. Such resistance often emerges through secondary mutations which either restore the activity of FLT3 in the presence of drugs or activate a key player in a signalling network such as NRAS. We had developed AML-specific cell lines resistant to two advanced FLT3 inhibitors: gilteritinib and FF-10101. Resistance in these cell lines proceeds though different mechanisms. In this study, we followed on the efficacy of five FLT3 inhibitors (gilteritinib, FF-10101 and three promising inhibitors that are being developed), two pan-PI3K inhibitors (one of which also inhibits mTOR) and two c-KIT inhibitors in order to examine the significance of different signalling cascades in FLT3+-AML. In addition, we used molecular modelling and quantum chemistry calculations to explain why specific FLT3 mutations affect some inhibitors more than others. Two novel FLT3 inhibitors were found to be only weakly affected by resistance mutations against gilteritinib and FF-10101. The efficacy of most FLT3 inhibitors was only weakly (or not at all) affected by the NRAS/G12C activating mutation. Finally, no non-FLT3 inhibitor has shown sufficient efficacy in the cells, suggesting the central role of FLT3 in FLT3-mutated AML.
Collapse
Affiliation(s)
- Jingmei Yang
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar SE-39231, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar SE-39231, Sweden.
| |
Collapse
|
4
|
Wen Y, Yang X, Li S, Huang L, Chen J, Tan L, Ma X, Zhu Y, Li Z, Shan C, Zhang C, Zhang Q, Liang M, Zhang H, Liu T. Targeting CDK4/6 suppresses colorectal cancer by destabilizing YAP1. MedComm (Beijing) 2025; 6:e70103. [PMID: 39968498 PMCID: PMC11832431 DOI: 10.1002/mco2.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025] Open
Abstract
Colorectal cancer (CRC) is among the most prevalent and deadly cancers worldwide. The Yes-associated protein 1 (YAP1) is frequently dysregulated in cancers, contributing to cancer stemness, chemoresistance, and cancer-related death. However, strategies directly targeting YAP1 have not yet been successful because of the lack of active binding pockets and unregulated toxicity. In this study, our Food and Drug Administration (FDA)-approved drug screening reveals that abemaciclib, a cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, dramatically promotes the proteasome-dependent degradation of YAP1, thereby inhibiting tumor progression in CRC cells and patient-derived xenograft models. We further identify deubiquitinating enzyme 3 (DUB3) as the bona fide deubiquitinase of YAP1 in CRC. Mechanistically, CDK4/6 directly phosphorylates DUB3 at Ser41, activating DUB3 to deubiquitinate and stabilize YAP1. Conversely, loss of Ser41 phosphorylation by CDK4/6 inhibition or Ser41A mutation, promotes YAP1 degradation and suppresses YAP1-driven tumor progression. Histological analysis shows a positive correlation between DUB3 and YAP1 expression in CRC specimens. Collectively, our study uncovers a novel oncogenic role of the CDK4/6-DUB3 pathway, which promotes YAP1 stabilization and tumor-promoting function, highlighting that targeting CDK4/6 offers a potential therapeutic strategy for CRC with aberrantly upregulated DUB3 and YAP1.
Collapse
Affiliation(s)
- Yalei Wen
- Research Institute for Maternal and Child Health, The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of PharmacyJinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Xiao Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Shengrong Li
- Research Institute for Maternal and Child Health, The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of PharmacyJinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Lei Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Jiayi Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Lirong Tan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Xiuqing Ma
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Yingjie Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Zhengqiu Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of PharmacyJinan UniversityGuangzhouChina
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug ResearchNankai UniversityTianjinChina
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical CenterNankai UniversityTianjinChina
| | - Qiushi Zhang
- Research Institute for Maternal and Child Health, The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of PharmacyJinan UniversityGuangzhouChina
- Research Institute for Maternal and Child Health, The Affiliated Guangdong Second Provincial General HospitalJinan UniversityGuangzhouChina
| | - Mingchao Liang
- The Affiliated Shunde Hospital of Jinan UniversityFoshanChina
| | - Haoxing Zhang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, College of Life Sciences and OceanographyShenzhen UniversityShenzhenChina
| | - Tongzheng Liu
- Research Institute for Maternal and Child Health, The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of PharmacyJinan UniversityGuangzhouChina
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
5
|
Scheiblecker L, Klampfl T, Doma E, Nebenfuehr S, Torres-Quesada O, Strich S, Heller G, Werdenich D, Tschulenk W, Zojer M, Bellutti F, Schirripa A, Zöchbauer-Müller S, Valent P, Walter I, Stefan E, Sexl V, Kollmann K. CDK6 kinase inhibition unmasks metabolic dependencies in BCR::ABL1+ leukemia. Cell Death Dis 2025; 16:107. [PMID: 39966356 PMCID: PMC11836434 DOI: 10.1038/s41419-025-07434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/15/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Metabolic reprogramming and cell cycle deregulation are hallmarks of cancer cells. The cell cycle kinase CDK6 has recently been implicated in a wide range of hematopoietic malignancies. We here investigate the role of CDK6 in the regulation of cellular metabolism in BCR::ABL1+ leukemic cells. Our study, using gene expression data and ChIP-Seq analysis, highlights the contribution of CDK6 kinase activity in the regulation of oxidative phosphorylation. Our findings imply a competition for promoter interaction of CDK6 with the master regulator of mitochondrial respiration, NRF-1. In line, cells lacking kinase active CDK6 display altered mitochondria morphology with a defective electron transport chain. The enhanced cytoplasm/mitochondria ATP ratio paralleled by high pyruvate and lactate levels indicate a metabolic switch to glycolysis. Accordingly, combinatorial treatment of leukemic cells including imatinib resistant cells with the CDK4/6 inhibitor palbociclib and the glycolysis inhibitor 2-deoxyglucose (2-DG) enhanced apoptosis, while blocking cell proliferation in leukemic cells. These data may open a new therapeutic avenue for hematologic malignancies with high CDK6 expression by exploiting metabolic vulnerabilities unmasked by blocking CDK6 kinase activity that might even be able to overcome imatinib resistance.
Collapse
MESH Headings
- Humans
- Cyclin-Dependent Kinase 6/metabolism
- Cyclin-Dependent Kinase 6/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Fusion Proteins, bcr-abl/genetics
- Glycolysis/drug effects
- Protein Kinase Inhibitors/pharmacology
- Piperazines/pharmacology
- Cell Line, Tumor
- Mitochondria/metabolism
- Mitochondria/drug effects
- Apoptosis/drug effects
- Oxidative Phosphorylation/drug effects
- Cell Proliferation/drug effects
- Deoxyglucose/pharmacology
- Pyridines/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- K562 Cells
Collapse
Affiliation(s)
- Lisa Scheiblecker
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thorsten Klampfl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eszter Doma
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Omar Torres-Quesada
- Institute of Medical Biochemistry, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| | - Sophie Strich
- Institute of Medical Biochemistry, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Gerwin Heller
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Daniela Werdenich
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Waltraud Tschulenk
- Institute of Morphology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Florian Bellutti
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Alessia Schirripa
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sabine Zöchbauer-Müller
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Ingrid Walter
- Institute of Morphology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eduard Stefan
- Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Sieberer H, Luciano M, Amend D, Blöchl C, Eglseer A, Steinkellner A, Rieser S, Andosch A, Steiner P, Hummer L, Krenn PW, Dang HH, Huber CG, Aberger F, Neuper T, Horejs-Hoeck J. Inhibition of NLRP3 enhances pro-apoptotic effects of FLT3 inhibition in AML. Cell Commun Signal 2025; 23:53. [PMID: 39875995 PMCID: PMC11773904 DOI: 10.1186/s12964-025-02046-w] [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: 09/30/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
FLT3 mutations occur in approximately 25% of all acute myeloid leukemia (AML) patients. While several FLT3 inhibitors have received FDA approval, their use is currently limited to combination therapies with chemotherapy, as resistance occurs, and efficacy decreases when the inhibitors are used alone. Given the highly heterogeneous nature of AML, there is an urgent need for novel targeted therapies that address the disease from multiple angles. Recent research has identified the NLRP3 inflammasome as a potential new driver in AML. Here, we investigated the efficacy of different NLRP3 inhibitors in targeting AML cells in vitro. Our findings reveal that NLRP3 inhibition induces cell cycle arrest as well as signs of senescence in multiple AML cell lines. In contrast, NLRP3 inhibition selectively induced apoptosis in FLT3 mutant AML cell lines, but not in FLT3 wild-type AML cells. Moreover, we show that NLRP3 inhibition impairs FLT3 signaling by reducing both FLT3 expression as well as downstream signaling in FLT3 mutant cells. A database analysis revealed a strong positive correlation between FLT3 and NLRP3 in cancer, which was particularly evident in AML patients. Strikingly, the simultaneous inhibition of NLRP3 and FLT3 markedly enhanced apoptosis in FLT3-ITD mutant AML cells, but not in FLT3 wild-type cells. In summary, this study reveals a promising combined therapeutic strategy specifically targeting NLRP3/FLT3-ITD positive AML blasts in vitro, highlighting a potential new avenue for AML treatment.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Apoptosis/drug effects
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- Cell Line, Tumor
- Signal Transduction/drug effects
- Mutation
Collapse
Affiliation(s)
- Helene Sieberer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Michela Luciano
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Diana Amend
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Constantin Blöchl
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Anna Eglseer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Alina Steinkellner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Sebastian Rieser
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Ancuela Andosch
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Philip Steiner
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, Linz, 4020, Austria
| | - Laura Hummer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Peter W Krenn
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Hieu-Hoa Dang
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Christian G Huber
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria.
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria.
- Cancer Cluster Salzburg, Salzburg, 5020, Austria.
| |
Collapse
|
7
|
Perzolli A, Steinebach C, Krönke J, Gütschow M, Zwaan CM, Barneh F, Heidenreich O. PROTAC-Mediated GSPT1 Degradation Impairs the Expression of Fusion Genes in Acute Myeloid Leukemia. Cancers (Basel) 2025; 17:211. [PMID: 39857993 PMCID: PMC11763475 DOI: 10.3390/cancers17020211] [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: 12/18/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Proteolysis targeting chimeras (PROTACs) are heterobifunctional small molecules that utilize the ubiquitin-proteasome system to selectively degrade target proteins. This innovative technology has shown remarkable efficacy and specificity in degrading oncogenic proteins and has progressed through various stages of preclinical and clinical development for hematologic malignancies, including adult acute myeloid leukemia (AML). However, the application of PROTACs in pediatric AML remains largely unexplored. METHODS In this study, we show the potent effect of GSPT1 degradation against AML cells induced by either a GSPT1-selective cereblon modulator CC-90009 or by an off-target effect caused by a CDK6-PROTAC named GU3341. RESULTS Both in vitro and ex vivo experiments revealed that GSPT1 degradation significantly inhibited tumor growth, induced cell cycle arrest, and triggered apoptosis in two pediatric AML subtypes characterized by RUNX1::RUNX1T1 and FUS::ERG fusion genes. Furthermore, the degradation of GSPT1 impaired the expression of RUNX1::RUNX1T1 and its cooperating transcription factors RUNX1 and ERG. Similarly, GSPT1 degradation also reduced FUS::ERG fusion transcript levels in AML cells harboring the translocation t(16;24)(p11:q22). CONCLUSIONS These findings suggest a new role of GSPT1 in regulating leukemic transcriptional networks and open a new therapeutic strategy to target leukemic fusion genes in pediatric AML patients.
Collapse
Affiliation(s)
- Alicia Perzolli
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (A.P.); (C.M.Z.); (F.B.)
- Department of Pediatric Oncology, Erasmus MC/Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands
| | - Christian Steinebach
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; (C.S.); (M.G.)
| | - Jan Krönke
- Department of Internal Medicine III, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany;
| | - Michael Gütschow
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; (C.S.); (M.G.)
| | - C. Michel Zwaan
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (A.P.); (C.M.Z.); (F.B.)
- Department of Pediatric Oncology, Erasmus MC/Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands
| | - Farnaz Barneh
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (A.P.); (C.M.Z.); (F.B.)
| | - Olaf Heidenreich
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (A.P.); (C.M.Z.); (F.B.)
