1
|
Liu B, Kang N, Yang F, Zhang W, Yan X, Wang H, Bai S, Cheng CC, Xu J, Hu W, Zhang Y. Discovery of FLT3-ITD Inhibitor Clifutinib: A Novel Biphenylacetylene Urea Derivative in Clinical Trials for the Treatment of Relapsed/Refractory FLT3-ITD + Acute Myeloid Leukemia. J Med Chem 2025; 68:7955-7972. [PMID: 40213868 DOI: 10.1021/acs.jmedchem.4c03023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Internal tandem duplication (ITD) mutations of FLT3 (FLT3-ITD) are a promising target for patients with acute myeloid leukemia (AML), given that they have been identified in the majority of AML patients and are associated with poor prognosis. Here, a series of biphenylacetylene derivatives was developed as selective FLT3-ITD inhibitors. Representative compound 9e exhibited excellent potency against FLT3-ITD kinase, with an IC50 value of 15.1 nM, and potently suppressed the proliferation of MV-4-11 and MOLM-13 AML cells harboring FLT3-ITD, with IC50 values of 1.5 and 1.4 nM, respectively. Moreover, compound 9e displayed favorable drug-like properties and significantly suppressed tumor growth in MV-4-11 (1.5 mg/kg, qd, tumor growth inhibition (TGI) = 193.5%) and MOLM-13 (4.5 mg/kg, qd, TGI = 94%) xenograft tumor models in mice without significant weight loss. Compound 9e (named Clifutinib) is currently being evaluated in a phase III clinical trial (NCT05586074) for the treatment of relapsed/refractory FLT3-ITD-positive AML.
Collapse
Affiliation(s)
- Bing Liu
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - Ning Kang
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - Fang Yang
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - WeiHong Zhang
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - XingGuo Yan
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - Heng Wang
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - Shun Bai
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - Cliff C Cheng
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - Juan Xu
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| | - WenHao Hu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - YingJun Zhang
- State Key Laboratory of Anti-Infective Drug Development (No. 2015DQ780357), Sunshine Lake Pharma Company, Ltd., Dongguan 523871, China
| |
Collapse
|
2
|
Yin F, Yang J, Luo H, Yu T, Lu W, Zhao M, Du H, Wen S, Huang P, Hu Y. Targeting oncogenic activation of FLT3/SREBP/FASN promotes the therapeutic effect of quizartinib involving disruption of mitochondrial phospholipids. Cell Death Dis 2025; 16:327. [PMID: 40263296 PMCID: PMC12015539 DOI: 10.1038/s41419-025-07661-6] [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: 08/12/2024] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
FMS-like tyrosine kinase 3-internal tandem duplication (FLT3/ITD) is a common driver mutation that presents with a high leukemic burden and its impact on metabolic homeostasis remains to be further investigated. Here, we revealed that the oncogenic activation of FLT3/ITD induced upregulation of target genes of sterol regulatory element-binding proteins (SREBPs) in vivo and in acute myeloid leukemia patients. Quizartinib is a second-generation FLT3 inhibitor that selectively inhibits the activating FLT3 mutations. We demonstrated the critical role of SREBP1 degradation in conferring the response of FLT3/ITD cells to quizartinib. Mechanistically, quizartinib facilitated degradation of the precursor form of SREBP1 via the FLT3/AKT/GSK3 axis and reduced protein levels of its target gene fatty acid synthase (FASN). Lipidomics analysis by Liquid Chromatography Mass Spectrometry (LC-MS) demonstrated that inhibition of FLT3 altered global levels of phospholipids including reduction of cardiolipin, leading to subsequent loss of mitochondrial membrane potential. Pharmacological inhibition of SREBP1 or FASN sensitized FLT3/ITD leukemia cells to quizartinib. Quizartinib combined with SREBP inhibitor fatostatin or FASN inhibitor orlistat provided substantial therapeutic benefit over monotherapies in the murine FLT3/ITD leukemia model. Our results indicated the mechanistic link between FLT3/ITD and SREBP degradation and suggested the combination therapy via targeting FLT3/SREBP/FASN axis.
Collapse
MESH Headings
- fms-Like Tyrosine Kinase 3/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- Humans
- Animals
- Benzothiazoles/pharmacology
- Benzothiazoles/therapeutic use
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Mice
- Mitochondria/metabolism
- Mitochondria/drug effects
- Phospholipids/metabolism
- Fatty Acid Synthase, Type I/metabolism
- Fatty Acid Synthase, Type I/genetics
- Sterol Regulatory Element Binding Protein 1/metabolism
- Sterol Regulatory Element Binding Protein 1/genetics
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
Collapse
Affiliation(s)
- Feng Yin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Jing Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Hao Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Tiantian Yu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
- Metabolomics Research Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Mingyue Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
- Metabolomics Research Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Yumin Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Cancer Metabolism and Intervention Research Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
- Metabolomics Research Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Brisco TA, De Kreijger S, Nair VN, Troian-Gautier L, Tambar UK. Photocatalytic Synthesis of Substituted 2-Aryl Morpholines via Diastereoselective Annulation. J Am Chem Soc 2025. [PMID: 40241339 DOI: 10.1021/jacs.5c01832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Morpholines are prevalent in medicinal chemistry due to their favorable pharmacokinetic properties and widespread presence in FDA-approved drugs. Existing methods for morpholine synthesis often require prefunctionalized or protected reagents, limiting their versatility and efficiency. Here, we present a photocatalytic, diastereoselective annulation strategy for the synthesis of morpholines directly from readily available starting materials. This method employs a visible-light-activated photocatalyst, Lewis acid, and Brønsted acid to achieve high yields and stereoselectivity. It also provides access to diverse substitution patterns, including challenging tri- and tetra-substituted morpholines. Mechanistic studies reveal that the reaction proceeds through the formation of a radical cation intermediate, with triflic acid playing critical roles in protonating the substrate, preserving the photocatalyst, and preventing product oxidation. Beyond morpholines, this strategy is extended to piperidines, pyrrolidines, and other privileged nitrogen heterocycles. Our findings provide a modular approach for constructing complex, medicinally valuable scaffolds, advancing both synthetic and medicinal chemistry.
Collapse
Affiliation(s)
- Tiffany A Brisco
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| | - Simon De Kreijger
- Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université Catholique de Louvain (UCLouvain), Place Louis Pasteur 1, bte L4.01.02, 1348 Louvain-la-Neuve, Belgium
| | - Vaishnavi N Nair
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| | - Ludovic Troian-Gautier
- Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université Catholique de Louvain (UCLouvain), Place Louis Pasteur 1, bte L4.01.02, 1348 Louvain-la-Neuve, Belgium
- Wel Research Institute, Avenue Pasteur 6, 1300 Wavre, Belgium
| | - Uttam K Tambar
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| |
Collapse
|
4
|
Zhu Y, Cao M, Tang Y, Liu Y, Wang H, Qi J, Huang C, Yan C, Liu X, Jiang S, Luo Y, Wang S, Zhou B, Xu H, Lu YY, Wang L. Inhibition of PINK1 senses ROS signaling to facilitate neuroblastoma cell pyroptosis. Autophagy 2025:1-20. [PMID: 40160153 DOI: 10.1080/15548627.2025.2487037] [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: 12/27/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
Mitochondria serve as the primary source of intracellular reactive oxygen species (ROS), which play a critical role in orchestrating cell death pathways such as pyroptosis in various types of cancers. PINK1-mediated mitophagy effectively removes damaged mitochondria and reduces detrimental ROS levels, thereby promoting cell survival. However, the regulation of pyroptosis by PINK1 and ROS in neuroblastoma remains unclear. In this study, we demonstrate that inhibition or deficiency of PINK1 sensitizes ROS signaling and promotes pyroptosis in neuroblastoma cells via the BAX-caspase-GSDME signaling pathway. Specifically, inhibition of PINK1 by AC220 or knockout of PINK1 impairs mitophagy and enhances ROS production, leading to oxidation and oligomerization of TOMM20, followed by mitochondrial recruitment and activation of BAX. Activated BAX facilitates the release of CYCS (cytochrome c, somatic) from the mitochondria into the cytosol, activating CASP3 (caspase 3). Subsequently, activated CASP3 cleaves and activates GSDME, inducing pyroptosis. Furthermore, inhibition or deficiency of PINK1 potentiates the anti-tumor effects of the clinical ROS-inducing drug ethacrynic acid (EA) to inhibit neuroblastoma progression in vivo. Therefore, our study provides a promising intervention strategy for neuroblastoma through the induction of pyroptosis.Abbreviation: AC220, quizartinib; ANOVA, analysis of variance; ANXA5, annexin A5; BAX, BCL2 associated X, apoptosis regulator; BAK1, BCL2 antagonist/killer 1; CCCP, carbonyl cyanide m-chlorophenyl hydrazone; COX4/COX IV, cytochrome c oxidase subunit 4; CS, citrate synthase; CSC, cancer stem cell; CYCS, cytochrome c, somatic; DTT, dithiothreitol; DNA, deoxyribonucleic acid; EA, ethacrynic acid; Fer-1, ferroptosis inhibitor ferrostatin-1; FLT3, fms related tyrosine kinase 3; GSDMD, gasdermin D; GSDME, gasdermin E; kDa, kilodalton; LDH, lactate dehydrogenase; MFN1, mitofusin 1; MFN2, mitofusin 2; mito, mitochondria; mito-ROS, mitochondrial ROS; mtKeima, mitochondria-targeted monomeric keima-red; ml, microliter; MT-CO2, mitochondrially encoded cytochrome c oxidase II; NAC, antioxidant N-acetyl-L-cysteine; Nec-1, necroptosis inhibitor necrostatin-1; OMA1, OMA1 zinc metallopeptidase; OMM, outer mitochondrial membrane; PARP, poly(ADP-ribose) polymerase; PBS, phosphate-buffered saline; PI, propidium iodide; PINK1, PTEN induced kinase 1; PRKN/Parkin, parkin RBR E3 ubiquitin protein ligase; Q-VD, Q-VD-OPH; ROS, reactive oxygen species; sg, single guide; sh, short hairpin; STS, staurosporine; TOMM20, translocase of outer mitochondrial membrane 20; TIMM23, translocase of inner mitochondrial membrane 23; μm, micrometer; μM, micromolar.
Collapse
Affiliation(s)
- Yuyuan Zhu
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Min Cao
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Yancheng Tang
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Yifan Liu
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Haiji Wang
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Jiaqi Qi
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Cainian Huang
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Chenghao Yan
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Xu Liu
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Sijia Jiang
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Yufei Luo
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Shaogui Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Zhou
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying-Ying Lu
- Department of Medical Research, Seventh Affiliated Hospital, Sun Yatsen University, Shenzhen, Guangdong, China
| | - Liming Wang
- The Affiliated XiangTan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Ferreira LM, García-García P, García PA, Castro MÁ. A review on quinolines: New green synthetic methods and bioactive potential. Eur J Pharm Sci 2025:107097. [PMID: 40221058 DOI: 10.1016/j.ejps.2025.107097] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Quinolines have been an interest of study for a few decades due to the importance of this system in natural and pharmaceutical products. Since their discovery in the nineteenth century, many medicinal properties have been found for quinoline compounds. Firstly, as an anti-parasitic agent against malaria and then against many other diseases, such as, other parasitic infections, HIV, bacterial infections and cancer. Consequently, many synthetic methods have been developed to afford the quinoline ring. In this review we look back at traditional methods and look forward to the most recent and promising "green" methods for the synthesis of quinolines. Also, we review the newest advances in therapeutic compounds based on the quinoline skeleton for the treatment of parasitic and cancer diseases and the most recent applications of quinoline derivatives in drug delivery systems.
Collapse
Affiliation(s)
- Laura M Ferreira
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Pilar García-García
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain.
| | - Pablo A García
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - María Ángeles Castro
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain.
| |
Collapse
|
6
|
Leigh RS, Kaynak BL, Ruskoaho H, Välimäki MJ. Development and comparison of single FLT3-inhibitors to dual FLT3/TAF1-inhibitors as an anti-leukemic approach. PLoS One 2025; 20:e0320443. [PMID: 40153395 PMCID: PMC11952222 DOI: 10.1371/journal.pone.0320443] [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: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 03/30/2025] Open
Abstract
Acute myeloid leukemia (AML) is characterized by several recurrent mutations that affect disease biology and phenotype, response to therapy and risk of subsequent relapse. Though tyrosine kinase inhibitors have gained regulatory approval for the treatment of AML, it is unclear whether single drugs targeting a specific genomic alteration will be sufficient to eradicate disease. Fortuitously, kinase/bromodomain inhibitors allow targeting of downstream transcriptional effectors of oncogenic pathways, allowing impediment of drug resistance at the transcriptional level. Successful development of combinatorial therapeutic strategies to inhibit both upstream oncogenic pathways and their downstream effectors could thus impede the onset of resistant disease. By using a combination of high-throughput cell-based screening assays and structure-based design, we have developed a novel anti-proliferative 3i-compound scaffold with a diverse range of single and dual FLT3/TAF1(2) activity against AML. Our novel approach to target both FLT3 kinase and TAF1(2) bromodomain efficiently maintained potency against haematological cancers. However, reference compounds and in vitro cell viability and cytotoxicity assays in cancer cell lines demonstrated superior effects of high affinity tyrosine kinase inhibition compared to inhibition of the TAF1 bromodomain. Our results highlight the feasibility of dual tyrosine kinase-bromodomain targeting to overcome disease mechanisms while also revealing the increased efficacy of FLT3-targeted compounds in AML.
