1
|
Fontecha MB, Del Rosario Anadón M, Lahitou IMM, Weich N, Bengió R, Moiraghi B, Larripa I, Fundia AF. Exploring the significance of MDM2 gene promoter variants in chronic myeloid leukemia. Leuk Res 2025; 149:107644. [PMID: 39823766 DOI: 10.1016/j.leukres.2025.107644] [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: 08/08/2024] [Revised: 01/02/2025] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) targeting BCR::ABL1 are highly successful in chronic myeloid leukemia (CML). However, extensive interpatient variability in therapeutic responses and resistance supports the need to find new prognostic biomarkers. We have previously reported that TP53 SNP215 variant affects CML risk and clinical outcome. We aimed to evaluate the role of MDM2 genetic variants in CML susceptibility and treatment response to TKIs. We genotyped five MDM2 promoter variants (del1518, SNP309, SNP285, SNP288 and SNP344) in 135 CML patients and 136 healthy individuals. Our study showed that MDM2 variants alone or in combination had no effect on CML susceptibility. The analysis of MDM2 genotypes in relation to patients' clinical parameters revealed that individuals with SNP309 G/G genotypes were at a significantly increased risk of undergoing molecular response failure (p = 0.044). Improved overall survival was also observed for non-responders with the alternative MDM2 del1518 del allele (p = 0.017) as well as for MDM2 del1518-SNP309 combinations with alternative genotypes (p = 0.014). In addition, combinatorial analysis demonstrated that alternative MDM2 SNP309 and TP53 SNP215 genotypes together are associated with faster achievement of MR2 (p = 0.029) and MMR (p = 0.042) in non-responders, suggesting a relationship with a favorable outcome. Overall, our study highlights the influence of MDM2 variants on clinical outcome, supporting that specific genotypes, alone or in combination, underlie the treatment-responsive phenotype.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-mdm2/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Female
- Male
- Middle Aged
- Promoter Regions, Genetic/genetics
- Polymorphism, Single Nucleotide
- Adult
- Aged
- Genotype
- Genetic Predisposition to Disease
- Protein Kinase Inhibitors/therapeutic use
- Prognosis
- Young Adult
- Case-Control Studies
- Aged, 80 and over
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
| | | | | | - Natalia Weich
- Laboratorio de Farmacogenómica, IMEX, CONICET-ANM, Buenos Aires, Argentina
| | - Raquel Bengió
- División Clínica Hematológica, IIHEMA, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Beatriz Moiraghi
- Servicio de Hematología, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Irene Larripa
- Laboratorio de Genética Hematológica, IMEX, CONICET-ANM, Buenos Aires, Argentina
| | - Ariela Freya Fundia
- Laboratorio de Farmacogenómica, IMEX, CONICET-ANM, Buenos Aires, Argentina; Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| |
Collapse
|
2
|
Hsu MT, Willimsky G, Hansmann L, Blankenstein T. T cell receptors specific for an imatinib-induced mutation in BCR-ABL for adoptive T cell therapy. Front Immunol 2025; 16:1518691. [PMID: 39931057 PMCID: PMC11807957 DOI: 10.3389/fimmu.2025.1518691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
BCR-ABL kinase is the major oncogenic driver of chronic myeloid leukemia (CML). Tyrosine kinase inhibitors (TKIs), which are highly potent in targeting BCR-ABL, are currently used as first-line treatment. Although TKIs are effective, drug resistance caused by the emergence of drug-selected secondary mutations in BCR-ABL remains a major problem for relapse, especially in patients with compound mutations. In this study, we aimed to investigate potential neoepitopes derived from mutated BCR-ABL and to generate neoepitope-specific TCRs for adoptive T cell therapy. Two candidate peptides derived from the E255V and the T315I mutation (designated ABL-E255V and ABL-T315I) were selected for study based on their in silico predicted binding affinity to HLA-A2. By immunizing transgenic mice that express a diverse human T cell receptor (TCR) repertoire restricted to HLA-A2, we detected CD8+ T cell responses against the ABL-E255V, but not the ABL-T315I peptide. From immune responding mice, two E255V-specific TCRs were isolated. Human CD8+ T cells were engineered to express the specific TCRs for characterization, in which one TCR was identified as a therapeutic candidate due to its superior avidity and lack of detectable off-target reactivity. Importantly, we demonstrated that the ABL-E255V neoepitope was naturally processed and presented. In summary, our results demonstrate that HLA-A2+ CML cells harboring the E255V mutation can be targeted by specific TCRs, which may benefit patients who are highly resistant to available TKIs due to compound mutations.