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Department of Hematology, University of Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
8
|
Wu M, Wang W, Mao X, Wu Y, Jin Y, Liu T, Lu Y, Dai H, Zeng S, Huang W, Wang Y, Yao X, Che J, Ying M, Dong X. Discovery of a potent CDKs/FLT3 PROTAC with enhanced differentiation and proliferation inhibition for AML. Eur J Med Chem 2024; 275:116539. [PMID: 38878515 DOI: 10.1016/j.ejmech.2024.116539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 07/12/2024]
Abstract
AML is an aggressive malignancy of immature myeloid progenitor cells. Discovering effective treatments for AML through cell differentiation and anti-proliferation remains a significant challenge. Building on previous studies on CDK2 PROTACs with differentiation-inducing properties, this research aims to enhance CDKs degradation through structural optimization to facilitate the differentiation and inhibit the proliferation of AML cells. Compound C3, featuring a 4-methylpiperidine ring linker, effectively degraded CDK2 with a DC50 value of 18.73 ± 10.78 nM, and stimulated 72.77 ± 3.51 % cell differentiation at 6.25 nM in HL-60 cells. Moreover, C3 exhibited potent anti-proliferative activity against various AML cell types. Degradation selectivity analysis indicated that C3 could be endowed with efficient degradation of CDK2/4/6/9 and FLT3, especially FLT3-ITD in MV4-11 cells. These findings propose that C3 combined targeting CDK2/4/6/9 and FLT3 with enhanced differentiation and proliferation inhibition, which holds promise as a potential treatment for AML.
Collapse
Affiliation(s)
- Mingfei Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Wei Wang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences; Zhejiang University, Cancer Center; Zhejiang University School of Medicine Children'sHospital, Division of Hematology-Oncology, Hangzhou, 310058, PR China
| | - Xinfei Mao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences; Zhejiang University, Cancer Center; Zhejiang University School of Medicine Children'sHospital, Division of Hematology-Oncology, Hangzhou, 310058, PR China
| | - Yiquan Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yuyuan Jin
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310058, PR China
| | - Tao Liu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yan Lu
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Haibin Dai
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Shenxin Zeng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310058, PR China
| | - Wenhai Huang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310058, PR China
| | - Yuwei Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macau, 999078, PR China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Meidan Ying
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences; Zhejiang University, Cancer Center; Zhejiang University School of Medicine Children'sHospital, Division of Hematology-Oncology, Hangzhou, 310058, PR China.
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China.
| |
Collapse
|
9
|
Yang T, Ke H, Liu J, An X, Xue J, Ning J, Hao F, Xiong L, Chen C, Wang Y, Zheng J, Gao B, Bao Z, Gong K, Zhang L, Zhang F, Guo S, Li QX. Narazaciclib, a novel multi-kinase inhibitor with potent activity against CSF1R, FLT3 and CDK6, shows strong anti-AML activity in defined preclinical models. Sci Rep 2024; 14:9032. [PMID: 38641704 PMCID: PMC11031590 DOI: 10.1038/s41598-024-59650-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 04/12/2024] [Indexed: 04/21/2024] Open
Abstract
CSF1R is a receptor tyrosine kinase responsible for the growth/survival/polarization of macrophages and overexpressed in some AML patients. We hypothesized that a novel multi-kinase inhibitor (TKi), narazaciclib (HX301/ON123300), with high potency against CSF1R (IC50 ~ 0.285 nM), would have anti-AML effects. We tested this by confirming HX301's high potency against CSF1R (IC50 ~ 0.285 nM), as well as other kinases, e.g. FLT3 (IC50 of ~ 19.77 nM) and CDK6 (0.53 nM). An in vitro proliferation assay showed that narazaciclib has a high growth inhibitory effect in cell cultures where CSF1R or mutant FLT3-ITD variants that may be proliferation drivers, including primary macrophages (IC50 of 72.5 nM) and a subset of AML lines (IC50 < 1.5 μM). In vivo pharmacology modeling of narazaciclib using five AML xenografts resulted in: inhibition of MV4-11 (FLT3-ITD) subcutaneous tumor growth and complete suppression of AM7577-PDX (FLT3-ITD/CSF1Rmed) systemic growth, likely due to the suppression of FLT3-ITD activity; complete suppression of AM8096-PDX (CSF1Rhi/wild-type FLT3) growth, likely due to the inhibition of CSF1R ("a putative driver"); and nonresponse of both AM5512-PDX and AM7407-PDX (wild-type FLT3/CSF1Rlo). Significant leukemia load reductions in bone marrow, where disease originated, were also achieved in both responders (AM7577/AM8096), implicating that HX301 might be a potentially more effective therapy than those only affecting peripheral leukemic cells. Altogether, narazaciclib can potentially be a candidate treatment for a subset of AML with CSF1Rhi and/or mutant FLT3-ITD variants, particularly second generation FLT3 inhibitor resistant variants.
Collapse
Affiliation(s)
- Tao Yang
- Hanx Biopharmaceuticals, Ltd., Wuhan, Hubei, PRC, China
| | - Hang Ke
- Hanx Biopharmaceuticals, Ltd., Wuhan, Hubei, PRC, China
| | - Jinping Liu
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | - Xiaoyu An
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | - Jia Xue
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | | | - Feng Hao
- Kyinno Biotechnology, Ltd., Beijing, PRC, China
| | | | - Cen Chen
- Hanx Biopharmaceuticals, Ltd., Wuhan, Hubei, PRC, China
| | - Yueying Wang
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | - Jia Zheng
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | - Bing Gao
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | | | - Kefeng Gong
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | - Lei Zhang
- Hanx Biopharmaceuticals, Ltd., Wuhan, Hubei, PRC, China
| | - Faming Zhang
- Hanx Biopharmaceuticals, Ltd., Wuhan, Hubei, PRC, China
| | - Sheng Guo
- Crown Bioscience, Inc., Taicang, Jiangsu, PRC, USA
| | - Qi-Xiang Li
- Hanx Biopharmaceuticals, Ltd., Wuhan, Hubei, PRC, China.
| |
Collapse
|
10
|
Foy R, Lew KX, Saurin AT. The search for CDK4/6 inhibitor biomarkers has been hampered by inappropriate proliferation assays. NPJ Breast Cancer 2024; 10:19. [PMID: 38438376 PMCID: PMC10912267 DOI: 10.1038/s41523-024-00624-8] [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: 04/19/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
CDK4/6 inhibitors are effective at treating advanced HR+ /HER2- breast cancer, however biomarkers that can predict response are urgently needed. We demonstrate here that previous large-scale screens designed to identify which tumour types or genotypes are most sensitive to CDK4/6 inhibitors have misrepresented the responsive cell lines because of a reliance on metabolic proliferation assays. CDK4/6-inhibited cells arrest in G1 but continue to grow in size, thereby producing more mitochondria. We show that this growth obscures the arrest using ATP-based proliferation assays but not if DNA-based assays are used instead. Furthermore, lymphoma lines, previously identified as the most sensitive, simply appear to respond the best using ATP-based assays because they fail to overgrow during the G1 arrest. Similarly, the CDK4/6 inhibitor abemaciclib appears to inhibit proliferation better than palbociclib because it also restricts cellular overgrowth through off-target effects. DepMap analysis of screening data using reliable assay types, demonstrates that palbociclib-sensitive cell types are also sensitive to Cyclin D1, CDK4 and CDK6 knockout/knockdown, whereas the palbociclib-resistant lines are sensitive to Cyclin E1, CDK2 and SKP2 knockout/knockdown. Potential biomarkers of palbociclib-sensitive cells are increased expression of CCND1 and RB1, and reduced expression of CCNE1 and CDKN2A. Probing DepMap with similar data from metabolic assays fails to reveal these associations. Together, this demonstrates why CDK4/6 inhibitors, and any other anti-cancer drugs that arrest the cell cycle but permit continued cell growth, must now be re-screened against a wide-range of cell types using an appropriate proliferation assay. This would help to better inform clinical trials and to identify much needed biomarkers of response.
Collapse
Affiliation(s)
- Reece Foy
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Kah Xin Lew
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian T Saurin
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
11
|
Long X, Jiang H, Liu Z, Liu J, Hu R. Long noncoding RNA LINC00675 drives malignancy in acute myeloid leukemia via the miR-6809 -CDK6 axis. Pathol Res Pract 2024; 255:155221. [PMID: 38422911 DOI: 10.1016/j.prp.2024.155221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Hematological malignancies such as acute myeloid leukemia (AML) have a low cure rate and a high recurrence rate. Long noncoding RNAs (LNCs) are essential regulators of tumorigenesis and progression. The role of lncRNA LINC00675 in AML has rarely been reported. This study revealed elevated LINC00675 expression in AML that promotes proliferation and inhibits apoptosis. Mechanistically, LINC00675 combines with miR-6809 to promote the expression of CDK6 in vitro and in vivo. Immune-checkpoint genes were expressed more highly in LINC00675-high patients. A high level of LINC00675 expression may make patients more susceptible to palbociclib treatments. In conclusion, our study demonstrated that LINC00675 is an oncogenic lncRNA that enhances the malignancy of AML by upregulating CDK6 expression through miR-6809 sponging, providing a new perspective and feasible target for the diagnosis and treatment of AML.
Collapse
Affiliation(s)
- Xinyi Long
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang 110000, China; Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Huinan Jiang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Zhuogang Liu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Rong Hu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
12
|
Wang H, Ba J, Kang Y, Gong Z, Liang T, Zhang Y, Qi J, Wang J. Recent Progress in CDK4/6 Inhibitors and PROTACs. Molecules 2023; 28:8060. [PMID: 38138549 PMCID: PMC10745860 DOI: 10.3390/molecules28248060] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Cell division in eukaryotes is a highly regulated process that is critical to the life of a cell. Dysregulated cell proliferation, often driven by anomalies in cell Cyclin-dependent kinase (CDK) activation, is a key pathological mechanism in cancer. Recently, selective CDK4/6 inhibitors have shown clinical success, particularly in treating advanced-stage estrogen receptor (ER)-positive and human epidermal growth factor receptor 2 (HER2)-negative breast cancer. This review provides an in-depth analysis of the action mechanism and recent advancements in CDK4/6 inhibitors, categorizing them based on their structural characteristics and origins. Furthermore, it explores proteolysis targeting chimers (PROTACs) targeting CDK4/6. We hope that this review could be of benefit for further research on CDK4/6 inhibitors and PROTACs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jianguo Qi
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University Jinming Campus, Kaifeng 475004, China
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University Jinming Campus, Kaifeng 475004, China
| |
Collapse
|
13
|
Tang H, Yeo D, De Souza K, Ahmad O, Shafiq T, Ofor O, Anand A, Karim S, Khan S, Madhusudan S. Clinical Impact of CDK4/6 Inhibitors in De Novo or PR- or Very Elderly Post-Menopausal ER+/HER2- Advanced Breast Cancers. Cancers (Basel) 2023; 15:5164. [PMID: 37958338 PMCID: PMC10647609 DOI: 10.3390/cancers15215164] [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: 09/11/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
The CDK4/6 inhibitors significantly increase progression-free survival (PFS) in ER+/HER2- advanced breast cancer patients. In clinical trials, overall survival (OS) improvement has been demonstrated for ribociclib and abemaciclib but not for palbociclib. We undertook a real-world evaluation of PFS and OS in 227 post-menopausal patients who received first-line CDK4/6 inhibitors. There is no significant difference in median PFS (27.5 months vs. 25.7 months, p = 0.3) or median OS (49.5 months vs. 50.2 months, p = 0.67) in patients who received either palbociclib or ribociclib, respectively. De novo disease is significantly associated with prolonged median PFS and median OS compared with recurrence disease (47.1 months vs. 20.3 months (p = 0.0002) and 77.4 months vs. 37.3 months (p = 0.0003), respectively). PR- tumours have significantly reduced median PFS and OS compared with PR+ disease (19.2 months vs. 38 months (p = 0.003) and 34.3 months vs. 62.6 months (p = 0.02), respectively). In the very elderly (>80 years), median PFS and OS are significantly shorter compared with patients who are 65 years or below (14.5 months vs. 30.2 months (p = 0.01), and 77.4 months vs. 29.6 months (p = 0.009), respectively) in the palbociclib group. Our data suggest that the benefit in the very elderly is limited, and PR+/de novo disease obtains the maximum survival benefit.