Collapse
Affiliation(s)
- Robert S. Leigh
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Bogac L. Kaynak
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Li N, Steiger S, Guo Y, Li M, Wen Z, Huang M, Xie C, Jiang S, Zhang D, Zhao Y, Yu L, Wang X, Zheng Z, Zhao ZJ, Chen Y. Tubular epithelial cell-derived Flt3L is required for type 1 conventional dendritic cell (cDC1) activation and expansion in promoting the recovery in acute kidney injury. J Adv Res 2025:S2090-1232(25)00134-1. [PMID: 40023248 DOI: 10.1016/j.jare.2025.02.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Dendritic cells (DCs) play a crucial role in the recovery following acute kidney injury (AKI). Fms-related tyrosine kinase 3 ligand (Flt3L) is essential for the generation and maintenance of DCs. However, the cellular source of Flt3L in the kidney and its contribution on renal DC function during AKI remain unclear. METHODS An online available dataset and specimens collected from AKI were used to analyze FLT3L expression. Wild type (WT) mice, T cell-deficient (TcraKO), and type 1 conventional DC (cDC1)-deficient (Irf8KO) mice underwent ischemia-reperfusion (IR) injury to induce AKI. These mice were treated with either mouse recombinant Flt3L (rFlt3L) or the Flt3 inhibitor gilteritinib. In vitro, experiments with human and murine bone marrow (BM) cells, HK-2 cell line, Jurkat T cells, the monocyte cell line THP1, CD4+ T cells and cDC1s were conducted to validate the link between Flt3L and DCs. RESULTS Circulating FLT3L levels were significantly elevated in patients with AKI. This correlated with the degree of kidney dysfunction observed in these patients. Flt3L was expressed in and released by tubular epithelial cells, with minimal expression in immune cells. Flt3L primarily promoted the activation and expansion of cDC1s and polarization of CD4+T cells in vitro, an effect that was blocked by dephosphorylation of AKT and ERK signaling with gilteritinib. In vivo, gilteritinib worsened the outcomes after AKI by decreasing kidney cDC1s expansion. Conversely, therapeutic administration of rFlt3L promoted renal cDC1 accumulation and improved kidney function in mice with AKI. However, in Irf8KO mice, rFlt3 failed to improve outcomes. CONCLUSION Flt3L is upregulated in both humans and mice during IRI-induced AKI and is likely produced by tubular epithelial cells. It mainly promotes the expansion and activation of kidney cDC1 cells, thereby reducing the severity of AKI in mice. These findings suggest that Flt3L-dependent, cDC1-targeted immunotherapy could be a promising strategy for treating AKI.
Collapse
Affiliation(s)
- Na Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich 80336, Germany
| | - Yao Guo
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Muzheng Li
- Department of Cardiology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zheqi Wen
- Department of Cardiovascular Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingcheng Huang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Chuyu Xie
- Emergency and Disaster Medical Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Shan Jiang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Dengyang Zhang
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Yuming Zhao
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Liuting Yu
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xiaohua Wang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, OK 73019, USA
| | - Yun Chen
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
8
|
Gorecki L, Reznickova E, Krystof V, Rezacova M, Ceckova M, Korabecny J. Strategies for the treatment of acute myeloid leukemia with FLT3 mutations: a patent review. Expert Opin Ther Pat 2025; 35:137-164. [PMID: 39718422 DOI: 10.1080/13543776.2024.2446224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/09/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Approximately one-third of all AML patients have a mutation in the Fms-like tyrosine kinase 3 (FLT3) gene, which is associated with a poor prognosis in these individuals. The 2017 approval of midostaurin, the first FLT3 inhibitor, spurred extensive development of more potent and selective inhibitors with an improved safety profile. AREAS COVERED This review analyzes patent inventions for the treatment of AML using FLT3 inhibitors, covering developments from the earliest to the most recent, disclosed in 2024. Our search using the global Espacenet database identified numerous compounds with low nanomolar inhibitory concentrations against FLT3-ITD and FLT3-TKD mutants. These compounds have shown promise in preclinical studies. Co-inhibition strategies and combinatorial therapies to overcome resistance and enhance anti-leukemic efficacy are also discussed. EXPERT OPINION Recent patents highlight advances in the field of FLT3 inhibitors with a focus on overcoming resistance, improving selectivity and potency. Future strategies may include third-generation inhibitors such as type III allosteric inhibitors, irreversible inhibitors, or PROTACs. Personalized medicine approaches utilizing genetic profiling to tailor therapies are emphasized. Exploration of novel combination regimens with emerging therapies like CAR T-cell therapy, immune checkpoint inhibitors, and small molecules targeting critical AML pathways is ongoing to further enhance anti-leukemic efficacy.
Collapse
Affiliation(s)
- Lukas Gorecki
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czech Republic
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Reznickova
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Vladimir Krystof
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Martina Rezacova
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Králové, Charles University, Hradec Kralove, Czech Republic
| | - Martina Ceckova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
9
|
Destro L, Crippa V, Gabbia D, Roverso M, Bogialli S, Zardi P, Marzaro G, Mologni L, Zambon A. Discovery of selective, metabolically stable pyrazole-based FLT3 inhibitors for the treatment of acute myeloid leukemia. RSC Med Chem 2025:d4md00956h. [PMID: 39990165 PMCID: PMC11843578 DOI: 10.1039/d4md00956h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/25/2025] [Indexed: 02/25/2025] Open
Abstract
Acute myeloid leukemia (AML) is the most prevalent form of acute leukemia in adults, representing a substantial medical need, as the standard of care has not changed for the past two decades, and the long-term outcome remains dismal for a large fraction of patients. Approximately 30% of AMLs carry activating mutations of the FLT3 kinase. Unfortunately, single-agent FLT3 inhibitor therapy has met limited clinical efficacy, underscoring a strong rationale for the development of more selective and more potent inhibitors. Here we present the design, synthesis and biological evaluation of a series of biphenyl substituted pyrazoyl-ureas, an underexplored scaffold in medicinal chemistry, as novel FLT3 inhibitors with a putative type II binding mode. Optimized compounds show nanomolar activity against isolated FLT3 (230 nM for compound 10q) and on FLT3-driven cell lines (280 nM and 18 nM for compound 10q against MV4.11 and MOLM-14 cells respectively), with no toxicity against control cell lines, limited metabolism in human microsomes and a reliable SAR; furthermore, profiling of compound 10q against a panel of kinases highlights c-Kit as the only other hit. Overall, we show that the series has a narrow selectivity profile and metabolic stability, and the mode of action of the inhibitors through FLT3 is confirmed by strong suppression of FLT3 and STAT5 phosphorylation.
Collapse
Affiliation(s)
- Lorenza Destro
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia Modena Italy
| | - Valentina Crippa
- Department of Medicine and Surgery, University of Milano-Bicocca Monza Italy
| | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo Italy
| | - Marco Roverso
- Department of Chemistry, University of Padova Via Marzolo 1 Italy
| | - Sara Bogialli
- Department of Chemistry, University of Padova Via Marzolo 1 Italy
| | - Paolo Zardi
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia Modena Italy
| | - Giovanni Marzaro
- Department of Diagnostic and Public Health, University of Verona Piazzale Antonio Scuro 10 Verona Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca Monza Italy
| | - Alfonso Zambon
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia Modena Italy
| |
Collapse
|
10
|
Chen YJ, Zhao Y, Yao MY, Wang YF, Ma M, Yu CC, Jiang HL, Wei W, Shen J, Xu XW, Xie CY. Concurrent inhibition of p300/CBP and FLT3 enhances cytotoxicity and overcomes resistance in acute myeloid leukemia. Acta Pharmacol Sin 2025:10.1038/s41401-025-01479-w. [PMID: 39885312 DOI: 10.1038/s41401-025-01479-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/22/2024] [Indexed: 02/01/2025]
Abstract
FMS-like tyrosine kinase-3 (FLT3), a class 3 receptor tyrosine kinase, can be activated by mutations of internal tandem duplication (FLT3-ITD) or point mutations in the tyrosine kinase domain (FLT3-TKD), leading to constitutive activation of downstream signaling cascades, including the JAK/STAT5, PI3K/AKT/mTOR and RAS/MAPK pathways, which promote the progression of leukemic cells. Despite the initial promise of FLT3 inhibitors, the discouraging outcomes in the treatment of FLT3-ITD-positive acute myeloid leukemia (AML) promote the pursuit of more potent and enduring therapeutic approaches. The histone acetyltransferase complex comprising the E1A binding protein P300 and its paralog CREB-binding protein (p300/CBP) is a promising therapeutic target, but the development of effective p300/CBP inhibitors faces challenges due to inherent resistance and low efficacy, often exacerbated by the absence of reliable clinical biomarkers for patient stratification. In this study we investigated the role of p300/CBP in FLT3-ITD AML and evaluated the therapeutic potential of targeting p300/CBP alone or in combination with FLT3 inhibitors. We showed that high expression of p300 was significantly associated with poor prognosis in AML patients and positively correlated with FLT3 expression. We unveiled that the p300/CBP inhibitors A485 or CCS1477 dose-dependently downregulated FLT3 transcription via abrogation of histone acetylation in FLT3-ITD AML cells; in contrast, the FLT3 inhibitor quizartinib reduced the level of H3K27Ac. Concurrent inhibition of p300/CBP and FLT3 enhanced the suppression of FLT3 signaling and H3K27 acetylation, concomitantly reducing the phosphorylation of STAT5, AKT, ERK and the expression of c-Myc, thereby leading to synergistic antileukemic effects both in vitro and in vivo. Moreover, we found that p300/CBP-associated transcripts were highly expressed in quizartinib-resistant AML cells with FLT3-TKD mutation. Targeting p300/CBP with A485 or CCS1477 retained the efficacy of quizartinib, suggesting marked synergy when combined with p300/CBP inhibitors in quizartinib-resistant AML models, as well as primary FLT3-ITD+ AML samples. These results demonstrate a potential therapeutic strategy of combining p300/CBP and FLT3 inhibitors to treat FLT3-ITD and FLT3-TKD AML.
Collapse
Affiliation(s)
- Yu-Jun Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Zhao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | | | - Ya-Fang Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Ming Ma
- Lingang Laboratory, Shanghai, 200031, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | - Hua-Liang Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wu Wei
- Lingang Laboratory, Shanghai, 200031, China
| | - Jie Shen
- Department of Pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xiao-Wei Xu
- Department of Hematology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai General Hospital, Shanghai, 200080, China.
| | - Cheng-Ying Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- Lingang Laboratory, Shanghai, 200031, China.
| |
Collapse
|
11
|
Chu D, Ji C, Zhang Y, Wei C, Zhang X, Zhong Q, Yan H, Wang J. Identification of inhibitors targeting the FLT3-ITD mutation through 4D-QSAR, in vitro, and in silico. Eur J Med Chem 2025; 282:117089. [PMID: 39602994 DOI: 10.1016/j.ejmech.2024.117089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) mutation is a key target for acute myeloid leukemia (AML) treatment. The second-generation inhibitors such as Gilteritinib still present off-target effects and associated side effects. Therefore, identifying novel FLT3-ITD inhibitors has become a promising strategy for AML treatment. In this study, a 4D-QSAR model was developed based on Gilteritinib and its analogues, and it was found that introducing hydrophobic bulky groups at the piperazine or piperidine of Gilteritinib would enhance the binding affinity to FLT3-ITD. So, three series of targeted compounds (A1-A5, B1-B5 and C1-C5) were designed and synthesized. The antiproliferative activity against MOLM-13 cells was evaluated in vitro. Compound A1 (IC50 = 25.65 nM), with a cubane group at the piperazine position; Compounds B2 (IC50 = 63.38 nM) and C2 (IC50 = 54.96 nM), with a norbornene group at the piperidine position, showed the strongest inhibition in their series. Their IC50 values were comparable to that of the positive control Gilteritinib (IC50 = 22.37 nM). FLT3-ITD was confirmed as the degradation target through a kinase inhibition assay, where the IC50 values were 2.12 nM (Compound A1), 1.29 nM (Compound B2), and 3.06 nM (Compound C2), which were comparable to that of Gilteritinib (IC50 = 0.43 nM). Additionally, molecular docking and molecular dynamics (MD) simulations showed that Compounds A1, B2, and C2 had similar binding modes to that of Gilteritinib with more stable affinities. Overall, these results demonstrated that Compounds A1, B2, and C2 were promising inhibitors for targeting AML with FLT3-ITD mutation.