Collapse
MESH Headings
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/immunology
- Animals
- Imatinib Mesylate/pharmacology
- Humans
- Mice
- Immunotherapy, Adoptive/methods
- Mutation
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Mice, Transgenic
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- HLA-A2 Antigen/immunology
- Epitopes, T-Lymphocyte/immunology
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Meng-Tung Hsu
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gerald Willimsky
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, partner site Berlin, Berlin, Germany
| | - Leo Hansmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, partner site Berlin, Berlin, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
3
|
Jahanbakhsh K, Ansari-Ahl R, Mashhadi B, Zare M, Samarkhazan NS, Kazemzadeh H, Dehghan G, Dehkordi MF, Gharaghani S, Mahdavi M. DNA binding, and apoptosis-inducing activities of a β-ionone-derived ester in human myeloid leukemia cells: multispectral and molecular dynamic simulation analyses. Sci Rep 2024; 14:27985. [PMID: 39543249 PMCID: PMC11564724 DOI: 10.1038/s41598-024-78690-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: 08/05/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
β-Ionone is the end-ring counterpart of β-carotenoids, which are widely found in fruits and vegetables. Recent studies have illustrated the antimetastatic, anti-proliferative, and apoptosis-inducing activities of β-ionone both in vitro and in vivo. We aimed to explore the anti-cancer potency of β-Ionone-derived ester, (E)-4-(2,6,6-trimethylcyclohex-1-enyl) but-3-en-2-ylpyrazine-2-carboxylate (4-TM.P). The cytotoxic effects of the compound on K562 cells were evaluated by MTT assay. The mechanisms of apoptosis induction were investigated by acridine orange/ethidium bromide (AO/EtBr) double staining, cell cycle analysis, and Annexin V/PI staining. Furthermore, the 4-TM.P-DNA interactions have been thoroughly elucidated by various methods, such as ultraviolet-visible spectroscopy, fluorescence assays, viscosity measurements, molecular docking, and dynamic simulation. The MTT cytotoxicity assay revealed that the growth of K562 cells was inhibited by treatment with β-ionone-derived ester, with an IC50 of 25 ± 5.0 µM at 72 h. Morphological studies revealed the occurrence of apoptosis in treated cells, and G0/G1 cell cycle arrest was observed after treatment of the cells with the IC50 value of the compound. Analyses of multi-spectroscopy and viscosity assays revealed that 4-TM.P binds to DNA in the minor groove mode, which was supported by molecular docking studies. The dynamic stability of the complex was also confirmed using molecular dynamic simulation analyses.