Collapse
Affiliation(s)
- Hiu Tang
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Daniel Yeo
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Karen De Souza
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK
| | - Omar Ahmad
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Tahir Shafiq
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Okezie Ofor
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Anjana Anand
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Syed Karim
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Sarah Khan
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
| | - Srinivasan Madhusudan
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK (S.K.)
- Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| |
Collapse
|
14
|
Takahashi S. Combination Therapies with Kinase Inhibitors for Acute Myeloid Leukemia Treatment. Hematol Rep 2023; 15:331-346. [PMID: 37367084 DOI: 10.3390/hematolrep15020035] [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: 12/05/2022] [Revised: 03/10/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Targeting kinase activity is considered to be an attractive therapeutic strategy to overcome acute myeloid leukemia (AML) since aberrant activation of the kinase pathway plays a pivotal role in leukemogenesis through abnormal cell proliferation and differentiation block. Although clinical trials for kinase modulators as single agents remain scarce, combination therapies are an area of therapeutic interest. In this review, the author summarizes attractive kinase pathways for therapeutic targets and the combination strategies for these pathways. Specifically, the review focuses on combination therapies targeting the FLT3 pathways, as well as PI3K/AKT/mTOR, CDK and CHK1 pathways. From a literature review, combination therapies with the kinase inhibitors appear more promising than monotherapies with individual agents. Therefore, the development of efficient combination therapies with kinase inhibitors may result in effective therapeutic strategies for AML.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Laboratory Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| |
Collapse
|
15
|
Perrone S, Ottone T, Zhdanovskaya N, Molica M. How acute myeloid leukemia (AML) escapes from FMS-related tyrosine kinase 3 (FLT3) inhibitors? Still an overrated complication? CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:223-238. [PMID: 37457126 PMCID: PMC10344728 DOI: 10.20517/cdr.2022.130] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/21/2023] [Accepted: 04/11/2023] [Indexed: 07/18/2023]
Abstract
FMS-related tyrosine kinase 3 (FLT3) mutations, present in about 25%-30% of acute myeloid leukemia (AML) patients, constitute one of the most frequently detected mutations in these patients. The binding of FLT3L to FLT3 activates the phosphatidylinositol 3-kinase (PI3K) and RAS pathways, producing increased cell proliferation and the inhibition of apoptosis. Two types of FLT3 mutations exist: FLT3-ITD and FLT3-TKD (point mutations in D835 and I836 or deletion of codon I836). A class of drugs, tyrosine-kinase inhibitors (TKI), targeting mutated FLT3, is already available with 1st and 2nd generation molecules, but only midostaurin and gilteritinib are currently approved. However, the emergence of resistance or the selection of clones not responding to FLT3 inhibitors has become an important clinical dilemma, as the duration of clinical responses is generally limited to a few months. This review analyzes the insights into mechanisms of resistance to TKI and poses a particular view on the clinical relevance of this phenomenon. Has resistance been overlooked? Indeed, FLT3 inhibitors have significantly contributed to reducing the negative impact of FLT3 mutations on the prognosis of AML patients who are no longer considered at high risk by the European LeukemiaNet (ELN) 2022. Finally, several ongoing efforts to overcome resistance to FLT3-inhibitors will be presented: new generation FLT3 inhibitors in monotherapy or combined with standard chemotherapy, hypomethylating drugs, or IDH1/2 inhibitors, Bcl2 inhibitors; novel anti-human FLT3 monoclonal antibodies (e.g., FLT3/CD3 bispecific antibodies); FLT3-CAR T-cells; CDK4/6 kinase inhibitor (e.g., palbociclib).
Collapse
Affiliation(s)
- Salvatore Perrone
- Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, Latina 04100, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, the University of Rome “Tor Vergata”, Rome 00100 Italy
- Neuro-Oncohematology, Santa Lucia Foundation, I.R.C.C.S., Rome 00100, Italy
| | - Nadezda Zhdanovskaya
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome 00161, Italy
| | - Matteo Molica
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, Rome 00144, Italy
| |
Collapse
|
16
|
Xie X, Zhang W, Zhou X, Ye Z, Wang H, Qiu Y, Pan Y, Hu Y, Li L, Chen Z, Yang W, Lu Y, Zou S, Li Y, Bai X. Abemaciclib drives the therapeutic differentiation of acute myeloid leukaemia stem cells. Br J Haematol 2023; 201:940-953. [PMID: 36916190 DOI: 10.1111/bjh.18735] [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: 12/06/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/15/2023]
Abstract
Self-renewal and differentiation arrest are two features of leukaemia stem cells (LSCs) responsible for the high relapse rate of acute myeloid leukaemia (AML). To screen drugs to overcome differentiation blockade for AML, we conducted screening of 2040 small molecules from a library of United States Food and Drug Administration-approved drugs and found that the cyclin-dependent kinase (CDK)4/6 inhibitor, abemaciclib, exerts high anti-leukaemic activity. Abemaciclib significantly suppressed proliferation and promoted the differentiation of LSCs in vitro. Abemaciclib also efficiently induced differentiation and impaired self-renewal of LSCs, thus reducing the leukaemic cell burden and improving survival in various preclinical animal models, including patient-derived xenografts. Importantly, abemaciclib strongly enhanced anti-tumour effects in combination with venetoclax, a B-cell lymphoma 2 (Bcl-2) inhibitor. This treatment combination led to a marked decrease in LSC-enriched populations and resulted in a synergistic anti-leukaemic effect. Target screening revealed that in addition to CDK4/6, abemaciclib bound to and inhibited CDK9, consequently attenuating the protein levels of global p-Ser2 RNA Polymerase II (Pol II) carboxy terminal domain (CTD), Myc, Bcl-2, and myeloid cell leukaemia-1 (Mcl-1), which was important for the anti-AML effect of abemaciclib. Collectively, these data provide a strong rationale for the clinical evaluation of abemaciclib to induce LSC differentiation and treat highly aggressive AML as well as other advanced haematological malignancies.
Collapse
Affiliation(s)
- Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wuju Zhang
- Department of Oncology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixin Ye
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hao Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yating Pan
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Luyao Li
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuanzhuan Chen
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wanwen Yang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yao Lu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuxin Zou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Ling VY, Straube J, Godfrey W, Haldar R, Janardhanan Y, Cooper L, Bruedigam C, Cooper E, Tavakoli Shirazi P, Jacquelin S, Tey SK, Baell J, Huang F, Jin J, Zhao Y, Bullinger L, Bywater MJ, Lane SW. Targeting cell cycle and apoptosis to overcome chemotherapy resistance in acute myeloid leukemia. Leukemia 2023; 37:143-153. [PMID: 36400926 DOI: 10.1038/s41375-022-01755-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022]
Abstract
Chemotherapy-resistant acute myeloid leukemia (AML), frequently driven by clonal evolution, has a dismal prognosis. A genome-wide CRISPR knockout screen investigating resistance to doxorubicin and cytarabine (Dox/AraC) in human AML cell lines identified gene knockouts involving AraC metabolism and genes that regulate cell cycle arrest (cyclin dependent kinase inhibitor 2A (CDKN2A), checkpoint kinase 2 (CHEK2) and TP53) as contributing to resistance. In human AML cohorts, reduced expression of CDKN2A conferred inferior overall survival and CDKN2A downregulation occurred at relapse in paired diagnosis-relapse samples, validating its clinical relevance. Therapeutically targeting the G1S cell cycle restriction point (with CDK4/6 inhibitor, palbociclib and KAT6A inhibitor, WM-1119, to upregulate CDKN2A) synergized with chemotherapy. Additionally, direct promotion of apoptosis with venetoclax, showed substantial synergy with chemotherapy, overcoming resistance mediated by impaired cell cycle arrest. Altogether, we identify defective cell cycle arrest as a clinically relevant contributor to chemoresistance and identify rationally designed therapeutic combinations that enhance response in AML, potentially circumventing chemoresistance.
Collapse
Affiliation(s)
- Victoria Y Ling
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Jasmin Straube
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - William Godfrey
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rohit Haldar
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Leanne Cooper
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Claudia Bruedigam
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Emily Cooper
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | | | - Siok-Keen Tey
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Jonathan Baell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Fei Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Jianwen Jin
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Yichao Zhao
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Lars Bullinger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Megan J Bywater
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| | - Steven W Lane
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.
| |
Collapse
|
18
|
Tomanová M, Kozlanská K, Jorda R, Jedinák L, Havlíková T, Řezníčková E, Peřina M, Klener P, Dolníková A, Cankař P, Kryštof V. Synthesis and Structural Optimization of 2,7,9-Trisubstituted purin-8-ones as FLT3-ITD Inhibitors. Int J Mol Sci 2022; 23:ijms232416169. [PMID: 36555810 PMCID: PMC9782245 DOI: 10.3390/ijms232416169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Therapy of FLT3-positive acute myeloid leukemia still remains complicated, despite the availability of newly approved kinase inhibitors. Various strategies to avoid the reduced efficacy of therapy have been explored, including the development of dual targeting compounds, which inhibit FLT3 and another kinase necessary for the survival and proliferation of AML cells. We have designed new 2,7,9-trisubstituted 8-oxopurines as FLT3 inhibitors and report here the structure-activity relationship studies. We demonstrated that substituents at positions 7 and 9 modulate activity between CDK4 and FLT3 kinase, and the isopropyl group at position 7 substantially increased the selectivity toward FLT3 kinase, which led to the discovery of compound 15a (9-cyclopentyl-7-isopropyl-2-((4-(piperazin-1-yl)phenyl)amino)-7,9-dihydro-8H-purin-8-one). Cellular analyses in MV4-11 cells revealed inhibition of autophosphorylation of FLT3 kinase in nanomolar doses, including the suppression of downstream STAT5 and ERK1/2 phosphorylation. We also describe mechanistic studies in cell lines and activity in a mouse xenograft model in vivo.
Collapse
Affiliation(s)
- Monika Tomanová
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
| | - Karolína Kozlanská
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech Republic
| | - Radek Jorda
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech Republic
| | - Lukáš Jedinák
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
| | - Tereza Havlíková
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
| | - Eva Řezníčková
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech Republic
| | - Miroslav Peřina
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech Republic
| | - Pavel Klener
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic
- First Department of Internal Medicine-Hematology, General University Hospital and First Faculty of Medicine, Charles University, 12808 Prague, Czech Republic
| | - Alexandra Dolníková
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic
| | - Petr Cankař
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
- Correspondence: (P.C.); (V.K.)
| | - Vladimír Kryštof
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, 77900 Olomouc, Czech Republic
- Correspondence: (P.C.); (V.K.)
| |
Collapse
|
19
|
Molica M, Perrone S. Molecular targets for the treatment of AML in the forthcoming 5th World Health Organization Classification of Haematolymphoid Tumours. Expert Rev Hematol 2022; 15:973-986. [PMID: 36271671 DOI: 10.1080/17474086.2022.2140137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a genetically heterogeneous disease for which the treatment armamentarium has been historically restricted to chemotherapy. However, genomic and epigenomic alterations that contribute to AML initiation, maintenance, and relapse have disclosed new insights to the 5th update in WHO Classification of Haematolymphoid Tumours. AREAS COVERED After four decades of intensive chemotherapy as a 'one-size-fits-all' concept, several targeted agents have been approved for the treatment of AML. Several compounds, directed against regulators of apoptotic, epigenetic, or micro-environmental pathways, and immune-system modulators, are currently in development and investigation in clinical trials. We review advances in target-based therapy for AML focusing on their mechanism of action, examining the intracellular events and pathways, and the results from published clinical trials. EXPERT OPINION To improve patient clinical outcomes, find new biomarkers for therapeutic response, and pinpoint patients who might benefit from novel targeted medicines, next-generation sequencing is being used to evaluate AML-associated mutations. In fact, the new 5th edition of WHO classification has reaffirmed the importance of genetically defined entities that have a prognostic impact, but not all have a specific treatment available. New class of target drugs are in clinical development and could be beneficial to improve the therapeutic armamentarium available.