Collapse
Affiliation(s)
- Dongchen Chu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, PR China
| | - CuiCui Ji
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, PR China
| | - Yu Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, PR China
| | - Chaochun Wei
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, PR China
| | - Xiaokun Zhang
- Department of Chemistry, Tsinghua University, Beijing, 100084, PR China.
| | - Qidi Zhong
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, PR China
| | - Hong Yan
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, PR China
| | - Juan Wang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, PR China
| |
Collapse
|
12
|
Rataj J, Gorecki L, Muthna D, Sorf A, Krystof V, Klener P, Ceckova M, Rezacova M, Korabecny J. Targeting FMS-like tyrosine kinase 3 (FLT3) in acute myeloid leukemia: Novel molecular approaches and therapeutic challenges. Biomed Pharmacother 2025; 182:117788. [PMID: 39733588 DOI: 10.1016/j.biopha.2024.117788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024] Open
Abstract
Acute myeloid leukemia (AML), a heterogeneous hematologic malignancy, has generally a poor prognosis despite the recent advancements in diagnostics and treatment. Genetic instability, particularly mutations in the FMS-like tyrosine kinase 3 (FLT3) gene, is associated with severe outcomes. Approximately 30 % of AML patients harbor FLT3 mutations, which have been linked to higher relapse and reduced survival rates. Traditional AML treatments employ cytarabine and anthracyclines drugs. Furthermore, the development of FLT3 inhibitors has significantly improved therapy for FLT3-mutated AML patients. For example, the introduction of midostaurin, the first FLT3 inhibitor, improved patient outcomes. However, resistant AML cell clones continue to pose a challenge to the success of AML treatment. This review discusses FLT3 kinase, mutations, and role in AML pathogenesis. It explores the molecular mechanisms of FLT3 activation, signaling pathways, and the structure and function of the FLT3 receptor. Current and emerging therapeutic approaches are presented, while highlighting the latest FLT3 inhibitors in clinical use, and strategies to overcome drug resistance. Future directions, including personalized therapies and novel drug designs, are examined to provide updated insights into FLT3-targeted treatments. This comprehensive review aims to guide clinicians and researchers in the development of innovative therapies to improve AML patient outcomes.
Collapse
Affiliation(s)
- Jan Rataj
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove 500 05, Czech Republic
| | - Lukas Gorecki
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, Hradec Kralove 500 01, Czech Republic; Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, Hradec Kralove 500 05, Czech Republic
| | - Darina Muthna
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, Hradec Kralove 500 03, Czech Republic
| | - Ales Sorf
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, Hradec Kralove 500 01, Czech Republic; Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, Czech Republic
| | - Vladimir Krystof
- Department of Experimental Biology, Faculty of Science, Palacký University, Slechtitelu 27, Olomouc 779 00, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Albertov 5/128 00, Prague 128 00, Czech Republic; First Department of Medicine, Department of Hematology, Charles University General Hospital, Katerinska 1660/32, Prague 121 08, Czech Republic
| | - Martina Ceckova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove 500 05, Czech Republic.
| | - Martina Rezacova
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, Hradec Kralove 500 03, Czech Republic.
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, Hradec Kralove 500 05, Czech Republic.
| |
Collapse
|
13
|
Schifferstein J, Bernatavicius A, Janssen APA. Docking-Informed Machine Learning for Kinome-wide Affinity Prediction. J Chem Inf Model 2024; 64:9196-9204. [PMID: 39657274 DOI: 10.1021/acs.jcim.4c01260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Kinase inhibitors are an important class of anticancer drugs, with 80 inhibitors clinically approved and >100 in active clinical testing. Most bind competitively in the ATP-binding site, leading to challenges with selectivity for a specific kinase, resulting in risks for toxicity and general off-target effects. Assessing the binding of an inhibitor for the entire kinome is experimentally possible but expensive. A reliable and interpretable computational prediction of kinase selectivity would greatly benefit the inhibitor discovery and optimization process. Here, we use machine learning on docked poses to address this need. To this end, we aggregated all known inhibitor-kinase affinities and generated the complete accompanying 3D interactome by docking all inhibitors to the respective high-quality X-ray structures. We then used this resource to train a neural network as a kinase-specific scoring function, which achieved an overall performance (R2) of 0.63-0.74 on unseen inhibitors across the kinome. The entire pipeline from molecule to 3D-based affinity prediction has been fully automated and wrapped in a freely available package. This has a graphical user interface that is tightly integrated with PyMOL to allow immediate adoption in the medicinal chemistry practice.
Collapse
Affiliation(s)
- Jordy Schifferstein
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2333CC, The Netherlands
- Oncode Institute, Utrecht 3521AL, The Netherlands
| | - Andrius Bernatavicius
- Leiden Institute of Advanced Computer Science, Leiden University, Leiden 2333CC, The Netherlands
| | - Antonius P A Janssen
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2333CC, The Netherlands
- Oncode Institute, Utrecht 3521AL, The Netherlands
| |
Collapse
|
14
|
Luo W, Liu Y, Qin H, Zhao Z, Wang S, He W, Tang S, Peng J. Nitrogen-containing heterocyclic drug products approved by the FDA in 2023: Synthesis and biological activity. Eur J Med Chem 2024; 279:116838. [PMID: 39255645 DOI: 10.1016/j.ejmech.2024.116838] [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: 06/21/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
This article profiles 13 newly approved nitrogen-containing heterocyclic drugs by the U.S. Food and Drug Administration (FDA) in 2023. These drugs target a variety of therapeutic areas including proteinuria in patients with IgA nephropathy, migraine in adults, Rett syndrome, PI3Kδ syndrome, vasomotor symptoms, alopecia areata, acute myeloid leukemia, postpartum depression, myelofibrosis, and various cancer and tumor types. The molecular structures of these approved drugs feature common aromatic heterocyclic compounds such as pyrrole, imidazole, pyrazole, isoxazole, pyridine, and pyrimidine, as well as aliphatic heterocyclic compounds like caprolactam, piperazine, and piperidine. Some compounds also contain multiple heteroatoms like 1,2,4-thiadiazole and 1,2,4-triazole. The article provides a comprehensive overview of the bioactivity spectrum, medicinal chemistry discovery, and synthetic methods for each compound.
Collapse
Affiliation(s)
- Weijiang Luo
- Department of Medicinal Chemistry, School of Pharmacy, Hengyang Medical School, University of South China, China
| | - Yiqi Liu
- Department of Medicinal Chemistry, School of Pharmacy, Hengyang Medical School, University of South China, China
| | - Hui Qin
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zeyan Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Hengyang Medical School, University of South China, China
| | - Suqi Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hengyang Medical School, University of South China, China
| | - Weimin He
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, Hunan, 421001, China.
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, China.
| | - Junmei Peng
- Department of Medicinal Chemistry, School of Pharmacy, Hengyang Medical School, University of South China, China.
| |
Collapse
|
15
|
Yan XY, Kang YY, Zhang ZY, Huang P, Yang C, Naranmandura H. Therapeutic approaches targeting oncogenic proteins in myeloid leukemia: challenges and perspectives. Expert Opin Ther Targets 2024; 28:1131-1148. [PMID: 39679536 DOI: 10.1080/14728222.2024.2443577] [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: 12/30/2023] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION Leukemia is typically categorized into myeloid leukemia and lymphoblastic leukemia based on the origins of leukemic cells. Myeloid leukemia is a group of clonal malignancies characterized by the presence of increased immature myeloid cells in both the bone marrow and peripheral blood. Of note, the aberrant expression of specific proteins or the generation of fusion proteins due to chromosomal abnormalities are well established drivers in various forms of myeloid leukemia. Therefore, these oncoproteins represent promising targets for drug development. AREAS COVERED In this review, we comprehensively discussed the pathogenesis of typical leukemia oncoproteins and the current landscape of small molecule drugs targeting these oncogenic proteins. Additionally, we elucidated novel strategies, including proteolysis-targeting chimeras (PROTACs), hyperthermia, and genomic editing, which specifically degrade oncogenic proteins in myeloid malignancies. EXPERT OPINION Although small molecule drugs have significantly improved the prognosis of oncoprotein-driven myeloid leukemia patients, drug resistance due to the mutations in oncoproteins is still a great challenge in the clinic. New approaches such as PROTACs, hyperthermia, and genomic editing are considered promising approaches for the treatment of oncoprotein-driven leukemia, especially for drug-resistant mutants.
Collapse
Affiliation(s)
- Xing Yi Yan
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Yuan Kang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ze Yan Zhang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Huang
- Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Chang Yang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Naranmandura
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Cortes J. Quizartinib: a potent and selective FLT3 inhibitor for the treatment of patients with FLT3-ITD-positive AML. J Hematol Oncol 2024; 17:111. [PMID: 39538314 PMCID: PMC11558990 DOI: 10.1186/s13045-024-01617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Mutations in FMS-related receptor tyrosine kinase 3 (FLT3) are among the most common alterations in acute myeloid leukemia (AML), present in ≈30% of newly diagnosed AML cases. Internal tandem duplications (ITD) in FLT3 (FLT3-ITD) occur in ≈25% of newly diagnosed AML cases and are associated with unfavorable outcomes. Quizartinib (formerly AC220) is a novel, second-generation, highly potent, and selective type II FLT3 inhibitor. Quizartinib is approved in Japan as monotherapy for the treatment of adult patients with FLT3-ITD-positive relapsed/refractory (R/R) AML. Quizartinib is also approved in the United States, Japan, Europe, and United Kingdom in combination with chemotherapy during induction and consolidation, and as maintenance monotherapy (but, in the United States, not after allogeneic hematopoietic cell transplantation [allo-HCT]), for the treatment of adult patients with newly diagnosed FLT3-ITD-positive AML. In this review, we summarize preclinical studies that established quizartinib as a potent and selective type II FLT3 inhibitor as well as early and pivotal phase 3 clinical studies (QuANTUM-R and QuANTUM-First) that led to the approvals of quizartinib. We also summarize mechanisms of resistance to quizartinib along with its safety profile. Furthermore, we review the ongoing post hoc analyses of the QuANTUM-First data elucidating the impact of allo-HCT, the presence of measurable residual disease, and number and length of ITD on the clinical outcomes of quizartinib. We also describe the impact of quizartinib on patient-reported outcomes. Finally, we highlight some of the ongoing studies that test quizartinib in patients with FLT3-ITD-positive AML, patients with FLT3-ITD-negative AML, in both the first-line and R/R settings, in patients fit or unfit for intensive chemotherapy, including studies for quizartinib-based combination with other compounds such as decitabine and venetoclax. Future research should aim to further optimize the clinical value of quizartinib and explore its use in additional clinical settings, which could be achieved by testing quizartinib with other drugs, better characterization of the mechanisms of resistance, identification of the role of quizartinib as a maintenance therapy after allo-HCT, and investigating quizartinib in patients with FLT3-ITD-negative AML.
Collapse
Affiliation(s)
- Jorge Cortes
- Georgia Cancer Center at Augusta University, 1410 Laney Walker Blvd., CN2222, Augusta, GA, 30912, USA.
| |
Collapse
|
17
|
Xia Y, Chen Q, Liu HN, Chi Y, Zhu Y, Shan LS, Dai B, Wu L, Shi X. Synthetic routes and clinical application of new drugs approved by EMA during 2023. Eur J Med Chem 2024; 277:116762. [PMID: 39151275 DOI: 10.1016/j.ejmech.2024.116762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
In 2023, the European Medicines Agency (EMA) granted approval to 77 new molecular entities (NMEs), consisting of 45 new chemical entities (NCEs) and 32 new biological entities (NBEs). These pharmacological agents encompass a broad spectrum of therapeutic domains, including oncology, cardiology, dermatology, diagnostic medicine, endocrinology, gastroenterology and hepatology, metabolic disorders, and neurology. Among the 77 approved pharmaceuticals, three received accelerated review status, and 17 (22 %) were granted orphan drug designation for the treatment of rare diseases. This review provides an overview of the clinical applications and synthetic routes of 42 newly approved NCEs by the EMA in 2023. The objective is to offer a comprehensive understanding of the synthetic approaches used in the development of these drug molecules, thereby inspiring the creation of novel, efficient, and applicable synthetic methodologies.