Collapse
Affiliation(s)
- Kamran Jahanbakhsh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Ramin Ansari-Ahl
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Benyamin Mashhadi
- Laboratory of Molecular Biology, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Monireh Zare
- Department of Biochemistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamid Kazemzadeh
- Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Majid Mahdavi
- Laboratory of Molecular Biology, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
4
|
García-Gutiérrez V, Gómez-Casares MT, Xicoy B, Casado-Montero F, Orti G, Giraldo P, Hernández-Boluda JC. Critical review of clinical data and expert-based recommendations for the use of bosutinib in the treatment of chronic myeloid leukemia. Front Oncol 2024; 14:1405467. [PMID: 39252937 PMCID: PMC11381280 DOI: 10.3389/fonc.2024.1405467] [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: 03/22/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Chronic myeloid leukemia (CML), characterized by the presence of the BCR::ABL1 fusion gene, has undergone a transformative shift with the introduction of tyrosine kinase inhibitors (TKIs). The current availability of six different TKIs (imatinib, dasatinib, nilotinib, bosutinib, ponatinib, and asciminib) in clinical practice makes it important to know their efficacy and toxicity profile for treatment optimization. This review examines the latest insights regarding the use of bosutinib in CML treatment. Clinical trials have demonstrated the effectiveness of bosutinib, positioning it as a first-line treatment that can induce sustained molecular responses. Importantly, it can also be effective in patients who have experienced treatment failure or intolerance with prior TKIs, revealing the potential of bosutinib also in second- and later-line settings. Even in the advanced phase of CML, bosutinib has demonstrated its capacity to achieve molecular responses, expanding its usefulness. Real-world evidence studies echo these findings, emphasizing bosutinib's effectiveness in achieving deep molecular responses, maintaining remissions, and serving as an alternative for patients intolerant or resistant to other TKIs as a second-line therapy. Notably, one of the greatest strengths of bosutinib is its favorable safety profile, in particular the low incidence of vascular complications with its use, which is undoubtedly a comparative advantage over other TKIs. In summary, the latest research highlights the versatility of bosutinib in CML treatment and underscores its pivotal role in optimizing patient management in challenging cases. Continuing research and investigation will further establish bosutinib's place in the evolving landscape of CML therapy, offering an alternative for CML patients across different treatment stages.
Collapse
Affiliation(s)
| | - María Teresa Gómez-Casares
- Servicio de Hematología, Hospital Universitario de Gran Canaria Dr. Negrin. Profesor asociado de la ULPGC, Las Palmas de Gran Canaria, Spain
| | - Blanca Xicoy
- Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Felipe Casado-Montero
- Servicio de Hematología y Hemoterapia, Hospital General Universitario de Toledo, Toledo, Spain
| | - Guillermo Orti
- Servicio de Hematologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pilar Giraldo
- Hematologia, Hospital Quironsalud, Fundación ZeroLMC, Zaragoza, Spain
| | | |
Collapse
|
5
|
Vicente ATS, Salvador JAR. PROteolysis-Targeting Chimeras (PROTACs) in leukemia: overview and future perspectives. MedComm (Beijing) 2024; 5:e575. [PMID: 38845697 PMCID: PMC11154823 DOI: 10.1002/mco2.575] [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: 01/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Leukemia is a heterogeneous group of life-threatening malignant disorders of the hematopoietic system. Immunotherapy, radiotherapy, stem cell transplantation, targeted therapy, and chemotherapy are among the approved leukemia treatments. Unfortunately, therapeutic resistance, side effects, relapses, and long-term sequelae occur in a significant proportion of patients and severely compromise the treatment efficacy. The development of novel approaches to improve outcomes is therefore an unmet need. Recently, novel leukemia drug discovery strategies, including targeted protein degradation, have shown potential to advance the field of personalized medicine for leukemia patients. Specifically, PROteolysis-TArgeting Chimeras (PROTACs) are revolutionary compounds that allow the selective degradation of a protein by the ubiquitin-proteasome system. Developed against a wide range of cancer targets, they show promising potential in overcoming many of the drawbacks associated with conventional therapies. Following the exponential growth of antileukemic PROTACs, this article reviews PROTAC-mediated degradation of leukemia-associated targets. Chemical structures, in vitro and in vivo activities, pharmacokinetics, pharmacodynamics, and clinical trials of PROTACs are critically discussed. Furthermore, advantages, challenges, and future perspectives of PROTACs in leukemia are covered, in order to understand the potential that these novel compounds may have as future drugs for leukemia treatment.