Collapse
Affiliation(s)
| | - Salvatore Perrone
- Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, Latina, Italy.,Division of Hematology, University Hospital Paolo Giaccone, Palermo, Italy
| |
Collapse
|
20
|
Izuegbuna OO. Polyphenols: Chemoprevention and therapeutic potentials in hematological malignancies. Front Nutr 2022; 9:1008893. [PMID: 36386899 PMCID: PMC9643866 DOI: 10.3389/fnut.2022.1008893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/02/2022] [Indexed: 01/25/2024] Open
Abstract
Polyphenols are one of the largest plant-derived natural product and they play an important role in plants' defense as well as in human health and disease. A number of them are pleiotropic molecules and have been shown to regulate signaling pathways, immune response and cell growth and proliferation which all play a role in cancer development. Hematological malignancies on the other hand, are cancers of the blood. While current therapies are efficacious, they are usually expensive and with unwanted side effects. Thus, the search for newer less toxic agents. Polyphenols have been reported to possess antineoplastic properties which include cell cycle arrest, and apoptosis via multiple mechanisms. They also have immunomodulatory activities where they enhance T cell activation and suppress regulatory T cells. They carry out these actions through such pathways as PI3K/Akt/mTOR and the kynurenine. They can also reverse cancer resistance to chemotherapy agents. In this review, i look at some of the molecular mechanism of action of polyphenols and their potential roles as therapeutic agents in hematological malignancies. Here i discuss their anti-proliferative and anti-neoplastic activities especially their abilities modulate signaling pathways as well as immune response in hematological malignancies. I also looked at clinical studies done mainly in the last 10-15 years on various polyphenol combination and how they enhance synergism. I recommend that further preclinical and clinical studies be carried out to ensure safety and efficacy before polyphenol therapies be officially moved to the clinics.
Collapse
Affiliation(s)
- Ogochukwu O. Izuegbuna
- Department of Haematology, Ladoke Akintola University of Technology (LAUTECH) Teaching Hospital, Ogbomoso, Nigeria
| |
Collapse
|
21
|
Wang A, Fang M, Jiang H, Wang D, Zhang X, Tang B, Zhu X, Hu W, Liu X. Palbociclib promotes the antitumor activity of Venetoclax plus Azacitidine against acute myeloid leukemia. Biomed Pharmacother 2022; 153:113527. [DOI: 10.1016/j.biopha.2022.113527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022] Open
|
22
|
Vorwerk J, Sun K, Frank D, Neumann F, Hüve J, Budde PM, Liu L, Xie X, Patnana PK, Ahmed HMM, Opalka B, Lenz G, Jayavelu AK, Khandanpour C. Presence of the GFI1-36N single nucleotide polymorphism enhances the response of MLL-AF9 leukemic cells to CDK4/6 inhibition. Front Oncol 2022; 12:903691. [PMID: 36003783 PMCID: PMC9393725 DOI: 10.3389/fonc.2022.903691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
The zinc finger protein Growth Factor Independence 1 (GFI1) acts as a transcriptional repressor regulating differentiation of myeloid and lymphoid cells. A single nucleotide polymorphism of GFI1, GFI1-36N, has a prevalence of 7% in healthy Caucasians and 15% in acute myeloid leukemia (AML) patients, hence most probably predisposing to AML. One reason for this is that GFI1-36N differs from the wildtype form GFI1-36S regarding its ability to induce epigenetic changes resulting in a derepression of oncogenes. Using proteomics, immunofluorescence, and immunoblotting we have now gained evidence that murine GFI1-36N leukemic cells exhibit a higher protein level of the pro-proliferative protein arginine N-methyltransferase 5 (PRMT5) as well as increased levels of the cell cycle propagating cyclin-dependent kinases 4 (CDK4) and 6 (CDK6) leading to a faster proliferation of GFI1-36N leukemic cells in vitro. As a therapeutic approach, we subsequently treated leukemic GFI1-36S and GFI1-36N cells with the CDK4/6 inhibitor palbociclib and observed that GFI1-36N leukemic cells were more susceptible to this treatment. The findings suggest that presence of the GFI1-36N variant increases proliferation of leukemic cells and could possibly be a marker for a specific subset of AML patients sensitive to CDK4/6 inhibitors such as palbociclib.
Collapse
Affiliation(s)
- Jan Vorwerk
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Kaiyan Sun
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Daria Frank
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Felix Neumann
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
- Evorion Biotechnologies GmbH, Münster, Germany
| | - Jana Hüve
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Paulina Marie Budde
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Longlong Liu
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Xiaoqing Xie
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Pradeep Kumar Patnana
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Helal Mohammed Mohammed Ahmed
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Bertram Opalka
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Ashok Kumar Jayavelu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Munich, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
- *Correspondence: Cyrus Khandanpour,
| |
Collapse
|
23
|
Qi K, Hu X, Yu X, Cheng H, Wang C, Wang S, Wang Y, Li Y, Cao J, Pan B, Wu Q, Qiao J, Zeng L, Li Z, Xu K, Fu C. Targeting cyclin-dependent kinases 4/6 inhibits survival of megakaryoblasts in acute megakaryoblastic leukaemia. Leuk Res 2022; 120:106920. [PMID: 35872339 DOI: 10.1016/j.leukres.2022.106920] [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: 02/11/2022] [Revised: 06/19/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022]
Abstract
Acute megakaryoblastic leukaemia (AMKL) is characterized by expansion of megakaryoblasts, which are hyper-proliferative cells that fail to undergo differentiation. Insight to the cell-cycle regulation revealed important events in early or late megakaryocytes (MKs) maturation; the cyclin-dependent kinases 4 and 6 (CDK4/6) have been reported to participate in the development of progenitor megakaryocytes, mainly by promoting cell cycle progression and DNA polyploidization. However, it remains unclear whether the continuous proliferation, but not differentiation, of megakaryoblasts is related to an aberrant regulation of CDK4/6 in AMKL. Here, we found that CDK4/6 were up regulated in patients with AMKL, and persistently maintained at a high level during the differentiation of abnormal megakaryocytes in vitro, according to a database and western blot. Additionally, AMKL cells were exceptionally reliant on the cell cycle regulators CDK4 or 6, as blocking their activity using an inhibitor or short hairpin RNA (shRNA) significantly reduced the proliferation of 6133/MPL megakaryocytes, reduced DNA polyploidy, induced apoptosis, decreased the level of phosphorylated retinoblastoma protein (p-Rb), and activation of caspase 3. Additionally, CDK4/6 inhibitors and shRNA reduced the numbers of leukemia cells in the liver and bone marrow (BM), alleviated hepatosplenomegaly, and prolonged the survival of AMKL-transplanted mice. These results suggested that blocking the activity of CDK4/6 may represent an effective approach to control megakaryoblasts in AMKL.
Collapse
Affiliation(s)
- Kunming Qi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Xueting Hu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Xiangru Yu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Hai Cheng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Chunqing Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shujin Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Ying Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Yanjie Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jiang Cao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Bin Pan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Qingyun Wu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.
| | - Chunling Fu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.
| |
Collapse
|
24
|
Harris R, Yang M, Schmidt C, Royet C, Singh S, Natarajan A, Morris M, Frezza C, Laman H. Fbxo7 promotes Cdk6 activity to inhibit PFKP and glycolysis in T cells. J Cell Biol 2022; 221:e202203095. [PMID: 35670764 PMCID: PMC9178409 DOI: 10.1083/jcb.202203095] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 12/24/2022] Open
Abstract
Fbxo7 is associated with cancer and Parkinson's disease. Although Fbxo7 recruits substrates for SCF-type ubiquitin ligases, it also promotes Cdk6 activation in a ligase-independent fashion. We discovered PFKP, the gatekeeper of glycolysis, in a screen for Fbxo7 substrates. PFKP is an essential Cdk6 substrate in some T-ALL cells. We investigated the molecular relationship between Fbxo7, Cdk6, and PFKP, and the effect of Fbxo7 on T cell metabolism, viability, and activation. Fbxo7 promotes Cdk6-independent ubiquitination and Cdk6-dependent phosphorylation of PFKP. Importantly, Fbxo7-deficient cells have reduced Cdk6 activity, and hematopoietic and lymphocytic cells show high expression and significant dependency on Fbxo7. CD4+ T cells with reduced Fbxo7 show increased glycolysis, despite lower cell viability and activation levels. Metabolomic studies of activated CD4+ T cells confirm increased glycolytic flux in Fbxo7-deficient cells, alongside altered nucleotide biosynthesis and arginine metabolism. We show Fbxo7 expression is glucose-responsive at the mRNA and protein level and propose Fbxo7 inhibits PFKP and glycolysis via its activation of Cdk6.
Collapse
Affiliation(s)
- Rebecca Harris
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ming Yang
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Christina Schmidt
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Chloe Royet
- Pôle Chimie Balard Recherche, Institut des Biomolécules Max Mousseron-UMR5247, Montpellier, France
| | - Sarbjit Singh
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Amarnath Natarajan
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - May Morris
- Pôle Chimie Balard Recherche, Institut des Biomolécules Max Mousseron-UMR5247, Montpellier, France
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Heike Laman
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Chen X, Wu Y, Wang X, Xu C, Wang L, Jian J, Wu D, Wu G. CDK6 is upregulated and may be a potential therapeutic target in enzalutamide-resistant castration-resistant prostate cancer. Eur J Med Res 2022; 27:105. [PMID: 35780240 PMCID: PMC9250190 DOI: 10.1186/s40001-022-00730-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/14/2022] [Indexed: 01/20/2023] Open
Abstract
Background Androgen deprivation therapy (ADT) is still the first-line treatment of prostate cancer (PCa). However, after a certain period of therapy, primary PCa inevitably progresses into castration-resistant PCa (CRPC). Enzalutamide (Enz) is an androgen receptor (AR) signal inhibitor which can delay the progression of CRPC and increase survival of patients with metastatic CRPC. However, the mechanisms involved in enzalutamide-resistant (EnzR) CRPC are still controversial. In the study, we used bioinformatic methods to find potential genes that correlated with the occurrence of EnzR CRPC. Methods We collected RNA sequencing data of the EnzR CRPC cell line LNCaP (EnzR LNCaP) from GSE44905, GSE78201, and GSE150807. We found the hub genes from the three datasets. Then we tested the expression of the hub genes in different databases and the potential drugs that can affect the hub genes. Finally, we verified the hub gene expression and drug function. Results From GSE44905, GSE78201 and GSE150807, we found 45 differentially expressed genes (DEGs) between LNCaP and EnzR LNCaP. Ten hub genes were found in the protein–protein interaction (PPI) network. The expression of hub gene and survival analysis were analyzed by different databases. We found that cyclin-dependent kinase 6 (CDK6) was highly expressed in both the EnzR LNCaP cell and PCa patients. Ten potential small molecules could suppress CDK6 expression as per “CLUE COMMAND” findings. Finally, we found the expression of CDK6 increased in both PCa patients’ samples, CRPC and EnzR PCa cell lines. Three potential CDK6 inhibitors, namely apigenin, chrysin and fisetin, can decrease cell proliferation. Conclusions The study proved that the abnormal overexpression of CDK6 may be a reason behind EnzR CRPC occurrence and suppression CDK6 expression may help treat EnzR CRPC. Supplementary Information The online version contains supplementary material available at 10.1186/s40001-022-00730-y.
Collapse
Affiliation(s)
- Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine,Tongji University, 389 Xincun Road, Shanghai, China
| | - Yechen Wu
- Department of Urology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinan Wang
- Department of Urology, Tongji Hospital, School of Medicine,Tongji University, 389 Xincun Road, Shanghai, China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine,Tongji University, 389 Xincun Road, Shanghai, China
| | - Licheng Wang
- Department of Urology, Tongji Hospital, School of Medicine,Tongji University, 389 Xincun Road, Shanghai, China
| | - Jingang Jian
- Suzhou Medical School of Soochow University, Jiangsu, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine,Tongji University, 389 Xincun Road, Shanghai, China.
| | - Gang Wu
- Department of Urology, Tongji Hospital, School of Medicine,Tongji University, 389 Xincun Road, Shanghai, China.
| |
Collapse
|
26
|
Abstract
Cyclin-dependent kinase 4 (CDK4) and CDK6 are critical mediators of cellular transition into S phase and are important for the initiation, growth and survival of many cancer types. Pharmacological inhibitors of CDK4/6 have rapidly become a new standard of care for patients with advanced hormone receptor-positive breast cancer. As expected, CDK4/6 inhibitors arrest sensitive tumour cells in the G1 phase of the cell cycle. However, the effects of CDK4/6 inhibition are far more wide-reaching. New insights into their mechanisms of action have triggered identification of new therapeutic opportunities, including the development of novel combination regimens, expanded application to a broader range of cancers and use as supportive care to ameliorate the toxic effects of other therapies. Exploring these new opportunities in the clinic is an urgent priority, which in many cases has not been adequately addressed. Here, we provide a framework for conceptualizing the activity of CDK4/6 inhibitors in cancer and explain how this framework might shape the future clinical development of these agents. We also discuss the biological underpinnings of CDK4/6 inhibitor resistance, an increasingly common challenge in clinical oncology.