Collapse
Affiliation(s)
- Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Nan Liu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Lin Wu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xiaobao Shi
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
18
|
Hesham HM, Dokla EME, Elrazaz EZ, Lasheen DS, Abou El Ella DA. FLT3-PROTACs for combating AML resistance: Analytical overview on chimeric agents developed, challenges, and future perspectives. Eur J Med Chem 2024; 277:116717. [PMID: 39094274 DOI: 10.1016/j.ejmech.2024.116717] [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: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
The urgent and unmet medical demand of acute myeloid leukemia (AML) patients has driven the drug discovery process for expansion of the landscape of AML treatment. Despite the several agents developed for treatment of AML, more than 60 % of treated patients undergo relapse again after re-emission, thus, no complete cure for this complex disease has been reached yet. Targeted oncoprotein degradation is a new paradigm that can be employed to solve drug resistance, disease relapse, and treatment failure in complex diseases as AML, the most lethal hematological malignancy. AML is an aggressive blood cancer form and the most common type of acute leukemia, with bad outcomes and a very poor 5-year survival rate. FLT3 mutations occur in about 30 % of AML cases and FLT3-ITD is associated with poor prognosis of this disease. Prevalent FLT3 mutations include internal tandem duplication and point mutations (e.g., D835) in the tyrosine kinase domain, which induce FLT3 kinase activation and result in survival and proliferation of AML cells again. Currently approved FLT3 inhibitors suffer from limited clinical efficacy due to FLT3 reactivation by mutations, therefore, alternative new treatments are highly needed. Proteolysis-targeting chimera (PROTAC) is a bi-functional molecule that consists of a ligand of the protein of interest, FLT3 inhibitor in our case, that is covalently linked to an E3 ubiquitin ligase ligand. Upon FLT3-specific PROTAC binding to FLT3, the PROTAC can recruit E3 for FLT3 ubiquitination, which is subsequently subjected to proteasome-mediated degradation. In this review we tried to address the question if PROTAC technology has succeeded in tackling the disease relapse and treatment failure of AML. Next, we explored the latest FLT3-targeting PROTACs developed in the past few years such as quizartinib-based PROTACs, dovitinib-based PROTACs, gilteritinib-based PROTACs, and others. Then, we followed with a deep analysis of their advantages regarding potency improvement and overcoming AML drug resistance. Finally, we discussed the challenges facing these chimeric molecules with proposed future solutions to circumvent them.
Collapse
Affiliation(s)
- Heba M Hesham
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Eman M E Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| | - Eman Z Elrazaz
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Deena S Lasheen
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Dalal A Abou El Ella
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| |
Collapse
|
19
|
Schlaweck S, Radcke A, Kampmann S, Becker BV, Brossart P, Heine A. The Immunomodulatory Effect of Different FLT3 Inhibitors on Dendritic Cells. Cancers (Basel) 2024; 16:3719. [PMID: 39518156 PMCID: PMC11545830 DOI: 10.3390/cancers16213719] [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: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND FMS-like tyrosine kinase 3 (FLT3) mutations or internal tandem duplication occur in 30% of acute myeloid leukemia (AML) cases. In these cases, FLT3 inhibitors (FLT3i) are approved for induction treatment and relapse. Allogeneic hematopoietic stem cell transplantation (alloHSCT) remains the recommended post-induction therapy for suitable patients. However, the role of FLT3i therapy after alloHSCT remains unclear. Therefore, we investigated the three currently available FLT3i, gilteritinib, midostaurin, and quizartinib, in terms of their immunosuppressive effect on dendritic cells (DCs). DCs are professional antigen-presenting cells inducing T-cell responses to infectious stimuli. Highly activated DCs can also cause complications after alloHSCT, such as triggering Graft versus Host disease, a serious and potentially life-threatening complication after alloHSCT. METHODS To study the immunomodulatory effects on DCs, we differentiated murine and human DCs in the presence of FLT3i and performed immunophenotyping by flow cytometry and cytokine measurements and investigated gene and protein expression. RESULTS We detected a dose-dependent immunosuppressive effect of midostaurin, which decreased the expression of costimulatory markers like CD86, and found a reduced secretion of pro-inflammatory cytokines such as IL-12, TNFα, and IL-6. Mechanistically, we show that midostaurin inhibits TLR and TNF signaling and NFκB, PI3K, and MAPK pathways. The immunosuppressive effect of gilteritinib was less pronounced, while quizartinib did not show truncation of relevant signaling pathways. CONCLUSIONS Our results suggest different immunosuppressive effects of these three FLT3i and may, therefore, provide an additional rationale for optimal maintenance therapy after alloHSCT of FLT3-positive AML patients to prevent infectious complications and GvHD mediated by DCs.
Collapse
Affiliation(s)
- Sebastian Schlaweck
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
- Faculty of Medicine, Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), University Hospital of Bonn, 53127 Bonn, Germany
| | - Alea Radcke
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| | - Sascha Kampmann
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| | - Benjamin V. Becker
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany;
| | - Peter Brossart
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| | - Annkristin Heine
- Medical Clinic III for Hematology, Oncology, Rheumatology, Immunoncology and Stem-Cell Transplantation, University of Bonn, 53127 Bonn, Germany; (S.S.); (A.R.); (S.K.); (P.B.)
| |
Collapse
|
20
|
Moallem FE, Gholami Chahkand MS, Dadkhah PA, Azarm E, Shahrokhi M, Deyhimi MS, Karimi MA. Quizartinib: a new hope in acute myeloid leukemia, an applied comprehensive review. Future Oncol 2024; 20:2791-2810. [PMID: 39297694 PMCID: PMC11572082 DOI: 10.1080/14796694.2024.2399425] [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: 12/12/2023] [Accepted: 08/29/2024] [Indexed: 11/16/2024] Open
Abstract
Acute myeloid leukemia (AML) is caused by a defective precursor leading to malignant clonal expansion, often with FMS-like tyrosine kinase-3 receptor (FLT3) mutations, particularly internal tandem duplication (ITD), which has a poor prognosis. Quizartinib, a second-generation FLT3 inhibitor, has FDA approval for relapsed/refractory AML with FLT3/ITD mutation. It has shown promise in clinical studies since 2013 due to its excellent oral absorption and potent activity on FLT3. This review explores Quizartinib's mechanism of action, efficacy in monotherapy or combination with chemotherapy, drug interactions, adverse events, resistance mechanisms and future research directions.
Collapse
Affiliation(s)
| | | | | | - Eftekhar Azarm
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Khamari R, Degand C, Fovez Q, Trinh A, Chomy A, Laine W, Dekiouk S, Ghesquiere B, Quesnel B, Marchetti P, Manier S, Kluza J. Key role of glutamine metabolism in persistence of leukemic cells upon exposition to FLT3 tyrosine kinase inhibitors. Exp Hematol 2024; 137:104253. [PMID: 38879112 DOI: 10.1016/j.exphem.2024.104253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 07/02/2024]
Abstract
Acute myeloid leukemias are a group of hematological malignancies characterized by a poor prognosis for survival. The discovery of oncogenic mutations in the FMS-like tyrosine kinase 3 (FLT3) gene has led to the development of tyrosine kinase inhibitors such as quizartinib. However, achieving complete remission in patients remains challenging because these new tyrosine kinase inhibitors (TKIs) are unable to completely eradicate all leukemic cells. Residual leukemic cells persist during quizartinib treatment, leading to the rapid emergence of drug-resistant leukemia. Given that mitochondrial oxidative metabolism promotes the survival of leukemic cells after exposure to multiple anticancer drugs, we characterized the metabolism of leukemic cells that persisted during quizartinib treatment and developed metabolic strategies to eradicate them. In our study, employing biochemical and metabolomics approaches, we confirmed that the survival of leukemic cells treated with FLT3 inhibitors critically depends on maintaining mitochondrial metabolism, specifically through glutamine oxidation. We uncovered a synergistic interaction between the FLT3 inhibitor quizartinib and L-asparaginase, operating through antimetabolic mechanisms. Utilizing various models of persistent leukemia, we demonstrated that leukemic cells resistant to quizartinib are susceptible to L-asparaginase. This combined therapeutic strategy shows promise in reducing the development of resistance to FLT3 inhibitors, offering a potential strategy to enhance treatment outcomes.
Collapse
MESH Headings
- fms-Like Tyrosine Kinase 3/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- Humans
- Glutamine/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Mitochondria/metabolism
- Mitochondria/drug effects
- Benzothiazoles/pharmacology
- Cell Line, Tumor
- Animals
- Mice
- Tyrosine Kinase Inhibitors
Collapse
Affiliation(s)
- Raeeka Khamari
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Claire Degand
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Quentin Fovez
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Anne Trinh
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Axel Chomy
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - William Laine
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Salim Dekiouk
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; Centre de Bio-Pathologie, Banque de Tissus, CHU of Lille, Lille, France
| | - Bart Ghesquiere
- Department of Oncology, Metabolomics Core Facility, Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; Hematology Department, CHU of Lille, Lille, France
| | - Philippe Marchetti
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; Centre de Bio-Pathologie, Banque de Tissus, CHU of Lille, Lille, France
| | - Salomon Manier
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; Hematology Department, CHU of Lille, Lille, France
| | - J Kluza
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France.
| |
Collapse
|
22
|
Zhao Y, Tian Y, Pang X, Li G, Shi S, Yan A. Classification of FLT3 inhibitors and SAR analysis by machine learning methods. Mol Divers 2024; 28:1995-2011. [PMID: 37142889 DOI: 10.1007/s11030-023-10640-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/17/2023] [Indexed: 05/06/2023]
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a type III receptor tyrosine kinase, which is an important target for anti-cancer therapy. In this work, we conducted a structure-activity relationship (SAR) study on 3867 FLT3 inhibitors we collected. MACCS fingerprints, ECFP4 fingerprints, and TT fingerprints were used to represent the inhibitors in the dataset. A total of 36 classification models were built based on support vector machine (SVM), random forest (RF), eXtreme Gradient Boosting (XGBoost), and deep neural networks (DNN) algorithms. Model 3D_3 built by deep neural networks (DNN) and TT fingerprints performed best on the test set with the highest prediction accuracy of 85.83% and Matthews correlation coefficient (MCC) of 0.72 and also performed well on the external test set. In addition, we clustered 3867 inhibitors into 11 subsets by the K-Means algorithm to figure out the structural characteristics of the reported FLT3 inhibitors. Finally, we analyzed the SAR of FLT3 inhibitors by RF algorithm based on ECFP4 fingerprints. The results showed that 2-aminopyrimidine, 1-ethylpiperidine,2,4-bis(methylamino)pyrimidine, amino-aromatic heterocycle, [(2E)-but-2-enyl]dimethylamine, but-2-enyl, and alkynyl were typical fragments among highly active inhibitors. Besides, three scaffolds in Subset_A (Subset 4), Subset_B, and Subset_C showed a significant relationship to inhibition activity targeting FLT3.
Collapse
Affiliation(s)
- Yunyang Zhao
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, 15 BeiSanHuan East Road, P.O. Box 53, Beijing, 100029, People's Republic of China
| | - Yujia Tian
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, 15 BeiSanHuan East Road, P.O. Box 53, Beijing, 100029, People's Republic of China
| | - Xiaoyang Pang
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, 15 BeiSanHuan East Road, P.O. Box 53, Beijing, 100029, People's Republic of China
| | - Guo Li
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, 15 BeiSanHuan East Road, P.O. Box 53, Beijing, 100029, People's Republic of China
| | - Shenghui Shi
- College of Information Science and Technology, Beijing University of Chemical Technology, 15 BeiSanHuan East Road, Beijing, 100029, People's Republic of China.
| | - Aixia Yan
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, 15 BeiSanHuan East Road, P.O. Box 53, Beijing, 100029, People's Republic of China.
| |
Collapse
|
23
|
Majirská M, Pilátová MB, Kudličková Z, Vojtek M, Diniz C. Targeting hematological malignancies with isoxazole derivatives. Drug Discov Today 2024; 29:104059. [PMID: 38871112 DOI: 10.1016/j.drudis.2024.104059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/18/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Compounds with a heterocyclic isoxazole ring are well known for their diverse biologic activities encompassing antimicrobial, antipsychotic, immunosuppressive, antidiabetic and anticancer effects. Recent studies on hematological malignancies have also shown that some of the isoxazole-derived compounds feature encouraging cancer selectivity, low toxicity to normal cells and ability to overcome cancer drug resistance of conventional treatments. These characteristics are particularly promising because patients with hematological malignancies face poor clinical outcomes caused by cancer drug resistance or relapse of the disease. This review summarizes the knowledge on isoxazole-derived compounds toward hematological malignancies and provides clues on their mechanism(s) of action (apoptosis, cell cycle arrest, ROS production) and putative pharmacological targets (c-Myc, BET, ATR, FLT3, HSP90, CARM1, tubulin, PD-1/PD-L1, HDACs) wherever known.
Collapse
Affiliation(s)
- Monika Majirská
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovakia
| | - Martina Bago Pilátová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovakia.
| | - Zuzana Kudličková
- NMR Laboratory, Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Slovakia
| | - Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Marshall CM, Federice JG, Bell CN, Cox PB, Njardarson JT. An Update on the Nitrogen Heterocycle Compositions and Properties of U.S. FDA-Approved Pharmaceuticals (2013-2023). J Med Chem 2024; 67:11622-11655. [PMID: 38995264 DOI: 10.1021/acs.jmedchem.4c01122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
This Perspective is a continuation of our analysis of U.S. FDA-approved small-molecule drugs (1938-2012) containing nitrogen heterocycles. In this study we report drug structure and property analyses of 321 unique new small-molecule drugs approved from January 2013 to December 2023 as well as information about frequency of important heteroatoms such as sulfur and fluorine and key small nitrogen substituents (CN and NO2). The most notable change is an incredible increase in drugs containing at least one nitrogen heterocycle─82%, compared to 59% from preceding decades─as well as a significant increase in the number of nitrogen heterocycles per drug. Pyridine has claimed the #1 high-frequency nitrogen heterocycle occurrence spot from piperidine (#2), with pyrimidine (#5), pyrazole (#6), and morpholine (#9) being the big top 10 climbers. Also notable is high number of fused nitrogen heterocycles, apparently driven largely by newly approved cancer drugs.