Collapse
Affiliation(s)
- André T. S. Vicente
- Laboratory of Pharmaceutical ChemistryFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical ChemistryFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| |
Collapse
|
6
|
Hekmatshoar Y, Rahbar Saadat Y, Ozkan T, Bozkurt S, Karadag Gurel A. Identification of common genes and pathways underlying imatinib and nilotinib treatment in CML: a Bioinformatics Study. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 43:664-684. [PMID: 38117080 DOI: 10.1080/15257770.2023.2296021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Imatinib (IMA) and nilotinib are the first and second generations of BCR-ABL tyrosine kinase inhibitors, which widely applied in chronic myeloid leukemia (CML) treatment. Here we aimed to provide new targets for CML treatment by transcriptome analysis. Microarray data GSE19567 was downloaded and analyzed from Gene Expression Omnibus (GEO) to identify common genes, which are downregulated or upregulated in K562-imatinib and K562-nilotinib treated cells. The differentially expressed genes (DEGs) were assessed, and STRING and Cytoscape were used to create the protein-protein interaction (PPI) network. In imatinib and nilotinib treated groups' comparison, there were common 626 upregulated and 268 downregulated genes, which were differentially expressed. The GO analysis represented the enrichment of DEGs in iron ion binding, protein tyrosine kinase activity, transcription factor activity, ATP binding, sequence-specific DNA binding, cytokine activity, the mitochondrion, sequence-specific DNA binding, plasma membrane and cell-cell adherens junction. KEGG pathway analysis revealed that downregulated DEGs were associated with pathways including microRNAs in cancer and PI3K-Akt signaling pathway. Furthermore, upregulated DEGs were involved in hematopoietic cell lineage, lysosome and chemical carcinogenesis. Among the upregulated genes, MYH9, MYH14, MYL10, MYL7, MYL5, RXRA, CYP1A1, FECH, AKR1C3, ALAD, CAT, CITED2, CPT1A, CYP3A5, CYP3A7, FABP1, HBD, HMBS and PPOX genes were found as hub genes. Moreover, 20 downregulated genes, YARS, AARS, SARS, GARS, CARS, IARS, RRP79, CEBPB, RRP12, UTP14A, PNO1, CCND1, DDX10, MYC, WDR43, CEBPG, DDIT3, VEGFA, PIM1 and TRIB3 were identified as hub genes. These genes have the potential to become target genes for diagnosis and therapy of CML patients.
Collapse
Affiliation(s)
- Yalda Hekmatshoar
- Department of Medical Biology, School of Medicine, Altinbas University, Istanbul, Turkey
| | | | - Tulin Ozkan
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | - Sureyya Bozkurt
- Department of Medical Biology, School of Medicine, Istinye University, Istanbul, Turkey
| | - Aynur Karadag Gurel
- Department of Medical Biology, School of Medicine, Usak University, Usak, Turkey
| |
Collapse
|
7
|
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
|
8
|
Vasileva VY, Khairullina ZM, Sudarikova AV, Chubinskiy-Nadezhdin VI. Role of Calcium-Activated Potassium Channels in Proliferation, Migration and Invasion of Human Chronic Myeloid Leukemia K562 Cells. MEMBRANES 2023; 13:583. [PMID: 37367787 DOI: 10.3390/membranes13060583] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
Calcium-activated potassium channels (KCa) are important participants in calcium signaling pathways due to their ability to be activated by an increase in intracellular free calcium concentration. KCa channels are involved in the regulation of cellular processes in both normal and pathophysiological conditions, including oncotransformation. Previously, using patch-clamp, we registered the KCa currents in the plasma membrane of human chronic myeloid leukemia K562 cells, whose activity was controlled by local Ca2+ entry via mechanosensitive calcium-permeable channels. Here, we performed the molecular and functional identification of KCa channels and have uncovered their role in the proliferation, migration and invasion of K562 cells. Using a combined approach, we identified the functional activity of SK2, SK3 and IK channels in the plasma membrane of the cells. Selective SK and IK channel inhibitors, apamin and TRAM-34, respectively, reduced the proliferative, migratory and invasive capabilities of human myeloid leukemia cells. At the same time, the viability of K562 cells was not affected by KCa channel inhibitors. Ca2+ imaging showed that both SK and IK channel inhibitors affect Ca2+ entry and this could underlie the observed suppression of pathophysiological reactions of K562 cells. Our data imply that SK/IK channel inhibitors could be used to slow down the proliferation and spreading of chronic myeloid leukemia K562 cells that express functionally active KCa channels in the plasma membrane.
Collapse
Affiliation(s)
- Valeria Y Vasileva
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 Saint-Petersburg, Russia
| | - Zuleikha M Khairullina
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 Saint-Petersburg, Russia
| | - Anastasia V Sudarikova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 Saint-Petersburg, Russia
| | | |
Collapse
|