Collapse
Affiliation(s)
- Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| | - Johann S Bergholz
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jean J Zhao
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
27
|
Sun J, Zhu YM, Liu Q, Hu YH, Li C, Jie HH, Xu GH, Xiao RJ, Xing XL, Yu SC, Liang YP. LncRNA ROR modulates myocardial ischemia-reperfusion injury mediated by the miR-185-5p/CDK6 axis. J Transl Med 2022; 102:505-514. [PMID: 35066566 DOI: 10.1038/s41374-021-00722-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023] Open
Abstract
LncRNAs and miRNAs are correlated with the pathogenesis of myocardial ischemia-reperfusion injury (MIRI). Whether lncRNA ROR or miR-185-5p plays a crucial role in MIRI is still unclear. In in-vitro, human cardiac myocytes (HCMs) were treated with hypoxia/reoxygenation (H/R). Wistar rats were used to set up an in-vitro I/R model by means of recanalization after ligation. Evaluation of the myocardial injury marker lactate dehydrogenase (LDH) in HCMs cells was performed. The expression of miR-185-5p and ROR, IL-1β, and IL-18 were detected by qRT-PCR. ELISA was also performed to evaluate the secretion of IL-1β and IL-18. Western blotting was carried out to determine CDK6, NLRP3, GSDMD-N, ASC, and cleaved-caspase1 protein expression. The relationship between miR-185-5p and CDK6 or ROR was confirmed by a dual-luciferase reporter assay. Our findings revealed that H/R treated HCMs showed a significantly decreased miR-185-5p expression and increased expression of CDK6 and ROR. ROR knockdown reduced H/R induced pyroptosis and inflammation, while knockdown of miR-185-5p accelerated the effect. Furthermore, miR-185-5p was negatively regulated and absorbed by ROR in HCMs. Overexpression of miR-185-5p reversed the H/R-induced cell pyroptosis and upregulation of LDH, IL-1β, and IL-18. In HCMs, miR-185-5p was also negatively regulated and related to CDK6 expression. Moreover, overexpression of CDK6 significantly inhibited the effects of miR-185-5p mimics on the inflammatory response and pyroptosis of HCMs. Knockdown of ROR alleviated H/R-induced myocardial injury by elevating miR-185-5p and inhibiting CDK6 expression. Taken together, our results show that the ROR/miR-185-5p/CDK6 axis modulates cell pyroptosis induced by H/R and the inflammatory response of HCMs.
Collapse
Affiliation(s)
- Jing Sun
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Yan-Meng Zhu
- Queen Mary School, Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Qin Liu
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Yan-Hui Hu
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Chang Li
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Huan-Huan Jie
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Guo-Hai Xu
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Ren-Jie Xiao
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Xian-Liang Xing
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Shu-Chun Yu
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China
| | - Ying-Ping Liang
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P.R. China.
| |
Collapse
|
28
|
Schmalzbauer BS, Thondanpallil T, Heller G, Schirripa A, Sperl CM, Mayer IM, Knab VM, Nebenfuehr S, Zojer M, Mueller AC, Fontaine F, Klampfl T, Sexl V, Kollmann K. CDK6 Degradation Is Counteracted by p16INK4A and p18INK4C in AML. Cancers (Basel) 2022; 14:cancers14061554. [PMID: 35326705 PMCID: PMC8946512 DOI: 10.3390/cancers14061554] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/17/2022] Open
Abstract
Simple Summary CDK4/6 kinase inhibitors show promising results in various subtypes of AML, which has been primarily assigned to the inhibition of CDK6. To bypass therapeutic resistances and tackle the kinase-dependent, as well as kinase-independent, functions of CDK6, new CDK6 degraders have been developed. Here, we present insights into the mechanistic requirements for the efficacy of a CDK6-specific degrader in AML. We show that the presence and levels of the INK4 proteins p16INK4A and p18INK4C determine the extent of CDK6 degradation. Our study reveals the importance of INK4 protein levels as predictive markers for CDK6-targeted therapy in AML. Abstract Cyclin-dependent kinase 6 (CDK6) represents a novel therapeutic target for the treatment of certain subtypes of acute myeloid leukaemia (AML). CDK4/6 kinase inhibitors have been widely studied in many cancer types and their effects may be limited by primary and secondary resistance mechanisms. CDK4/6 degraders, which eliminate kinase-dependent and kinase-independent effects, have been suggested as an alternative therapeutic option. We show that the efficacy of the CDK6-specific protein degrader BSJ-03-123 varies among AML subtypes and depends on the low expression of the INK4 proteins p16INK4A and p18INK4C. INK4 protein levels are significantly elevated in KMT2A-MLLT3+ cells compared to RUNX1-RUNX1T1+ cells, contributing to the different CDK6 degradation efficacy. We demonstrate that CDK6 complexes containing p16INK4A or p18INK4C are protected from BSJ-mediated degradation and that INK4 levels define the proliferative response to CDK6 degradation. These findings define INK4 proteins as predictive markers for CDK6 degradation-targeted therapies in AML.
Collapse
Affiliation(s)
- Belinda S. Schmalzbauer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Teresemary Thondanpallil
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Gerwin Heller
- Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| | - Alessia Schirripa
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Clio-Melina Sperl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Isabella M. Mayer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Vanessa M. Knab
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - André C. Mueller
- CeMM—Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (A.C.M.); (F.F.)
| | - Frédéric Fontaine
- CeMM—Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (A.C.M.); (F.F.)
| | - Thorsten Klampfl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (B.S.S.); (T.T.); (A.S.); (C.-M.S.); (I.M.M.); (V.M.K.); (S.N.); (M.Z.); (T.K.); (V.S.)
- Correspondence:
| |
Collapse
|
29
|
Li X, Yang T, Hu M, Yang Y, Tang M, Deng D, Liu K, Fu S, Tan Y, Wang H, Chen Y, Zhang C, Guo Y, Peng B, Si W, Yang Z, Chen L. Synthesis and biological evaluation of 6-(pyrimidin-4-yl)-1H-pyrazolo[4,3-b]pyridine derivatives as novel dual FLT3/CDK4 inhibitors. Bioorg Chem 2022; 121:105669. [DOI: 10.1016/j.bioorg.2022.105669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/02/2022]
|
30
|
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4 and CDK6) and their activating partners, D-type cyclins, link the extracellular environment with the core cell cycle machinery. Constitutive activation of cyclin D–CDK4/6 represents the driving force of tumorigenesis in several cancer types. Small-molecule inhibitors of CDK4/6 have been used with great success in the treatment of hormone receptor–positive breast cancers and are in clinical trials for many other tumor types. Unexpectedly, recent work indicates that inhibition of CDK4/6 affects a wide range of cellular functions such as tumor cell metabolism and antitumor immunity. We discuss how recent advances in understanding CDK4/6 biology are opening new avenues for the future use of cyclin D–CDK4/6 inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yan Geng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
31
|
Yuan K, Ji M, Xie S, Qiu Z, Chen W, Min W, Xia F, Zheng M, Wang X, Li J, Hou Y, Kuang W, Wang L, Gu W, Li Z, Yang P. Discovery of Dual CDK6/PIM1 Inhibitors with a Novel Structure, High Potency, and Favorable Druggability for the Treatment of Acute Myeloid Leukemia. J Med Chem 2021; 65:857-875. [PMID: 34958208 DOI: 10.1021/acs.jmedchem.1c02019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nowadays, the simultaneous inhibition of two or more pathways plays an increasingly important role in cancer treatment due to the complex and diverse pathogenesis of cancer, and the combination of the cyclin-dependent kinase 6 (CDK6) inhibitor and PIM1 inhibitor was found to generate synergistic effects in acute myeloid leukemia (AML) treatment. Therefore, we discovered a novel lead 1 targeting CDK6/PIM1 via pharmacophore-based and structure-based virtual screening, synthesized five different series of new derivates, and obtained a potent and balanced dual CDK6/PIM1 inhibitor 51, which showed high kinase selectivity. Meanwhile, 51 displayed an excellent safety profile and great pharmacokinetic properties. Furthermore, 51 displayed stronger potency in reducing the burden of AML than palbociclib and SMI-4a in vivo. In summary, we offered a new direction for AML treatment and provided a great lead compound for AML preclinical studies.
Collapse
Affiliation(s)
- Kai Yuan
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Minghui Ji
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shengnan Xie
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhixia Qiu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Weijiao Chen
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fei Xia
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mingming Zheng
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiaxing Li
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Hou
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenbin Kuang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liping Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing 211198, China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
32
|
Lengefeld J, Cheng CW, Maretich P, Blair M, Hagen H, McReynolds MR, Sullivan E, Majors K, Roberts C, Kang JH, Steiner JD, Miettinen TP, Manalis SR, Antebi A, Morrison SJ, Lees JA, Boyer LA, Yilmaz ÖH, Amon A. Cell size is a determinant of stem cell potential during aging. SCIENCE ADVANCES 2021; 7:eabk0271. [PMID: 34767451 PMCID: PMC8589318 DOI: 10.1126/sciadv.abk0271] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/24/2021] [Indexed: 05/05/2023]
Abstract
Stem cells are remarkably small. Whether small size is important for stem cell function is unknown. We find that hematopoietic stem cells (HSCs) enlarge under conditions known to decrease stem cell function. This decreased fitness of large HSCs is due to reduced proliferation and was accompanied by altered metabolism. Preventing HSC enlargement or reducing large HSCs in size averts the loss of stem cell potential under conditions causing stem cell exhaustion. Last, we show that murine and human HSCs enlarge during aging. Preventing this age-dependent enlargement improves HSC function. We conclude that small cell size is important for stem cell function in vivo and propose that stem cell enlargement contributes to their functional decline during aging.
Collapse
Affiliation(s)
- Jette Lengefeld
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Chia-Wei Cheng
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pema Maretich
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marguerite Blair
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hannah Hagen
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Melanie R. McReynolds
- Department of Chemistry, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton, NJ, USA
| | - Emily Sullivan
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kyra Majors
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina Roberts
- Max Planck Institute for Biology of Ageing and CECAD, University of Cologne, Cologne, Germany
| | - Joon Ho Kang
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joachim D. Steiner
- Max Planck Institute for Biology of Ageing and CECAD, University of Cologne, Cologne, Germany
- Department II of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Teemu P. Miettinen
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Scott R. Manalis
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing and CECAD, University of Cologne, Cologne, Germany
| | - Sean J. Morrison
- Children’s Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jacqueline A. Lees
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laurie A. Boyer
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ömer H. Yilmaz
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Angelika Amon
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
33
|
Izuegbuna O, Otunola GA, Bradley G. GC-MS Profiling and Antineoplastic Activity of Pelargonium Inquinans Ait Leaves on Acute Leukaemia Cell Lines U937 and Jurkat. Nutr Cancer 2021; 74:1849-1871. [PMID: 34477039 DOI: 10.1080/01635581.2021.1969417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We investigated the antineoplastic activities of extracts of Pelargonium inquinans leaves, a plant native to South Africa on acute leukemia cell lines, U937 and Jurkat and the inflammatory effect (nitric oxide and cyclo-oxygenase-2) on RAW 264.7 cells. The extracts of Pelargonium inquinans have significant cytotoxicity especially on U937 cells and pro-inflammatory release of nitric oxide on RAW 264.7 macrophages. The GC-MS study of the essential oil showed it had more than a hundred compounds. This study showed that Pelargonium inquinans have antineoplastic and anti-inflammatory activities which can be further explored in In Vivo studies.