Collapse
Affiliation(s)
- Christopher M Marshall
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - John G Federice
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Chloe N Bell
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Philip B Cox
- Discovery Research, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jon T Njardarson
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
25
|
Hu C, Zhang Y, Yang J, Xu Y, Deng T, Li Y, Xu S, Wang S, Wang P. Ningetinib, a novel FLT3 inhibitor, overcomes secondary drug resistance in acute myeloid leukemia. Cell Commun Signal 2024; 22:355. [PMID: 38978049 PMCID: PMC11229190 DOI: 10.1186/s12964-024-01729-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) is a common mutation type in acute myeloid leukemia (AML) and is usually associated with poor patient prognosis. With advancements in molecular diagnostics and the development of tyrosine kinase inhibitors (TKI), the overall survival (OS) of AML patients with FLT3-ITD mutations has been prolonged to some extent, but relapse and drug resistance are still substantial challenges. Ningetinib is a novel TKI against various kinases in relation to tumour pathogenesis and is undergoing clinical trials of lung cancer. In this study, we explored the antitumor activity of ningetinib against AML with FLT3 mutations both in vivo and in vitro. METHODS Cell proliferation assays were performed in AML cell lines and Ba/F3 cells expressing various FLT3 mutations to validate the antileukemic activity of ningetinib in vitro. Immunoblot assays were used to verify the effect of ningetinib on the FLT3 protein and downstream pathways. Molecular docking and CETSA were used to validate the interaction of ningetinib with target proteins. The survival benefit of ningetinib in vivo was assessed in Ba/F3-FLT3-ITD-, MOLM13, Ba/F3-FLT3-ITD-F691L-, MOLM13-FLT3-ITD-F691L-induced leukemia mouse models. We also used patient-derived primary cells to determine the efficacy of ningetinib. RESULTS Ningetinib inhibited cell proliferation, blocked the cell cycle, induced apoptosis and bound FLT3 to inhibit its downstream signaling pathways, including the STAT5, AKT and ERK pathways, in FLT3-ITD AML cell lines. In the mouse models with FLT3-ITD and FLT3-ITD-F691L mutation, ningetinib showed superior anti-leukemia activity to existing clinical drugs gilteritinib and quizartinib, significantly prolongating the survival of mice. In addition, ningetinib exhibited activity against patient-derived primary cells harboring FLT3-ITD mutations. CONCLUSION Overall, our study confirmed the therapeutic role of ningetinib in AML with FLT3-ITD mutations, providing a potential new option for clinically resistant patients.
Collapse
Affiliation(s)
- Chuhong Hu
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China
| | - Yvyin Zhang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Jie Yang
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China
| | - Yanli Xu
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China
| | - Tingfen Deng
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China
| | - Yumiao Li
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China
| | - Shilin Xu
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China
| | - Shunqing Wang
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China.
| | - Peihong Wang
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, 510180, Guangdong, China.
| |
Collapse
|
26
|
Jen WY, Kantarjian H, Kadia TM, DiNardo CD, Issa GC, Short NJ, Yilmaz M, Borthakur G, Ravandi F, Daver NG. Combination therapy with novel agents for acute myeloid leukaemia: Insights into treatment of a heterogenous disease. Br J Haematol 2024; 205:30-47. [PMID: 38724457 DOI: 10.1111/bjh.19519] [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: 01/22/2024] [Accepted: 04/27/2024] [Indexed: 07/13/2024]
Abstract
The treatment landscape of acute myeloid leukaemia (AML) is evolving rapidly. Venetoclax in combination with intensive chemotherapy or doublets or triplets with targeted or immune therapies is the focus of numerous ongoing trials. The development of mutation-targeted therapies has greatly enhanced the treatment armamentarium, with FLT3 inhibitors and isocitrate dehydrogenase inhibitors improving outcomes in frontline and relapsed/refractory (RR) AML, and menin inhibitors showing efficacy in RR NPM1mut and KMT2A-rearranged AML. With so many new drugs approved, the number of potential combinatorial approaches to leverage the maximal benefit of these agents has increased dramatically, while at the same time introducing clinical challenges, such as key preclinical and clinical data supporting the development of combinatorial therapy, how to optimally combine or sequence these novel agents, how to optimise dose and duration to maintain safety while enhancing efficacy, the optimal duration of therapy and the role of measurable residual disease in decision-making in both intensive and low-intensity therapy settings. In this review, we will outline the evidence leading to the approval of key agents in AML, their on-label current approvals and how they may be optimally combined in a safe and deliverable fashion to further improve outcomes in AML.
Collapse
Affiliation(s)
- Wei-Ying Jen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
27
|
Wang Z, An Y, Wang J, Lu J. Deciphering the mechanism of HM43239 inhibiting the mutant F691L resistant to gilteritinib in FMS-like tyrosine kinase 3. J Biomol Struct Dyn 2024; 42:5817-5826. [PMID: 37382586 DOI: 10.1080/07391102.2023.2229447] [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: 05/18/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
FMS-like tyrosine kinase (FLT3) has become the legitimate molecular therapeutic target for acute myeloid leukemia therapy. Though FLT3 inhibitors have impact on disease progression, drug resistance induced by secondary point mutations is the primary mechanism and urgent to overcome. Herein, we sought to decipher the mechanism of HM43239 inhibiting the mutant F691L resistant to gilteritinib in FLT3. A series of molecular modeling studies, including molecular dynamics (MD) simulation, dynamic cross-correlation (DCC) analysis, binding free energy (MM-GBSA) and docking study were explored to elucidate the differential tolerance mechanisms of two inhibitors to the same mutant. The F691L mutation had relatively larger effect on gilteritinib than HM43239, which showed as the changed and fixed conformation, respectively. These observations rationalized that the binding affinity of gilteritinib decreased more than that of HM43239 in the F691L mutant.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zhiwei Wang
- College of Pharmacy, Jinzhou Medical University, Linghe District, Jinzhou, China
| | - Yu An
- Department of Open Education, Jinzhou Open University, Linghe District, Jinzhou, China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenhe District, Shenyang, China
| | - Jinghua Lu
- College of Pharmacy, Jinzhou Medical University, Linghe District, Jinzhou, China
| |
Collapse
|
28
|
Wang X, DeFilippis RA, Yan W, Shah NP, Li HY. Overcoming Secondary Mutations of Type II Kinase Inhibitors. J Med Chem 2024; 67:9776-9788. [PMID: 38837951 PMCID: PMC11586107 DOI: 10.1021/acs.jmedchem.3c01629] [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] [Indexed: 06/07/2024]
Abstract
Type II kinase inhibitors bind in the "DFG-out" kinase conformation and are generally considered to be more potent and selective than type I inhibitors, which target a DFG-in conformation. Nine type II inhibitors are currently clinically approved, with more undergoing clinical development. Resistance-conferring secondary mutations emerged with the first series of type II inhibitors, most commonly at residues within the kinase activation loop and at the "gatekeeper" position. Recently, new inhibitors have been developed to overcome such mutations; however, mutations activating other pathways (and/or other targets) have subsequently emerged on occasion. Here, we systematically summarize the secondary mutations that confer resistance to type II inhibitors, the structural basis for resistance, newer inhibitors designed to overcome resistance, as well as the challenges and opportunities for the development of new inhibitors to overcome secondary kinase domain mutations.
Collapse
Affiliation(s)
- Xiuqi Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Rosa Anna DeFilippis
- Division of Hematology/Oncology, University of California, San Francisco, California 94143, United States
| | - Wei Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
- Department of Pharmacology, School of Medicine, The University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Neil P Shah
- Division of Hematology/Oncology, University of California, San Francisco, California 94143, United States
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
- Department of Pharmacology, School of Medicine, The University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| |
Collapse
|
29
|
Liu J, Gu J. Importance of PTM of FLT3 in acute myeloid leukemia. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1199-1207. [PMID: 38915288 PMCID: PMC11399421 DOI: 10.3724/abbs.2024112] [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: 04/27/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase expressed in hematopoietic cells. Internal-tandem duplication domain (ITD) mutation and tyrosine kinase domain (TKD) mutation are the two most common mutations in acute myeloid leukemia (AML). Post-translational modifications (PTMs) of FLT3, such as glycosylation and ubiquitination, have been shown to impact various aspects of the protein in both wild-type (WT) and mutant forms of FLT3. In this review, we describe how the glycosylation status of FLT3 affects its subcellular localization, which significantly impacts the activation of downstream signaling, and the impact of specific ubiquitination on FLT3 function and stability, which may be associated with disease progression. Moreover, potential novel therapeutic strategies involving a combination of FLT3 tyrosine kinase inhibitors and drugs targeting glycosylation or ubiquitination are discussed.
Collapse
Affiliation(s)
- Jianwei Liu
- />Division of Regulatory GlycobiologyInstitute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical University4-4-1 KomatsushimaAoba-kuSendai Miyagi981-8558Japan
| | - Jianguo Gu
- />Division of Regulatory GlycobiologyInstitute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical University4-4-1 KomatsushimaAoba-kuSendai Miyagi981-8558Japan
| |
Collapse
|
30
|
Sulaibi MA, Zahra J, Bardaweel S, El Abadleh M, Taha MO. Docking-guided exploration of the anti-flt3 potential of isoindigo derivatives towards potential treatments of acute myeloid leukemia. Med Chem Res 2024. [DOI: 10.1007/s00044-024-03259-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/07/2024] [Indexed: 07/10/2024]
|
31
|
Tecik M, Adan A. Emerging DNA Methylome Targets in FLT3-ITD-Positive Acute Myeloid Leukemia: Combination Therapy with Clinically Approved FLT3 Inhibitors. Curr Treat Options Oncol 2024; 25:719-751. [PMID: 38696033 PMCID: PMC11222205 DOI: 10.1007/s11864-024-01202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2024] [Indexed: 07/04/2024]
Abstract
OPINION STATEMENT The internal tandem duplication (ITD) mutation of the FMS-like receptor tyrosine kinase 3 (FLT3-ITD) is the most common mutation observed in approximately 30% of acute myeloid leukemia (AML) patients. It represents poor prognosis due to continuous activation of downstream growth-promoting signaling pathways such as STAT5 and PI3K/AKT. Hence, FLT3 is considered an attractive druggable target; selective small FLT3 inhibitors (FLT3Is), such as midostaurin and quizartinib, have been clinically approved. However, patients possess generally poor remission rates and acquired resistance when FLT3I used alone. Various factors in patients could cause these adverse effects including altered epigenetic regulation, causing mainly abnormal gene expression patterns. Epigenetic modifications are required for hematopoietic stem cell (HSC) self-renewal and differentiation; however, critical driver mutations have been identified in genes controlling DNA methylation (such as DNMT3A, TET2, IDH1/2). These regulators cause leukemia pathogenesis and affect disease diagnosis and prognosis when they co-occur with FLT3-ITD mutation. Therefore, understanding the role of different epigenetic alterations in FLT3-ITD AML pathogenesis and how they modulate FLT3I's activity is important to rationalize combinational treatment approaches including FLT3Is and modulators of methylation regulators or pathways. Data from ongoing pre-clinical and clinical studies will further precisely define the potential use of epigenetic therapy together with FLT3Is especially after characterized patients' mutational status in terms of FLT3 and DNA methlome regulators.
Collapse
Affiliation(s)
- Melisa Tecik
- Bioengineering Program, Graduate School of Engineering and Science, Abdullah Gul University, Kayseri, Turkey
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Turkey.
| |
Collapse
|
32
|
Mohebbi A, Shahriyary F, Farrokhi V, Bandar B, Saki N. A systematic review of second-generation FLT3 inhibitors for treatment of patients with relapsed/refractory acute myeloid leukemia. Leuk Res 2024; 141:107505. [PMID: 38692232 DOI: 10.1016/j.leukres.2024.107505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a complex disease with diverse mutations, including prevalent mutations in the FMS-like receptor tyrosine kinase 3 (FLT3) gene that lead to poor prognosis. Recent advancements have introduced FLT3 inhibitors that have improved outcomes for FLT3-mutated AML patients, however, questions remain on their application in complex conditions such as relapsed/refractory (R/R) disease. Therefore, we aimed to evaluate the clinical effectiveness of second-generation FLT3 inhibitors in treating patients with R/R AML. METHODS A systematic literature search of PubMed, MEDLINE, SCOPUS and Google Scholar databases was made to identify relevant studies up to January 30, 2024. This study was conducted following the guidelines of the PRISMA. RESULTS The ADMIRAL trial revealed significantly improved overall survival and complete remission rates with gilteritinib compared to salvage chemotherapy, with manageable adverse effects. Ongoing research explores its potential in combination therapies, showing synergistic effects with venetoclax and promising outcomes in various clinical trials. The QuANTUM-R trial suggested longer overall survival with quizartinib compared to standard chemotherapy, although concerns were raised regarding trial design and cardiotoxicity. Ongoing research explores combination therapies involving quizartinib, such as doublet or triplet regimens with venetoclax, showing promising outcomes in FLT3-mutated AML patients. CONCLUSION These targeted therapies offer promise for managing this subgroup of AML patients, but further research is needed to optimize their use. This study underscores the importance of personalized treatment based on genetic mutations in AML, paving the way for more effective and tailored approaches to combat the disease.