Collapse
Affiliation(s)
- Ogochukwu Izuegbuna
- Department of Biochemistry, Faculty of Science & Agriculture, University of Fort Hare, Alice, South Africa
| | - Gloria A Otunola
- Medicinal Plant and Economic Development (MPED), Department of Botany, University of Fort Hare, Alice, South Africa
| | - Graeme Bradley
- Department of Biochemistry, Faculty of Science & Agriculture, University of Fort Hare, Alice, South Africa
| |
Collapse
|
34
|
Esa E, Hashim AK, Mohamed EHM, Zakaria Z, Abu Hassan AN, Mat Yusoff Y, Kamaluddin NR, Abdul Rahman AZ, Chang KM, Mohamed R, Subbiah I, Jamian E, Ho CSL, Lim SM, Lau PC, Pung YF, Zain SM. Construction of a microRNA-mRNA Regulatory Network in De Novo Cytogenetically Normal Acute Myeloid Leukemia Patients. Genet Test Mol Biomarkers 2021; 25:199-210. [PMID: 33734890 DOI: 10.1089/gtmb.2020.0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The association between dysregulated microRNAs (miRNAs) and acute myeloid leukemia (AML) is well known. However, our understanding of the regulatory role of miRNAs in the cytogenetically normal AML (CN-AML) subtype pathway is still poor. The current study integrated miRNA and mRNA profiles to explore novel miRNA-mRNA interactions that affect the regulatory patterns of de novo CN-AML. Methods: We utilized a multiplexed nanoString nCounter platform to profile both miRNAs and mRNAs using similar sets of patient samples (n = 24). Correlations were assessed, and an miRNA-mRNA network was constructed. The underlying biological functions of the mRNAs were predicted by gene enrichment. Finally, the interacting pairs were assessed using TargetScan and microT-CDS. We identified 637 significant negative correlations (false discovery rate <0.05). Results: Network analysis revealed a cluster of 12 miRNAs representing the majority of mRNA targets. Within the cluster, five miRNAs (miR-495-3p, miR-185-5p, let-7i-5p, miR-409-3p, and miR-127-3p) were posited to play a pivotal role in the regulation of CN-AML, as they are associated with the negative regulation of myeloid leukocyte differentiation, negative regulation of myeloid cell differentiation, and positive regulation of hematopoiesis. Conclusion: Three novel interactions in CN-AML were predicted as let-7i-5p:HOXA9, miR-495-3p:PIK3R1, and miR-495-3p:CDK6 may be responsible for regulating myeloid cell differentiation in CN-AML.
Collapse
Affiliation(s)
- Ezalia Esa
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, Kuala Lumpur, Malaysia
| | | | | | - Zubaidah Zakaria
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Alifah Nadia Abu Hassan
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Yuslina Mat Yusoff
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Nor Rizan Kamaluddin
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Ahmad Zuhairi Abdul Rahman
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Kian-Meng Chang
- Hospital Ampang, Jalan Mewah Utara, Pandan Mewah, Ampang, Malaysia
| | - Rashidah Mohamed
- Hospital Ampang, Jalan Mewah Utara, Pandan Mewah, Ampang, Malaysia
| | - Indhira Subbiah
- Hospital Sultanah Aminah, Bangunan Induk, Jalan Persiaran Abu Bakar Sultan, Johor Bahru, Malaysia
| | - Ehram Jamian
- Hospital Sultanah Aminah, Bangunan Induk, Jalan Persiaran Abu Bakar Sultan, Johor Bahru, Malaysia
| | - Caroline Siew-Ling Ho
- Hospital Sultanah Aminah, Bangunan Induk, Jalan Persiaran Abu Bakar Sultan, Johor Bahru, Malaysia
| | - Soo-Min Lim
- Hospital Sultanah Aminah, Bangunan Induk, Jalan Persiaran Abu Bakar Sultan, Johor Bahru, Malaysia
| | - Peng-Choon Lau
- Department of Surgery, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Yuh-Fen Pung
- Department of Biomedical Science, University of Nottingham, Semenyih, Malaysia
| | - Shamsul Mohd Zain
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
35
|
Spirin P, Shyrokova E, Lebedev T, Vagapova E, Smirnova P, Kantemirov A, Dyshlovoy SA, von Amsberg G, Zhidkov M, Prassolov V. Cytotoxic Marine Alkaloid 3,10-Dibromofascaplysin Induces Apoptosis and Synergizes with Cytarabine Resulting in Leukemia Cell Death. Mar Drugs 2021; 19:md19090489. [PMID: 34564151 PMCID: PMC8468638 DOI: 10.3390/md19090489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 01/24/2023] Open
Abstract
Myeloid leukemia is a hematologic neoplasia characterized by a clonal proliferation of hematopoietic stem cell progenitors. Patient prognosis varies depending on the subtype of leukemia as well as eligibility for intensive treatment regimens and allogeneic stem cell transplantation. Although significant progress has been made in the therapy of patients including novel targeted treatment approaches, there is still an urgent need to optimize treatment outcome. The most common therapy is based on the use of chemotherapeutics cytarabine and anthrayclines. Here, we studied the effect of the recently synthesized marine alkaloid 3,10-dibromofascaplysin (DBF) in myeloid leukemia cells. Unsubstituted fascaplysin was early found to affect cell cycle via inhibiting CDK4/6, thus we compared the activity of DBF and other brominated derivatives with known CDK4/6 inhibitor palbociclib, which was earlier shown to be a promising candidate to treat leukemia. Unexpectedly, the effect DBF on cell cycle differs from palbociclib. In fact, DBF induced leukemic cells apoptosis and decreased the expression of genes responsible for cancer cell survival. Simultaneously, DBF was found to activate the E2F1 transcription factor. Using bioinformatical approaches we evaluated the possible molecular mechanisms, which may be associated with DBF-induced activation of E2F1. Finally, we found that DBF synergistically increase the cytotoxic effect of cytarabine in different myeloid leukemia cell lines. In conclusion, DBF is a promising drug candidate, which may be used in combinational therapeutics approaches to reduce leukemia cell growth.
Collapse
Affiliation(s)
- Pavel Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (E.S.); (T.L.); (E.V.); (V.P.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
- Correspondence:
| | - Elena Shyrokova
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (E.S.); (T.L.); (E.V.); (V.P.)
- Moscow Institute of Physics and Technology (National Research University), Institutskiy Per. 9, 141701 Dolgoprudny, Russia
| | - Timofey Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (E.S.); (T.L.); (E.V.); (V.P.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Elmira Vagapova
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (E.S.); (T.L.); (E.V.); (V.P.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Polina Smirnova
- School of Natural Sciences, Far Eastern Federal University, FEFU Campus, Ajax Bay 10, Russky Island, 690922 Vladivostok, Russia; (P.S.); (A.K.); (M.Z.)
| | - Alexey Kantemirov
- School of Natural Sciences, Far Eastern Federal University, FEFU Campus, Ajax Bay 10, Russky Island, 690922 Vladivostok, Russia; (P.S.); (A.K.); (M.Z.)
| | - Sergey A. Dyshlovoy
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (S.A.D.); (G.v.A.)
- Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevskogo Str. 17, 690041 Vladivostok, Russia
| | - Gunhild von Amsberg
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (S.A.D.); (G.v.A.)
- Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany
| | - Maxim Zhidkov
- School of Natural Sciences, Far Eastern Federal University, FEFU Campus, Ajax Bay 10, Russky Island, 690922 Vladivostok, Russia; (P.S.); (A.K.); (M.Z.)
| | - Vladimir Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (E.S.); (T.L.); (E.V.); (V.P.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| |
Collapse
|
36
|
Marofi F, Rahman HS, Al-Obaidi ZMJ, Jalil AT, Abdelbasset WK, Suksatan W, Dorofeev AE, Shomali N, Chartrand MS, Pathak Y, Hassanzadeh A, Baradaran B, Ahmadi M, Saeedi H, Tahmasebi S, Jarahian M. Novel CAR T therapy is a ray of hope in the treatment of seriously ill AML patients. Stem Cell Res Ther 2021; 12:465. [PMID: 34412685 PMCID: PMC8377882 DOI: 10.1186/s13287-021-02420-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a serious, life-threatening, and hardly curable hematological malignancy that affects the myeloid cell progenies and challenges patients of all ages but mostly occurs in adults. Although several therapies are available including chemotherapy, allogeneic hematopoietic stem cell transplantation (alloHSCT), and receptor-antagonist drugs, the 5-year survival of patients is quietly disappointing, less than 30%. alloHSCT is the major curative approach for AML with promising results but the treatment has severe adverse effects such as graft-versus-host disease (GVHD). Therefore, as an alternative, more efficient and less harmful immunotherapy-based approaches such as the adoptive transferring T cell therapy are in development for the treatment of AML. As such, chimeric antigen receptor (CAR) T cells are engineered T cells which have been developed in recent years as a breakthrough in cancer therapy. Interestingly, CAR T cells are effective against both solid tumors and hematological cancers such as AML. Gradually, CAR T cell therapy found its way into cancer therapy and was widely used for the treatment of hematologic malignancies with successful results particularly with somewhat better results in hematological cancer in comparison to solid tumors. The AML is generally fatal, therapy-resistant, and sometimes refractory disease with a disappointing low survival rate and weak prognosis. The 5-year survival rate for AML is only about 30%. However, the survival rate seems to be age-dependent. Novel CAR T cell therapy is a light at the end of the tunnel. The CD19 is an important target antigen in AML and lymphoma and the CAR T cells are engineered to target the CD19. In addition, a lot of research goes on the discovery of novel target antigens with therapeutic efficacy and utilizable for generating CAR T cells against various types of cancers. In recent years, many pieces of research on screening and identification of novel AML antigen targets with the goal of generation of effective anti-cancer CAR T cells have led to new therapies with strong cytotoxicity against cancerous cells and impressive clinical outcomes. Also, more recently, an improved version of CAR T cells which were called modified or smartly reprogrammed CAR T cells has been designed with less unwelcome effects, less toxicity against normal cells, more safety, more specificity, longer persistence, and proliferation capability. The purpose of this review is to discuss and explain the most recent advances in CAR T cell-based therapies targeting AML antigens and review the results of preclinical and clinical trials. Moreover, we will criticize the clinical challenges, side effects, and the different strategies for CAR T cell therapy.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Heshu Sulaiman Rahman
- College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq.,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaise, Sulaimaniyah, Iraq
| | - Zaid Mahdi Jaber Al-Obaidi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Alkafeel, Najaf, 54001, Iraq.,Department of Chemistry and Biochemistry, College of Medicine, University of Kerbala, Karbala, 56001, Iraq
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia.,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | | | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Yashwant Pathak
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA.,Department of Pharmaceutics, Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Ali Hassanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy, No. 2, Floor 4 Unit (G401), 69120, Heidelberg, Germany.
| |
Collapse
|
37
|
Cao J, Zhang Y, Mu J, Yang D, Gu X, Zhang J. Exosomal miR-21-5p contributes to ovarian cancer progression by regulating CDK6. Hum Cell 2021; 34:1185-1196. [PMID: 33813728 DOI: 10.1007/s13577-021-00522-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/17/2021] [Indexed: 12/23/2022]
Abstract
Ovarian cancer is a predominant gynecologic malignancy and correlated with high mortality and severe morbidity. Exosomal microRNAs (miRNAs) play crucial roles in various processes during the progression of ovarian cancer, such as cell proliferation, apoptosis, and invasion. However, the function of exosomal miR-21-5p in ovarian cancer is still unknown. Here, we found that miR-21-5p was upregulated in ovarian cancer tissues, plasma exosomes of ovarian cancer patients, and exosomes from ovarian cancer cells. MiR-21-5p was incorporated in the exosomes from the ovarian cancer cells. In addition, 5-ethynyl-2'-deoxyuridine (Edu), a marker of cancer cell proliferation, was enhanced by miR-21-5p mimic while reduced by miR-21-5p inhibitor in ovarian cancer cells. MiR-21-5p mimic could increase, but miR-21-5p inhibitor could decrease the migration and invasion of cancer cells. Ovarian cancer cell apoptosis was induced by miR-21-5p inhibitor. Moreover, miR-21-5p inhibitor could up-regulate the expression of pro-apoptotic cleaved caspase3 and Bax while downregulate the expression of anti-apoptotic Bcl2 in the cells. Exosomal miR-21-5p inhibited the expression of cyclin-dependent kinase 6 (CDK6) by targeting its 3'-untranslated region (3'-UTR) at both the mRNA and protein levels. Tumorigenicity analysis in nude mice revealed that exosomal miR-21-5p could increase tumor volume, size, and weight of ovarian cancer in vivo. Besides, miR-21-5p targeted CDK6 in tumor tissues of nude mice. In conclusion, exosomal miR-21-5p contributes to the progression of ovarian cancer by regulating CDK6. Our findings will provide novel insights into the mechanism of exosomal miR-21-5p in the development of ovarian cancer. Exosomal miR-21-5p may serve as a potential target for the therapy of ovarian cancer.
Collapse
Affiliation(s)
- Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tianfei Lane, Mochou Road, Nanjing City, Jiangsu Province, 210004, People's Republic of China
| | - Yuan Zhang
- Department of Gynecology Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, Anhui Province, 233000, People's Republic of China
| | - Juan Mu
- Department of Nutrition, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, People's Republic of China
| | - Dazhen Yang
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tianfei Lane, Mochou Road, Nanjing City, Jiangsu Province, 210004, People's Republic of China
| | - Xiaoyan Gu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tianfei Lane, Mochou Road, Nanjing City, Jiangsu Province, 210004, People's Republic of China.
| | - Jing Zhang
- Department of Gynecology Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, Anhui Province, 233000, People's Republic of China.
| |
Collapse
|
38
|
Klein K, Witalisz-Siepracka A, Gotthardt D, Agerer B, Locker F, Grausenburger R, Knab VM, Bergthaler A, Sexl V. T Cell-Intrinsic CDK6 Is Dispensable for Anti-Viral and Anti-Tumor Responses In Vivo. Front Immunol 2021; 12:650977. [PMID: 34248938 PMCID: PMC8264666 DOI: 10.3389/fimmu.2021.650977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022] Open
Abstract
The cyclin-dependent kinase 6 (CDK6) regulates the transition through the G1-phase of the cell cycle, but also acts as a transcriptional regulator. As such CDK6 regulates cell survival or cytokine secretion together with STATs, AP-1 or NF-κB. In the hematopoietic system, CDK6 regulates T cell development and promotes leukemia and lymphoma. CDK4/6 kinase inhibitors are FDA approved for treatment of breast cancer patients and have been reported to enhance T cell-mediated anti-tumor immunity. The involvement of CDK6 in T cell functions remains enigmatic. We here investigated the role of CDK6 in CD8+ T cells, using previously generated CDK6 knockout (Cdk6-/-) and kinase-dead mutant CDK6 (Cdk6K43M) knock-in mice. RNA-seq analysis indicated a role of CDK6 in T cell metabolism and interferon (IFN) signaling. To investigate whether these CDK6 functions are T cell-intrinsic, we generated a T cell-specific CDK6 knockout mouse model (Cdk6fl/fl CD4-Cre). T cell-intrinsic loss of CDK6 enhanced mitochondrial respiration in CD8+ T cells, but did not impact on cytotoxicity and production of the effector cytokines IFN-γ and TNF-α by CD8+ T cells in vitro. Loss of CDK6 in peripheral T cells did not affect tumor surveillance of MC38 tumors in vivo. Similarly, while we observed an impaired induction of early responses to type I IFN in CDK6-deficient CD8+ T cells, we failed to observe any differences in the response to LCMV infection upon T cell-intrinsic loss of CDK6 in vivo. This apparent contradiction might at least partially be explained by the reduced expression of Socs1, a negative regulator of IFN signaling, in CDK6-deficient CD8+ T cells. Therefore, our data are in line with a dual role of CDK6 in IFN signaling; while CDK6 promotes early IFN responses, it is also involved in the induction of a negative feedback loop. These data assign CDK6 a role in the fine-tuning of cytokine responses.
Collapse
Affiliation(s)
- Klara Klein
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Benedikt Agerer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Felix Locker
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Vanessa Maria Knab
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
39
|
Zhang Y, Yuan L. Fms-like tyrosine kinase 3-internal tandem duplications epigenetically activates checkpoint kinase 1 in acute myeloid leukemia cells. Sci Rep 2021; 11:13236. [PMID: 34168220 PMCID: PMC8225911 DOI: 10.1038/s41598-021-92566-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/04/2021] [Indexed: 01/15/2023] Open
Abstract
It is not clear how Fms-like tyrosine kinase 3-internal tandem duplications (FLT3-ITD) regulates checkpoint kinase 1 (CHK1) in acute myeloid leukemia (AML). In this study, we investigated the regulatory effect of FLT3-ITD on CHK1. Our results showed that CHK1 was highly expressed in FLT3-ITD positive AML. The overall survival rate and disease-free survival rate of AML patients with high CHK1 level were lower than those of patients with low CHK1 level. Mechanistically, FLT3-ITD recruited p300 to the CHK1 promoter and subsequently acetylated H3K27, thereby enhancing the transcription of CHK1. Interfering with the expression of CHK1 significantly inhibited the cell proliferation and induced cell apoptosis in FLT3-ITD positive MV4-11 cells. In addition, CHK1 knockdown promoted the sensitivity of MV4-11 cells to the epigenetic inhibitors JQ1 and C646. This study discovers a new therapeutic target for FLT3-ITD + AML and provided evidence for the combination of epigenetic inhibitors for AML treatment.
Collapse
Affiliation(s)
- Yudong Zhang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Lingli Yuan
- Department of Hematology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
40
|
Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, Yang W, Tian C, Miao Z, Wang T, Yang S. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct Target Ther 2021; 6:201. [PMID: 34054126 PMCID: PMC8165101 DOI: 10.1038/s41392-021-00572-w] [Citation(s) in RCA: 782] [Impact Index Per Article: 195.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Due to the advantages in efficacy and safety compared with traditional chemotherapy drugs, targeted therapeutic drugs have become mainstream cancer treatments. Since the first tyrosine kinase inhibitor imatinib was approved to enter the market by the US Food and Drug Administration (FDA) in 2001, an increasing number of small-molecule targeted drugs have been developed for the treatment of malignancies. By December 2020, 89 small-molecule targeted antitumor drugs have been approved by the US FDA and the National Medical Products Administration (NMPA) of China. Despite great progress, small-molecule targeted anti-cancer drugs still face many challenges, such as a low response rate and drug resistance. To better promote the development of targeted anti-cancer drugs, we conducted a comprehensive review of small-molecule targeted anti-cancer drugs according to the target classification. We present all the approved drugs as well as important drug candidates in clinical trials for each target, discuss the current challenges, and provide insights and perspectives for the research and development of anti-cancer drugs.
Collapse
Affiliation(s)
- Lei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, People's Republic of China
| | - Yueshan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Liang Xiong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wenjing Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ming Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ting Yuan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, People's Republic of China
| | - Wei Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chenyu Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhuang Miao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Tianqi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
41
|
Cyclin-Dependent Kinase Inhibitors in Hematological Malignancies-Current Understanding, (Pre-)Clinical Application and Promising Approaches. Cancers (Basel) 2021; 13:cancers13102497. [PMID: 34065376 PMCID: PMC8161389 DOI: 10.3390/cancers13102497] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Cyclin-dependent kinases are involved in the regulation of cancer-initiating processes like cell cycle progression, transcription, and DNA repair. In hematological neoplasms, these enzymes are often overexpressed, resulting in increased cell proliferation and cancer progression. Early (pre-)clinical data using cyclin-dependent kinase inhibitors are promising but identifying the right drug for each subgroup and patient is challenging. Certain chromosomal abnormalities and signaling molecule activities are considered as potential biomarkers. We therefore summarized relevant studies investigating cyclin-dependent kinase inhibitors in hematological malignancies and further discuss molecular mechanisms of resistance and other open questions. Abstract Genetically altered stem or progenitor cells feature gross chromosomal abnormalities, inducing modified ability of self-renewal and abnormal hematopoiesis. Cyclin-dependent kinases (CDK) regulate cell cycle progression, transcription, DNA repair and are aberrantly expressed in hematopoietic malignancies. Incorporation of CDK inhibitors (CDKIs) into the existing therapeutic regimens therefore constitutes a promising strategy. However, the complex molecular heterogeneity and different clinical presentation is challenging for selecting the right target and defining the ideal combination to mediate long-term disease control. Preclinical and early clinical data suggest that specific CDKIs have activity in selected patients, dependent on the existing rearrangements and mutations, potentially acting as biomarkers. Indeed, CDK6, expressed in hematopoietic cells, is a direct target of MLL fusion proteins often observed in acute leukemia and thus contributes to leukemogenesis. The high frequency of aberrancies in the retinoblastoma pathway additionally warrants application of CDKIs in hematopoietic neoplasms. In this review, we describe the preclinical and clinical advances recently made in the use of CDKIs. These include the FDA-approved CDK4/6 inhibitors, traditional and novel pan-CDKIs, as well as dual kinase inhibitors. We additionally provide an overview on molecular mechanisms of response vs. resistance and discuss open questions.
Collapse
|
42
|
A robust approach for the generation of functional hematopoietic progenitor cell lines to model leukemic transformation. Blood Adv 2021; 5:39-53. [PMID: 33570624 DOI: 10.1182/bloodadvances.2020003022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
Studies of molecular mechanisms of hematopoiesis and leukemogenesis are hampered by the unavailability of progenitor cell lines that accurately mimic the situation in vivo. We now report a robust method to generate and maintain LSK (Lin-, Sca-1+, c-Kit+) cells, which closely resemble MPP1 cells. HPCLSKs reconstitute hematopoiesis in lethally irradiated recipient mice over >8 months. Upon transformation with different oncogenes including BCR/ABL, FLT3-ITD, or MLL-AF9, their leukemic counterparts maintain stem cell properties in vitro and recapitulate leukemia formation in vivo. The method to generate HPCLSKs can be applied to transgenic mice, and we illustrate it for CDK6-deficient animals. Upon BCR/ABLp210 transformation, HPCLSKsCdk6-/- induce disease with a significantly enhanced latency and reduced incidence, showing the importance of CDK6 in leukemia formation. Studies of the CDK6 transcriptome in murine HPCLSK and human BCR/ABL+ cells have verified that certain pathways depend on CDK6 and have uncovered a novel CDK6-dependent signature, suggesting a role for CDK6 in leukemic progenitor cell homing. Loss of CDK6 may thus lead to a defect in homing. The HPCLSK system represents a unique tool for combined in vitro and in vivo studies and enables the production of large quantities of genetically modifiable hematopoietic or leukemic stem/progenitor cells.
Collapse
|
43
|
B Cell Receptor signaling and genetic lesions in TP53 and CDKN2A/CDKN2B cooperate in Richter Transformation. Blood 2021; 138:1053-1066. [PMID: 33900379 DOI: 10.1182/blood.2020008276] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/06/2021] [Indexed: 11/20/2022] Open
Abstract
B cell receptor (BCR) signals play a critical role in the pathogenesis of chronic lymphocytic leukemia (CLL), but their role in regulating CLL cell proliferation has still not been firmly established. Unlike normal B cells, CLL cells do not proliferate in vitro upon engagement of the BCR, suggesting that CLL cell proliferation is regulated by other signals from the microenvironment, such as those provided by Toll-like receptors or T cells. Here, we report that BCR engagement of human and murine CLL cells induces several positive regulators of the cell cycle, but simultaneously induces the negative regulators CDKN1A, CDKN2A and CDKN2B, which block cell cycle progression. We further show that introduction of genetic lesions that downregulate these cell cycle inhibitors, such as inactivating lesions in CDKN2A, CDKN2B and the CDKN1A regulator TP53, leads to more aggressive disease in a murine in vivo CLL model and spontaneous proliferation in vitro that is BCR-dependent but independent of costimulatory signals. Importantly, inactivating lesions in CDKN2A, CDKN2B and TP53 frequently co-occur in Richter syndrome, and BCR stimulation of human Richter syndrome cells with such lesions is sufficient to induce proliferation. We also show that tumor cells with combined TP53 and CDKN2A/2B abnormalities remain sensitive to BCR inhibitor treatment and are synergistically sensitive to the combination of a BCR and CDK4/6 inhibitor both in vitro and in vivo. These data provide evidence that BCR signals are directly involved in driving CLL cell proliferation and reveal a novel mechanism of Richter transformation.