Collapse
Affiliation(s)
- Alireza Mohebbi
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Shahriyary
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Vida Farrokhi
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Bita Bandar
- Department of Medical Laboratory, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Department of Medical Laboratory, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
33
|
Zheng X, Chen Z, Guo M, Liang H, Song X, Liu Y, Liao Z, Zhang Y, Guo J, Zhou Y, Zhang ZM, Tu Z, Zhang Y, Chen Y, Zhang Z, Lu X. Structure-Based Optimization of Pyrazinamide-Containing Macrocyclic Derivatives as Fms-like Tyrosine Kinase 3 (FLT3) Inhibitors to Overcome Clinical Mutations. ACS Pharmacol Transl Sci 2024; 7:1485-1506. [PMID: 38751627 PMCID: PMC11092118 DOI: 10.1021/acsptsci.4c00071] [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: 02/08/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 05/18/2024]
Abstract
Secondary mutations in Fms-like tyrosine kinase 3-tyrosine kinase domain (FLT3-TKD) (e.g., D835Y and F691L) have become a major on-target resistance mechanism of FLT3 inhibitors, which present a significant clinical challenge. To date, no effective drugs have been approved to simultaneously overcome clinical resistance caused by these two mutants. Thus, a series of pyrazinamide macrocyclic compounds were first designed and evaluated to overcome the secondary mutations of FLT3. The representative 8v exhibited potent inhibitory activities against FLT3D835Y and FLT3D835Y/F691L with IC50 values of 1.5 and 9.7 nM, respectively. 8v also strongly suppressed the proliferation against Ba/F3 cells transfected with FLT3-ITD, FLT3-ITD-D835Y, FLT3-ITD-F691L, FLT3-ITD-D835Y-F691L, and MV4-11 acute myeloid leukemia (AML) cell lines with IC50 values of 12.2, 10.5, 24.6, 16.9, and 6.8 nM, respectively. Furthermore, 8v demonstrated ideal anticancer efficacy in a Ba/F3-FLT3-ITD-D835Y xenograft model. The results suggested that 8v can serve as a promising macrocycle-based FLT3 inhibitor for the treatment of AML.
Collapse
Affiliation(s)
- Xuan Zheng
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhiwen Chen
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Ming Guo
- Department
of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local
Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hong Liang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Xiaojuan Song
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yiling Liu
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhenling Liao
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yan Zhang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Jing Guo
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yang Zhou
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhi-min Zhang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhengchao Tu
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Ye Zhang
- Guangxi
Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Yongheng Chen
- Department
of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local
Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhang Zhang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Xiaoyun Lu
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
- Department
of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510632, China
| |
Collapse
|
34
|
Qi J, Choi I, Ota S, Ichikawa S, Fujishima N, Iida H, Sugiura I, Sugiura K, Murata Y, Inoue H, Ohwada S, Wang J. Safety and Pharmacokinetics of Quizartinib Combination Therapy With Standard Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia: Results from Two Phase 1 Trials in Japan and China. Clin Pharmacol Drug Dev 2024; 13:560-571. [PMID: 38284515 DOI: 10.1002/cpdd.1353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/28/2023] [Indexed: 01/30/2024]
Abstract
Quizartinib is a potent, oral, second-generation, selective type II FMS-like receptor tyrosine kinase 3 (FLT3) inhibitor. It has shown improved overall survival in a randomized, multinational, Phase 3 (QuANTUM-First) study in patients with FLT3-internal tandem duplication (ITD)-positive newly diagnosed acute myeloid leukemia. We conducted 2 Phase 1b studies in Japan and China to evaluate the safety, pharmacokinetics, and efficacy of quizartinib in combination with standard induction and consolidation chemotherapy in patients with newly diagnosed acute myeloid leukemia. Quizartinib was started at a dose level of 20 mg/day and then escalated to 40 mg/day, the dose used in the Phase 3 study. Seven patients were enrolled according to the 3 + 3 dose-escalation method in each study, including 3 patients who were FLT3-ITD positive. No dose-limiting toxicities were observed at dose levels up to 40 mg/day in both studies. Grade 3 or higher, quizartinib-related, treatment-emergent adverse events included febrile neutropenia, hematologic toxicities, and infections. QT prolongation on electrocardiogram was observed in 5 patients. The pharmacokinetics of quizartinib and its metabolite AC886 were similar between the studies and consistent with previous findings in the United States. We confirmed the tolerability of Japanese and Chinese patients to the dose of quizartinib and chemotherapy regimens used in the QuANTUM-First study.
Collapse
Affiliation(s)
- Junyuan Qi
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Ilseung Choi
- National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | | | | | | | - Hiroatsu Iida
- National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | | | | | | | | | | | - Jianxiang Wang
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
35
|
Wang J, Rong Q, Ye L, Fang B, Zhao Y, Sun Y, Zhou H, Wang D, He J, Cui Z, Zhang Q, Kang D, Hu L. Discovery of a Novel Orally Bioavailable FLT3-PROTAC Degrader for Efficient Treatment of Acute Myeloid Leukemia and Overcoming Resistance of FLT3 Inhibitors. J Med Chem 2024. [PMID: 38655686 DOI: 10.1021/acs.jmedchem.4c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Fms-like tyrosine receptor kinase 3 (FLT3) proteolysis-targeting chimeras (PROTACs) represent a promising approach to eliminate the resistance of FLT3 inhibitors. However, due to the poor druggability of PROTACs, the development of orally bioavailable FLT3-PROTACs faces great challenges. Herein, a novel orally bioavailable FLT3-ITD degrader A20 with excellent pharmacokinetic properties was discovered through reasonable design. A20 selectively inhibited the proliferation of FLT3-ITD mutant acute myeloid leukemia (AML) cells and potently induced FLT3-ITD degradation through the ubiquitin-proteasome system. Notably, oral administration of A20 resulted in complete tumor regression on subcutaneous AML xenograft models. Furthermore, on systemic AML xenograft models, A20 could completely eliminate the CD45+CD33+ human leukemic cells in murine and significantly prolonged the survival time of mice. Most importantly, A20 exerted significantly improved antiproliferative activity against drug-resistant AML cells compared to existing FLT3 inhibitors. These findings suggested that A20 could serve as a promising drug candidate for relapsed or refractory AML.
Collapse
Affiliation(s)
- Junwei Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Quanjin Rong
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Lei Ye
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Bingqian Fang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yifan Zhao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yu Sun
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Haikun Zhou
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Dan Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Jinting He
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Zhenzhen Cui
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Qijian Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Di Kang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
36
|
Coleman DJ, Keane P, Chin PS, Ames L, Kellaway S, Blair H, Khan N, Griffin J, Holmes E, Maytum A, Potluri S, Strate L, Koscielniak K, Raghavan M, Bushweller J, Heidenreich O, Rabbitts T, Cockerill PN, Bonifer C. Pharmacological inhibition of RAS overcomes FLT3 inhibitor resistance in FLT3-ITD+ AML through AP-1 and RUNX1. iScience 2024; 27:109576. [PMID: 38638836 PMCID: PMC11024925 DOI: 10.1016/j.isci.2024.109576] [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: 10/23/2023] [Revised: 02/16/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
AML is characterized by mutations in genes associated with growth regulation such as internal tandem duplications (ITD) in the receptor kinase FLT3. Inhibitors targeting FLT3 (FLT3i) are being used to treat patients with FLT3-ITD+ but most relapse and become resistant. To elucidate the resistance mechanism, we compared the gene regulatory networks (GRNs) of leukemic cells from patients before and after relapse, which revealed that the GRNs of drug-responsive patients were altered by rewiring their AP-1-RUNX1 axis. Moreover, FLT3i induces the upregulation of signaling genes, and we show that multiple cytokines, including interleukin-3 (IL-3), can overcome FLT3 inhibition and send cells back into cycle. FLT3i leads to loss of AP-1 and RUNX1 chromatin binding, which is counteracted by IL-3. However, cytokine-mediated drug resistance can be overcome by a pan-RAS inhibitor. We show that cytokines instruct AML growth via the transcriptional regulators AP-1 and RUNX1 and that pan-RAS drugs bypass this barrier.
Collapse
Affiliation(s)
- Daniel J.L. Coleman
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Peter Keane
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Paulynn S. Chin
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Luke Ames
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Sophie Kellaway
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Helen Blair
- Wolfson Childhood Cancer Research Centre, University of Newcastle, Newcastle upon Tyne, UK
| | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - James Griffin
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Elizabeth Holmes
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Alexander Maytum
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Sandeep Potluri
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Lara Strate
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Kinga Koscielniak
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Manoj Raghavan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - John Bushweller
- School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Olaf Heidenreich
- Wolfson Childhood Cancer Research Centre, University of Newcastle, Newcastle upon Tyne, UK
- Princess Máxima Centrum of Pediatric Oncology, Utrecht, the Netherlands
| | - Terry Rabbitts
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Peter N. Cockerill
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Constanze Bonifer
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
37
|
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
|
38
|
Abaza Y, McMahon C, Garcia JS. Advancements and Challenges in the Treatment of AML. Am Soc Clin Oncol Educ Book 2024; 44:e438662. [PMID: 38662975 DOI: 10.1200/edbk_438662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The therapeutic arsenal for the management of AML has expanded significantly in recent years. Before 2017, newly diagnosed AML was treated with either standard cytarabine- and anthracycline-based induction chemotherapy (for all fit patients) or a single-agent hypomethylating agent (in unfit patients or those 75 years and older). While assessing patient fitness remains important, characterizing the disease biology has become critical to select the optimal initial therapy for each patient with more options available. FLT3 inhibitors, gemtuzumab ozogamicin, and CPX-351 have been shown to improve outcomes for specific subsets of patients. Venetoclax (VEN) with a hypomethylating agent (HMA) is the standard-of-care frontline regimen for most older patients, except perhaps for those with an IDH1 mutation where ivosidenib with azacitidine may also be considered. On the basis of the success seen with HMA/VEN in older patients, there is now increasing interest in incorporating VEN into frontline regimens in younger patients, with promising data from multiple early phase studies. This article focuses on recent updates and ongoing challenges in the management of AML, with a particular focus on the ongoing challenge of secondary AML and considerations regarding the selection of initial therapy in younger patients. An overview of common side effects and toxicities associated with targeted therapies is also presented here, along with recommended strategies to mitigate these risks.
Collapse
Affiliation(s)
- Yasmin Abaza
- Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Christine McMahon
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | | |
Collapse
|
39
|
Monogiou Belik D, Bernasconi R, Xu L, Della Verde G, Lorenz V, Grüterich V, Balzarolo M, Mochizuki M, Pfister O, Kuster GM. The Flt3-inhibitor quizartinib augments apoptosis and promotes maladaptive remodeling after myocardial infarction in mice. Apoptosis 2024; 29:357-371. [PMID: 37945814 PMCID: PMC10873224 DOI: 10.1007/s10495-023-01911-8] [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] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) targeting fms-like tyrosine kinase 3 (Flt3) such as quizartinib were specifically designed for acute myeloid leukemia treatment, but also multi-targeting TKIs applied to solid tumor patients inhibit Flt3. Flt3 is expressed in the heart and its activation is cytoprotective in myocardial infarction (MI) in mice. OBJECTIVES We sought to test whether Flt3-targeting TKI treatment aggravates cardiac injury after MI. METHODS AND RESULTS Compared to vehicle, quizartinib (10 mg/kg/day, gavage) did not alter cardiac dimensions or function in healthy mice after four weeks of therapy. Pretreated mice were randomly assigned to MI or sham surgery while receiving quizartinib or vehicle for one more week. Quizartinib did not aggravate the decline in ejection fraction, but significantly enhanced ventricular dilatation one week after infarction. In addition, apoptotic cell death was significantly increased in the myocardium of quizartinib-treated compared to vehicle-treated mice. In vitro, quizartinib dose-dependently decreased cell viability in neonatal rat ventricular myocytes and in H9c2 cells, and increased apoptosis as assessed in the latter. Together with H2O2, quizartinib potentiated the phosphorylation of the pro-apoptotic mitogen activated protein kinase p38 and augmented H2O2-induced cell death and apoptosis beyond additive degree. Pretreatment with a p38 inhibitor abolished apoptosis under quizartinib and H2O2. CONCLUSION Quizartinib potentiates apoptosis and promotes maladaptive remodeling after MI in mice at least in part via a p38-dependent mechanism. These findings are consistent with the multi-hit hypothesis of cardiotoxicity and make cardiac monitoring in patients with ischemic heart disease under Flt3- or multi-targeting TKIs advisable.