Collapse
|
44
|
Zhou B, Qin Y, Zhou J, Ruan J, Xiong F, Dong J, Huang X, Yu Z, Gao S. Bortezomib suppresses self-renewal and leukemogenesis of leukemia stem cell by NF-ĸB-dependent inhibition of CDK6 in MLL-rearranged myeloid leukemia. J Cell Mol Med 2021; 25:3124-3135. [PMID: 33599085 PMCID: PMC7957264 DOI: 10.1111/jcmm.16377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukaemia (AML) with chromosomal rearrangements involving the H3K4 methyltransferase mixed‐lineage leukaemia (MLL) is an aggressive subtype with low overall survival. Bortezomib (Bort) is first applied in multiple myeloma. However, whether bort possesses anti‐self‐renewal and leukemogenesis of leukaemia stem cell (LSC) in AML with MLL rearrangements is still unclear. Here, we found that bort suppressed cell proliferation and decreased colony formation in human and murine leukaemic blasts. Besides, bort reduced the frequency and function of LSC, inhibited the progression, and extended the overall survival in MLL‐AF9 (MF9) ‐transformed leukaemic mice. Furthermore, bort decreased the percentage of human LSC (CD34+CD38‐) cells and extended the overall survival in AML blasts‐xenografted NOD/SCID‐IL2Rγ (NSG) mice. Mechanistically, cyclin dependent kinase 6 (CDK6) was identified as a bort target by RNA sequencing. Bort reduced the expressions of CDK6 by inhibiting NF ĸB recruitment to the promoter of CDK6, leading to the abolishment of NF ĸB DNA‐binding activity for CDK6 promoter. Overexpression of CDK6 partially rescued bort‐induced anti‐leukemogenesis. Most importantly, bort had little side‐effect against the normal haematological stem and progenitor cell (HSPC) and did not affect CDK6 expression in normal HSPC. In conclusion, our results suggest that bort selectively targets LSC in MLL rearrangements. Bort might be a prospective drug for AML patients bearing MLL rearrangements.
Collapse
Affiliation(s)
- Bin Zhou
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaqian Qin
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingying Zhou
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jichen Ruan
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fang Xiong
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinglai Dong
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xingzhou Huang
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhijie Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenmeng Gao
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
45
|
Huang C, Chen Z, He Y, He Z, Ban Z, Zhu Y, Ding L, Yang C, Jeong JH, Yuan W, Yang L. EphA2 promotes tumorigenicity of cervical cancer by up-regulating CDK6. J Cell Mol Med 2021; 25:2967-2975. [PMID: 33586348 PMCID: PMC7957165 DOI: 10.1111/jcmm.16337] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/22/2020] [Accepted: 01/09/2021] [Indexed: 12/18/2022] Open
Abstract
Erythropoietin‐producing hepatocellular receptor A2 (EphA2) receptor tyrosine kinase plays an important role in tissue organization and homeostasis in normal organs. EphA2 is overexpressed in a variety of types of solid tumours with oncogenic functions. However, the role of EphA2 in cervical cancer (CC) is still needed to be further explored. Here, we examined the role of EphA2 by establishing a stable EphA2 knock‐down CC cell lines or a stable EphA2‐overexpressed CC cells lines. Overexpression of EphA2 increased cell proliferation and migration of CC while EphA2 knock‐down decreased the CC tumorigenicity. In addition, EphA2 knock‐down suppressed CC tumour development in the xenograft mouse model. Inhibition of EphA2 by AWL‐II‐41‐27, EphA2‐specific tyrosine kinase inhibitor, or knock‐down of EphA2 decreased mRNA and protein expression of cyclin‐dependent kinase (CDK) 6 in CC cells, which increased cellular susceptibility to epirubicin (EPI), an anti‐cancer chemotherapy drug. A clinicopathological study of EphA2 was conducted on a cohort of 158 human CC patients. EphA2 protein expression was positively correlated with CDK6 protein expression, invasion depth, lymph node metastasis and clinicopathological stage (P < .05). This study demonstrates the oncogenic activity of EphA2 in vitro and in vivo, which provides insights into the relevant mechanisms that might lead to novel treatments for CC.
Collapse
Affiliation(s)
- Changhao Huang
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - Zihua Chen
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, China.,Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yihong He
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengxi He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenying Ban
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanhang Zhu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Leilei Ding
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Yang
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, China.,Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ji-Hak Jeong
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Korea
| | - Weijie Yuan
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, China.,Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Li Yang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
46
|
Ernst P, Heidel FH. Molecular Mechanisms of Senescence and Implications for the Treatment of Myeloid Malignancies. Cancers (Basel) 2021; 13:612. [PMID: 33557090 PMCID: PMC7913823 DOI: 10.3390/cancers13040612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 01/07/2023] Open
Abstract
Senescence is a cellular state that is involved in aging-associated diseases but may also prohibit the development of pre-cancerous lesions and tumor growth. Senescent cells are actively secreting chemo- and cytokines, and this senescence-associated secretory phenotype (SASP) can contribute to both early anti-tumorigenic and long-term pro-tumorigenic effects. Recently, complex mechanisms of cellular senescence and their influence on cellular processes have been defined in more detail and, therefore, facilitate translational development of targeted therapies. In this review, we aim to discuss major molecular pathways involved in cellular senescence and potential therapeutic strategies, with a specific focus on myeloid malignancies.
Collapse
Affiliation(s)
- Philipp Ernst
- Internal Medicine 2, Hematology and Oncology, Jena University Hospital, 07747 Jena, Germany;
- Research Program “Else Kröner-Forschungskolleg AntiAge“, Jena University Hospital, 07747 Jena, Germany
| | - Florian H. Heidel
- Internal Medicine C, Hematology and Oncology, Stem Cell Transplantation and Palliative Care, Greifswald University Medicine, 17475 Greifswald, Germany
- Leibniz Institute on Aging, Fritz-Lipmann Institute, 07745 Jena, Germany
| |
Collapse
|
47
|
Swart LE, Heidenreich O. The RUNX1/RUNX1T1 network: translating insights into therapeutic options. Exp Hematol 2021; 94:1-10. [PMID: 33217477 PMCID: PMC7854360 DOI: 10.1016/j.exphem.2020.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022]
Abstract
RUNX1/RUNX1T1 is the most common fusion gene found in acute myeloid leukemia. Seminal contributions by many different research groups have revealed a complex regulatory network promoting leukemic self-renewal and propagation. Perturbation of RUNX1/RUNX1T1 levels and its DNA binding affects chromatin accessibility and transcription factor occupation at multiple gene loci associated with changes in gene expression levels. Exploration of this transcriptional program by targeted RNAi screens uncovered a crucial role of RUNX1/RUNX1T1 in cell cycle progression by regulating CCND2. This dependency results in a high vulnerability toward inhibitors of CDK4 and CDK6 and suggests new avenues for therapeutic intervention against acute myeloid leukemia.
Collapse
MESH Headings
- Animals
- Cell Cycle
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/metabolism
- Gene Expression Regulation, Leukemic
- Gene Regulatory Networks
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Protein Interaction Maps
- RUNX1 Translocation Partner 1 Protein/genetics
- RUNX1 Translocation Partner 1 Protein/metabolism
- Transcriptional Activation
Collapse
Affiliation(s)
- Laura E Swart
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Olaf Heidenreich
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
48
|
Liu W, Yi JM, Liu Y, Chen C, Zhang KX, Zhou C, Zhan HE, Zhao L, Morales S, Zhao XL, Zeng H. CDK6 Is a Potential Prognostic Biomarker in Acute Myeloid Leukemia. Front Genet 2021; 11:600227. [PMID: 33597968 PMCID: PMC7882723 DOI: 10.3389/fgene.2020.600227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a threatening hematological malignant disease in which new successful approaches in therapy are needed. Cyclin-dependent kinase 6 (CDK6), a regulatory enzyme of the cell cycle that plays an important role in leukemogenesis and the maintenance of leukemia stem cells (LSC), has the potential to predict the prognosis of AML. By analyzing public databases, we observed that the messenger RNA (mRNA) levels of CDK6 were significantly overexpressed in AML cell lines and non-acute promyelocytic leukemia (non-APL) AML patients when compared to healthy donors. Furthermore, CDK6 expression was significantly reduced in AML patients who achieved complete remission (CR) compared to that at the time of diagnosis in our validated cohort. The expression of CDK6 was tightly correlated with peripheral blood blasts, French-American-British (FAB) subtypes, CCAAT-enhancer-binding protein α (CEBPA) mutation, and chromosomal abnormalities of t(8;21). However, the clinical significance and effects of CDK6 expression on the prognosis of non-APL AML patients remain uncertain. We found that CDK6 expression was inversely correlated with overall survival (OS) among non-APL AML patients using the Kaplan-Meier analysis. CDK6 was also found to be positively associated with genes identified to contribute to the development of leukemia, including CCND2, DNMT3B, SOX4, and IKZF2, as well as being negatively associated with anticancer microRNAs, including miR-187, miR-9, miR-582, miR708, and miR-362. In summary, our study revealed that CDK6 might be a potential diagnostic and prognostic biomarker in non-APL AML patients.
Collapse
Affiliation(s)
- Wei Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Jin-Mou Yi
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Kai-Xuan Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Zhou
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui-En Zhan
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liang Zhao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Stephanie Morales
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Xie-Lan Zhao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
49
|
Kennedy VE, Smith CC. FLT3 Mutations in Acute Myeloid Leukemia: Key Concepts and Emerging Controversies. Front Oncol 2021; 10:612880. [PMID: 33425766 PMCID: PMC7787101 DOI: 10.3389/fonc.2020.612880] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022] Open
Abstract
The FLT3 receptor is overexpressed on the majority of acute myeloid leukemia (AML) blasts. Mutations in FLT3 are the most common genetic alteration in AML, identified in approximately one third of newly diagnosed patients. FLT3 internal tandem duplication mutations (FLT3-ITD) are associated with increased relapse and inferior overall survival. Multiple small molecule inhibitors of FLT3 signaling have been identified, two of which (midostaurin and gilteritinib) are currently approved in the United States, and many more of which are in clinical trials. Despite significant advances, resistance to FLT3 inhibitors through secondary FLT3 mutations, upregulation of parallel pathways, and extracellular signaling remains an ongoing challenge. Novel therapeutic strategies to overcome resistance, including combining FLT3 inhibitors with other antileukemic agents, development of new FLT3 inhibitors, and FLT3-directed immunotherapy are in active clinical development. Multiple questions regarding FLT3-mutated AML remain. In this review, we highlight several of the current most intriguing controversies in the field including the role of FLT3 inhibitors in maintenance therapy, the role of hematopoietic cell transplantation in FLT3-mutated AML, use of FLT3 inhibitors in FLT3 wild-type disease, significance of non-canonical FLT3 mutations, and finally, emerging concerns regarding clonal evolution.
Collapse
Affiliation(s)
- Vanessa E Kennedy
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Catherine C Smith
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
50
|
Kellaway S, Chin PS, Barneh F, Bonifer C, Heidenreich O. t(8;21) Acute Myeloid Leukemia as a Paradigm for the Understanding of Leukemogenesis at the Level of Gene Regulation and Chromatin Programming. Cells 2020; 9:E2681. [PMID: 33322186 PMCID: PMC7763303 DOI: 10.3390/cells9122681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous disease with multiple sub-types which are defined by different somatic mutations that cause blood cell differentiation to go astray. Mutations occur in genes encoding members of the cellular machinery controlling transcription and chromatin structure, including transcription factors, chromatin modifiers, DNA-methyltransferases, but also signaling molecules that activate inducible transcription factors controlling gene expression and cell growth. Mutant cells in AML patients are unable to differentiate and adopt new identities that are shaped by the original driver mutation and by rewiring their gene regulatory networks into regulatory phenotypes with enhanced fitness. One of the best-studied AML-subtypes is the t(8;21) AML which carries a translocation fusing the DNA-binding domain of the hematopoietic master regulator RUNX1 to the ETO gene. The resulting oncoprotein, RUNX1/ETO has been studied for decades, both at the biochemical but also at the systems biology level. It functions as a dominant-negative version of RUNX1 and interferes with multiple cellular processes associated with myeloid differentiation, growth regulation and genome stability. In this review, we summarize our current knowledge of how this protein reprograms normal into malignant cells and how our current knowledge could be harnessed to treat the disease.
Collapse
Affiliation(s)
- Sophie Kellaway
- Institute of Cancer and Genomica Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B152TT, UK; (S.K.); (P.S.C.)
| | - Paulynn S. Chin
- Institute of Cancer and Genomica Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B152TT, UK; (S.K.); (P.S.C.)
| | - Farnaz Barneh
- Princess Máxima Centrum for Pediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, The Netherlands;
| | - Constanze Bonifer
- Institute of Cancer and Genomica Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham B152TT, UK; (S.K.); (P.S.C.)
| | - Olaf Heidenreich
- Princess Máxima Centrum for Pediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, The Netherlands;
| |
Collapse
|