Collapse
Affiliation(s)
- Daria Monogiou Belik
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Riccardo Bernasconi
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vera Lorenz
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vivienne Grüterich
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Melania Balzarolo
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Michika Mochizuki
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland.
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
40
|
Liu J, Isaji T, Komatsu S, Sun Y, Xu X, Fukuda T, Fujimura T, Takahashi S, Gu J. BRCC36 associates with FLT3-ITD to regulate its protein stability and intracellular signaling in acute myeloid leukemia. Cancer Sci 2024; 115:1196-1208. [PMID: 38288901 PMCID: PMC11007003 DOI: 10.1111/cas.16090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 04/12/2024] Open
Abstract
Fms-like tyrosine kinase-3 (FLT3) is a commonly mutated gene in acute myeloid leukemia (AML). The two most common mutations are the internal-tandem duplication domain (ITD) mutation and the tyrosine kinase domain (TKD) mutation. FLT3-ITD and FLT3-TKD exhibit distinct protein stability, cellular localization, and intracellular signaling. To understand the underlying mechanisms, we performed proximity labeling with TurboID to identify proteins that regulate FLT3-ITD or -TKD differently. We found that BRCA1/BRCA2-containing complex subunit 36 (BRCC36), a specific K63-linked polyubiquitin deubiquitinase, was exclusively associated with ITD, not the wild type of FLT3 and TKD. Knockdown of BRCC36 resulted in decreased signal transducers and activators of transcription 5 phosphorylation and cell proliferation in ITD cells. Consistently, treatment with thiolutin, an inhibitor of BRCC36, specifically suppressed cell proliferation and induced cell apoptosis in ITD cells. Thiolutin efficiently affected leukemia cell lines expressing FLT3-ITD cell viability and exhibited mutual synergies with quizartinib, a standard clinical medicine for AML. Furthermore, mutation of the lysine at 609 of ITD led to significant suppression of K63 polyubiquitination and decreased its stability, suggesting that K609 is a critical site for K63 ubiquitination specifically recognized by BRCC36. These data indicate that BRCC36 is a specific regulator for FLT3-ITD, which may shed light on developing a novel therapeutic approach for AML.
Collapse
Affiliation(s)
- Jianwei Liu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Sachiko Komatsu
- Division of Bioanalytical ChemistryTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Yuhan Sun
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Xing Xu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Tsutomu Fujimura
- Division of Bioanalytical ChemistryTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Shinichiro Takahashi
- Division of Laboratory Medicine, Faculty of MedicineTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and GlycobiologyTohoku Medical and Pharmaceutical UniversitySendaiMiyagiJapan
| |
Collapse
|
41
|
Jin J, Hou S, Yao Y, Liu M, Mao L, Yang M, Tong H, Zeng T, Huang J, Zhu Y, Wang H. Phosphoproteomic Characterization and Kinase Signature Predict Response to Venetoclax Plus 3+7 Chemotherapy in Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305885. [PMID: 38161214 PMCID: PMC10953567 DOI: 10.1002/advs.202305885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Resistance to chemotherapy remains a formidable obstacle in acute myeloid leukemia (AML) therapeutic management, necessitating the exploration of optimal strategies to maximize therapeutic benefits. Venetoclax with 3+7 daunorubicin and cytarabine (DAV regimen) in young adult de novo AML patients is evaluated. 90% of treated patients achieved complete remission, underscoring the potential of this regimen as a compelling therapeutic intervention. To elucidate underlying mechanisms governing response to DAV in AML, quantitative phosphoproteomics to discern distinct molecular signatures characterizing a subset of DAV-sensitive patients is used. Cluster analysis reveals an enrichment of phosphoproteins implicated in chromatin organization and RNA processing within DAV-susceptible and DA-resistant AML patients. Furthermore, kinase activity profiling identifies AURKB as a candidate indicator of DAV regimen efficacy in DA-resistant AML due to AURKB activation. Intriguingly, AML cells overexpressing AURKB exhibit attenuated MCL-1 expression, rendering them receptive to DAV treatment and maintaining them resistant to DA treatment. Moreover, the dataset delineates a shared kinase, AKT1, associated with DAV response. Notably, AKT1 inhibition augments the antileukemic efficacy of DAV treatment in AML. Overall, this phosphoproteomic study identifies the role of AURKB as a predictive biomarker for DA, but not DAV, resistance and proposes a promising strategy to counteract therapy resistance in AML.
Collapse
Affiliation(s)
- Jie Jin
- Department of Hematologythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- Zhejiang Provincial Key Lab of Hematopoietic MalignancyZhejiang UniversityHangzhouZhejiangP. R. China
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouChina
- Zhejiang University Cancer CenterHangzhouZhejiangP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanP. R. China
| | - Shangyu Hou
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092P.R. China
| | - Yiyi Yao
- Department of Hematologythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- Zhejiang Provincial Key Lab of Hematopoietic MalignancyZhejiang UniversityHangzhouZhejiangP. R. China
| | - Miaomiao Liu
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092P.R. China
| | - Liping Mao
- Department of Hematologythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- Zhejiang Provincial Key Lab of Hematopoietic MalignancyZhejiang UniversityHangzhouZhejiangP. R. China
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouChina
| | - Min Yang
- Department of Hematologythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- Zhejiang Provincial Key Lab of Hematopoietic MalignancyZhejiang UniversityHangzhouZhejiangP. R. China
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouChina
| | - Hongyan Tong
- Department of Hematologythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- Zhejiang Provincial Key Lab of Hematopoietic MalignancyZhejiang UniversityHangzhouZhejiangP. R. China
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouChina
- Zhejiang University Cancer CenterHangzhouZhejiangP. R. China
| | - Tao Zeng
- Biomedical big data centerthe First Affiliated HospitalZhejiang University School of MedicineHangzhou, Zhejiang310003P.R. China
| | - Jinyan Huang
- Biomedical big data centerthe First Affiliated HospitalZhejiang University School of MedicineHangzhou, Zhejiang310003P.R. China
| | - Yinghui Zhu
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092P.R. China
- Frontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchTongji UniversityShanghai200092P.R. China
| | - Huafeng Wang
- Department of Hematologythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- Zhejiang Provincial Key Lab of Hematopoietic MalignancyZhejiang UniversityHangzhouZhejiangP. R. China
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouChina
- Zhejiang University Cancer CenterHangzhouZhejiangP. R. China
| |
Collapse
|
42
|
Wang YT, Yang PC, Zhang YF, Sun JF. Synthesis and clinical application of new drugs approved by FDA in 2023. Eur J Med Chem 2024; 265:116124. [PMID: 38183778 DOI: 10.1016/j.ejmech.2024.116124] [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: 12/29/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
In 2023, the U.S. Food and Drug Administration (FDA) granted approval to a total of 55 new drugs, comprising 29 new chemical entities (NCEs) and 25 new biological entities (NBEs). These drugs primarily focus on oncology, the central nervous system, anti-infection, hematology, cardiovascular, ophthalmology, immunomodulatory and other therapeutic areas. Out of the 55 drugs, 33 (60 %) underwent an accelerated review process and received approval, while 25 (45 %) were specifically approved for the treatment of rare diseases. The purpose of this review is to provide an overview of the clinical uses and production techniques of 29 newly FDA-approved NCEs in 2023. Our intention is to offer a comprehensive understanding of the synthetic approaches employed in the creation of these drug molecules, with the aim of inspiring the development of novel, efficient, and applicable synthetic methodologies.
Collapse
Affiliation(s)
- Ya-Tao Wang
- First People's Hospital of Shangqiu, Henan Province, Shangqiu, 476100, China.
| | - Peng-Cheng Yang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin, 133002, China
| | - Yan-Feng Zhang
- Shangqiu Municipal Hospital, Henan Province, Shangqiu, 476100, China.
| | - Jin-Feng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin, 133002, China; Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| |
Collapse
|
43
|
Wang X, DeFilippis RA, Leung YK, Shah NP, Li HY. N-(3-Methoxyphenyl)-6-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)pyridin-2-amine is an inhibitor of the FLT3-ITD and BCR-ABL pathways, and potently inhibits FLT3-ITD/D835Y and FLT3-ITD/F691L secondary mutants. Bioorg Chem 2024; 143:106966. [PMID: 37995643 PMCID: PMC11586108 DOI: 10.1016/j.bioorg.2023.106966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023]
Abstract
Activating mutations within FLT3 make up 30 % of all newly diagnosed acute myeloid leukemia (AML) cases, with the most common mutation being an internal tandem duplication (FLT3-ITD) in the juxtamembrane region (25 %). Currently, two generations of FLT3 kinase inhibitors have been developed, with three inhibitors clinically approved. However, treatment of FLT3-ITD mutated AML is limited due to the emergence of secondary clinical resistance, caused by multiple mechanism including on-target FLT3 secondary mutations - FLT3-ITD/D835Y and FLT3-ITD/F691L being the most common, as well as the off-target activation of alternative pathways including the BCR-ABL pathway. Through the screening of imidazo[1,2-a]pyridine derivatives, N-(3-methoxyphenyl)-6-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)pyridin-2-amine (compound 1) was identified as an inhibitor of both the FLT3-ITD and BCR-ABL pathways. Compound 1 potently inhibits clinically related leukemia cell lines driven by FLT3-ITD, FLT3-ITD/D835Y, FLT3-ITD/F691L, or BCR-ABL. Studies indicate that it mediates proapoptotic effects on cells by inhibiting FLT3 and BCR-ABL pathways, and other possible targets. Compound 1 is more potent against FLT3-ITD than BCR-ABL, and it may have other possible targets; however, compound 1 is first step for further optimization for the development of a balanced FLT3-ITD/BCR-ABL dual inhibitor for the treatment of relapsed FLT3-ITD mutated AML with multiple secondary clinical resistant subtypes such as FLT3-ITD/D835Y, FLT3-ITD/F691L, and cells co-expressing FLT3-ITD and BCR-ABL.
Collapse
Affiliation(s)
- Xiuqi Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rosa Anna DeFilippis
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Neil P Shah
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
44
|
Wang X, DeFilippis RA, Weldemichael T, Gunaganti N, Tran P, Leung YK, Shah NP, Li HY. An imidazo[1,2-a]pyridine-pyridine derivative potently inhibits FLT3-ITD and FLT3-ITD secondary mutants, including gilteritinib-resistant FLT3-ITD/F691L. Eur J Med Chem 2024; 264:115977. [PMID: 38056299 PMCID: PMC11590664 DOI: 10.1016/j.ejmech.2023.115977] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/07/2023] [Accepted: 11/18/2023] [Indexed: 12/08/2023]
Abstract
FLT3 activating mutations are detected in approximately 30 % of newly diagnosed acute myeloid leukemia (AML) cases, most commonly consisting of internal tandem duplication (ITD) mutations in the juxtamembrane region. Recently, several FLT3 inhibitors have demonstrated clinical activity and three are currently approved - midostaurin, quizartinib, and gilteritinib. Midostaurin is a first-generation FLT3 inhibitor with minimal activity as monotherapy. Midostaurin lacks selectivity and is only approved by the USFDA for use in combination with other chemotherapy agents. The second-generation inhibitors quizartinib and gilteritinib display improved specificity and selectivity, and have been approved for use as monotherapy. However, their clinical efficacies are limited in part due to the emergence of drug-resistant FLT3 secondary mutations in the tyrosine kinase domain at positions D835 and F691. Therefore, in order to overcome drug resistance and further improve outcomes, new compounds targeting FLT3-ITD with secondary mutants are urgently needed. In this study, through the structural modification of a reported compound Ling-5e, we identified compound 24 as a FLT3 inhibitor that is equally potent against FLT3-ITD and the clinically relevant mutants FLT3-ITD/D835Y, and FLT3-ITD/F691L. Its inhibitory effects were demonstrated in both cell viability assays and western blots analyses. When tested against cell lines lacking activating mutations in FLT3, no non-specific cytotoxicity effect was observed. Interestingly, molecular docking results showed that compound 24 may adopt different binding conformations with FLT3-F691L compared to FLT3, which may explain its retained activity against FLT3-ITD/F691L. In summary, compound 24 has inhibition potency on FLT3 comparable to gilteritinib, but a more balanced inhibition on FLT3 secondary mutations, especially FLT3-ITD/F691L which is gilteritinib resistant. Compound 24 may serve as a promising lead for the drug development of either primary or relapsed AML with FLT3 secondary mutations.
Collapse
Affiliation(s)
- Xiuqi Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rosa Anna DeFilippis
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Tsigereda Weldemichael
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Naresh Gunaganti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Phuc Tran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Neil P Shah
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
45
|
Bergeron J, Capo-Chichi JM, Tsui H, Mahe E, Berardi P, Minden MD, Brandwein JM, Schuh AC. The Clinical Utility of FLT3 Mutation Testing in Acute Leukemia: A Canadian Consensus. Curr Oncol 2023; 30:10410-10436. [PMID: 38132393 PMCID: PMC10742150 DOI: 10.3390/curroncol30120759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are detected in approximately 20-30% of patients with acute myeloid leukemia (AML), with the presence of a FLT3 internal tandem duplication (FLT3-ITD) mutation being associated with an inferior outcome. Assessment of FLT3 mutational status is now essential to define optimal upfront treatment in both newly diagnosed and relapsed AML, to support post-induction allogeneic hematopoietic stem cell transplantation (alloSCT) decision-making, and to evaluate treatment response via measurable (minimal) residual disease (MRD) evaluation. In view of its importance in AML diagnosis and management, the Canadian Leukemia Study Group/Groupe canadien d'étude sur la leucémie (CLSG/GCEL) undertook the development of a consensus statement on the clinical utility of FLT3 mutation testing, as members reported considerable inter-center variability across Canada with respect to testing availability and timing of use, methodology, and interpretation. The CLSG/GCEL panel identified key clinical and hematopathological questions, including: (1) which patients should be tested for FLT3 mutations, and when?; (2) which is the preferred method for FLT3 mutation testing?; (3) what is the clinical relevance of FLT3-ITD size, insertion site, and number of distinct FLT3-ITDs?; (4) is there a role for FLT3 analysis in MRD assessment?; (5) what is the clinical relevance of the FLT3-ITD allelic burden?; and (6) how should results of FLT3 mutation testing be reported? The panel followed an evidence-based approach, taken together with Canadian clinical and laboratory experience and expertise, to create a consensus document to facilitate a more uniform approach to AML diagnosis and treatment across Canada.
Collapse
Affiliation(s)
- Julie Bergeron
- CEMTL Installation Maisonneuve-Rosemont, Institut Universitaire d’Hématologie-Oncologie et de Thérapie Cellulaire, Université de Montréal, Montréal, QC H1T 2M4, Canada
| | - Jose-Mario Capo-Chichi
- Division of Clinical Laboratory Genetics, Department of Laboratory Medicine and Pathobiology, Laboratory Medicine Program, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada;
| | - Hubert Tsui
- Division of Hematological Pathology, Department of Laboratory Medicine and Molecular Diagnostics, Precision Diagnostics and Therapeutics Program, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Laboratory Medicine and Pathobiology, Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Etienne Mahe
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Division of Hematology and Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Philip Berardi
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital/Eastern Ontario Regional Laboratory Association, Ottawa, ON K1H 8M2, Canada;
- Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mark D. Minden
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.D.M.); (A.C.S.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Joseph M. Brandwein
- Division of Hematology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada;
| | - Andre C. Schuh
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.D.M.); (A.C.S.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
46
|
Niu ZX, Wang YT, Sun JF, Nie P, Herdewijn P. Recent advance of clinically approved small-molecule drugs for the treatment of myeloid leukemia. Eur J Med Chem 2023; 261:115827. [PMID: 37757658 DOI: 10.1016/j.ejmech.2023.115827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Myeloid leukemia denotes a hematologic malignancy characterized by aberrant proliferation and impaired differentiation of blood progenitor cells within the bone marrow. Despite the availability of several treatment options, the clinical outlook for individuals afflicted with myeloid leukemia continues to be unfavorable, making it a challenging disease to manage. Over the past, substantial endeavors have been dedicated to the identification of novel targets and the advancement of enhanced therapeutic modalities to ameliorate the management of this disease, resulting in the discovery of many clinically approved small-molecule drugs for myeloid leukemia, including histone deacetylase inhibitors, hypomethylating agents, and tyrosine kinase inhibitors. This comprehensive review succinctly presents an up-to-date assessment of the application and synthetic routes of clinically sanctioned small-molecule drugs employed in the treatment of myeloid leukemia. Additionally, it provides a concise exploration of the pertinent challenges and prospects encompassing drug resistance and toxicity. Overall, this review effectively underscores the considerable promise exhibited by clinically endorsed small-molecule drugs in the therapeutic realm of myeloid leukemia, while concurrently shedding light on the prospective avenues that may shape the future landscape of drug development within this domain.
Collapse
Affiliation(s)
- Zhen-Xi Niu
- Department of Pharmacy, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Ya-Tao Wang
- First People's Hospital of Shangqiu, Henan Province, Shangqiu, 476100, China; Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| | - Jin-Feng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin, 133002, China.
| | - Peng Nie
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| |
Collapse
|
47
|
Ma H, Cui J, Liu Z, Fang W, Lu S, Cao S, Zhang Y, Chen JA, Lu L, Xie Q, Wang Y, Huang Y, Li K, Tong H, Huang J, Lu W. Blockade of de novo pyrimidine biosynthesis triggers autophagic degradation of oncoprotein FLT3-ITD in acute myeloid leukemia. Oncogene 2023; 42:3331-3343. [PMID: 37752234 DOI: 10.1038/s41388-023-02848-7] [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: 05/23/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
The internal tandem duplication of the FMS-like tyrosine kinase 3 (FLT3-ITD) is one of the most frequent genetic alterations in acute myeloid leukemia (AML). Limited and transient clinical benefit of FLT3 kinase inhibitors (FLT3i) emphasizes the need for alternative therapeutic options for this subset of myeloid malignancies. Herein, we showed that FLT3-ITD mutant (FLT3-ITD+) AML cells were susceptible toward inhibitors of DHODH, a rate-limiting enzyme of de novo pyrimidine biosynthesis. Genetic and pharmacological blockade of DHODH triggered downregulation of FLT3-ITD protein, subsequently suppressed activation of downstream ERK and STAT5, and promoted cell death of FLT3-ITD+ AML cells. Mechanistically, DHODH blockade triggered autophagy-mediated FLT3-ITD degradation via inactivating mTOR, a potent autophagy repressor. Notably, blockade of DHODH synergized with an FDA-approved FLT3i quizartinib in significantly impairing the growth of FLT3-ITD+ AML cells and improving tumor-bearing mice survival. We further demonstrated that DHODH blockade exhibited profound anti-proliferation effect on quizartinib-resistant cells in vitro and in vivo. In summary, this study demonstrates that the induction of degradation of FLT3-ITD protein by DHODH blockade may offer a promising therapeutic strategy for AML patients harboring FLT3-ITD mutation.
Collapse
Affiliation(s)
- Hui Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Jiayan Cui
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Zehui Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Wenqing Fang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Sisi Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Shuying Cao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Yuanyuan Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - Ji-An Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 201203, Shanghai, China
| | - Lixue Lu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 201203, Shanghai, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 201203, Shanghai, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 201203, Shanghai, China
| | - Ying Huang
- NMPA Key Laboratory of Rapid Drug Inspection Technology, Guangdong Institute for Drug Control, 510663, Guangzhou, China
| | - Kongfei Li
- Department of Hematology, People's Hospital Affiliated to Ningbo University, 315000, Ningbo, China
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, 310003, Hangzhou, China
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, 310003, Hangzhou, China
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China.
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, 200241, Shanghai, China.
| |
Collapse
|
48
|
Kannampuzha S, Murali R, Gopalakrishnan AV, Mukherjee AG, Wanjari UR, Namachivayam A, George A, Dey A, Vellingiri B. Novel biomolecules in targeted cancer therapy: a new approach towards precision medicine. Med Oncol 2023; 40:323. [PMID: 37804361 DOI: 10.1007/s12032-023-02168-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/18/2023] [Indexed: 10/09/2023]
Abstract
Cancer is a major threat to human life around the globe, and the discovery of novel biomolecules continue to be an urgent therapeutic need that is still unmet. Precision medicine relies on targeted therapeutic strategies. Researchers are better equipped to develop therapies that target proteins as they understand more about the genetic alterations and molecules that cause progression of cancer. There has been a recent diversification of the sorts of targets exploited in treatment. Therapeutic antibody and biotechnology advancements enabled curative treatments to reach previously inaccessible sites. New treatment strategies have been initiated for several undruggable targets. The application of tailored therapy has been proven to have efficient results in controlling cancer progression. Novel biomolecules like SMDCs, ADCs, mABs, and PROTACS has gained vast attention in the recent years. Several studies have shown that using these novel technology helps in reducing the drug dosage as well as to overcome drug resistance in different cancer types. Therefore, it is crucial to fully untangle the mechanism and collect evidence to understand the significance of these novel drug targets and strategies. This review article will be discussing the importance and role of these novel biomolecules in targeted cancer therapies.
Collapse
Affiliation(s)
- Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - Abhijit Dey
- Department of Medical Services, MGM Cancer Institute, Chennai, Tamil Nadu, 600029, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| |
Collapse
|
49
|
Li M, Wei J, Zhu G, Fu S, He X, Hu X, Yu Y, Mou Y, Wang J, You X, Xiao X, Gu T, Ye Z, Zha Y. Quizartinib inhibits necroptosis by targeting receptor-interacting serine/threonine protein kinase 1. FASEB J 2023; 37:e23178. [PMID: 37698367 DOI: 10.1096/fj.202300600rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023]
Abstract
Systemic inflammatory response syndrome (SIRS), at least in part driven by necroptosis, is characterized by life-threatening multiple organ failure. Blocking the progression of SIRS and consequent multiple organ dysfunction is challenging. Receptor-interacting serine/threonine protein kinase 1 (RIPK1) is an important cell death and inflammatory mediator, making it a potential treatment target in several diseases. Here, using a drug repurposing approach, we show that inhibiting RIPK1 is also an effective treatment for SIRS. We performed cell-based high-throughput drug screening of an US Food and Drug Administration (FDA)-approved drug library that contains 1953 drugs to identify effective inhibitors of necroptotic cell death by SYTOX green staining. Dose-response validation of the top candidate, quizartinib, was conducted in two cell lines of HT-22 and MEFs. The effect of quizartinib on necroptosis-related proteins was evaluated using western blotting, immunoprecipitation, and an in vitro RIPK1 kinase assay. The in vivo effects of quizartinib were assessed in a murine tumor necrosis factor α (TNFα)-induced SIRS model. High-throughput screening identified quizartinib as the top "hit" in the compound library that rescued cells from necroptosis in vitro. Quizartinib inhibited necroptosis by directly inhibiting RIPK1 kinase activity and blocking downstream complex IIb formation. Furthermore, quizartinib protected mice against TNFα-induced SIRS. Quizartinib, as an FDA-approved drug with proven safety and efficacy, was repurposed for targeted inhibition of RIPK1. This work provides essential preclinical data for transferring quizartinib to the treatment of RIPK1-dependent necroptosis-induced inflammatory diseases, including SIRS.
Collapse
Affiliation(s)
- Min Li
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Jun Wei
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Guofeng Zhu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Shufang Fu
- Yichang Central People's Hospital, Yichang, China
- Department of Paediatrics, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Xiaoyan He
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Xinqian Hu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Yajie Yu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Yan Mou
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Jia Wang
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Xiaoling You
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Xin Xiao
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Tanrong Gu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Zhi Ye
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Yunhong Zha
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
50
|
Chen PY, Huang BJ, Harris M, Boone C, Wang W, Carias H, Mesiona B, Mavrici D, Kohler AC, Bollag G, Zhang C, Zhang Y, Shannon K. Structural and functional analyses of a germline KRAS T50I mutation provide insights into Raf activation. JCI Insight 2023; 8:e168445. [PMID: 37681415 PMCID: PMC10544224 DOI: 10.1172/jci.insight.168445] [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: 01/04/2023] [Accepted: 08/02/2023] [Indexed: 09/09/2023] Open
Abstract
A T50I substitution in the K-Ras interswitch domain causes Noonan syndrome and emerged as a third-site mutation that restored the in vivo transforming activity and constitutive MAPK pathway activation by an attenuated KrasG12D,E37G oncogene in a mouse leukemia model. Biochemical and crystallographic data suggested that K-RasT50I increases MAPK signal output through a non-GTPase mechanism, potentially by promoting asymmetric Ras:Ras interactions between T50 and E162. We generated a "switchable" system in which K-Ras mutant proteins expressed at physiologic levels supplant the fms like tyrosine kinase 3 (FLT3) dependency of MOLM-13 leukemia cells lacking endogenous KRAS and used this system to interrogate single or compound G12D, T50I, D154Q, and E162L mutations. These studies support a key role for the asymmetric lateral assembly of K-Ras in a plasma membrane-distal orientation that promotes the formation of active Ras:Raf complexes in a membrane-proximal conformation. Disease-causing mutations such as T50I are a valuable starting point for illuminating normal Ras function, elucidating mechanisms of disease, and identifying potential therapeutic opportunities for Rasopathy disorders and cancer.
Collapse
Affiliation(s)
- Pan-Yu Chen
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | | | - Max Harris
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | | | - Weijie Wang
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Heidi Carias
- Plexxikon Inc., South San Francisco, California, USA
| | - Brian Mesiona
- Plexxikon Inc., South San Francisco, California, USA
| | | | | | - Gideon Bollag
- Plexxikon Inc., South San Francisco, California, USA
| | - Chao Zhang
- Plexxikon Inc., South San Francisco, California, USA
| | - Ying Zhang
- Plexxikon Inc., South San Francisco, California, USA
| | - Kevin Shannon
- Department of Pediatrics, UCSF, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| |
Collapse
|