1
|
Zhang H, Song T, Song Y, Hu Z, Soliman MES, Jiang M, Yin F, Yang Z, Wang Z, Zhang Z. A kinase-independent Bcr-Abl function mediating an Hsp70-Bim protein-protein interaction in chronic myeloid leukemia. Int J Biol Macromol 2025; 310:143249. [PMID: 40250683 DOI: 10.1016/j.ijbiomac.2025.143249] [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: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Despite the transformative impact of tyrosine kinase inhibitors (TKIs) on chronic myeloid leukemia (CML), a subset of TKIs-resistant CML cells survives independent of Bcr-Abl kinase activity, forming a persistent therapeutic challenge. In this study, we present the first direct evidence that Bcr-Abl, through its DNA-binding domain (DBD), interacts with the nucleotide-binding domain (NBD) of Hsp70 to mediate the formation of a Bcr-Abl/Hsp70/Bim tri-complex, independent of its kinase function. Using a combination of in vitro biophysical assays-including fluorescent polarization assays (FPAs), isothermal titration calorimetry (ITC), circular dichroism spectroscopy, ATPase activity measurement, and rhodanese aggregation suppression-and cell-based co-immunoprecipitation (Co-IP), we demonstrate that this interaction induces a conformational change in Hsp70 that enhances its affinity for Bim and significantly elevates its ATPase activity. The resulting complex stabilizes oncogenic survival proteins such as AKT and eIF4E, thereby protecting TKIs-resistant CML cells from apoptosis in a Bcr-Abl kinase-independent manner. Importantly, pharmacologic disruption of this complex using the Hsp70/Bim inhibitor S1g-10 or Bcr-Abl PROTAC molecule effectively suppresses TKIs-resistant CML cell proliferation. These findings reveal a novel non-canonical function of Bcr-Abl and provide a new therapeutic strategy for overcoming TKIs-resistance in CML.
Collapse
Affiliation(s)
- Hong Zhang
- Central Hospital of Dalian University of Technology, School of Pharmacy, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Ting Song
- Central Hospital of Dalian University of Technology, School of Pharmacy, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yang Song
- Department of Hematology, Central Hospital of Dalian University of Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zhiyuan Hu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Mahmoud E S Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus 4001, Durban, South Africa
| | - Maojun Jiang
- Central Hospital of Dalian University of Technology, School of Pharmacy, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Fangkui Yin
- Central Hospital of Dalian University of Technology, School of Pharmacy, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zixuan Yang
- School of Pharmacy, ZhongShan College of Dalian Medical University, Dalian, Liaoning 116085, China
| | - Ziqian Wang
- Central Hospital of Dalian University of Technology, School of Pharmacy, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning 116024, China.
| | - Zhichao Zhang
- Central Hospital of Dalian University of Technology, School of Pharmacy, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning 116024, China.
| |
Collapse
|
2
|
Khamidullina AI, Yastrebova MA, Bruter AV, Nuzhina JV, Vorobyeva NE, Khrustaleva AM, Varlamova EA, Tyakht AV, Abramenko IE, Ivanova ES, Zamkova MA, Li J, Lim CU, Chen M, Broude EV, Roninson IB, Shtil AA, Tatarskiy VV. CDK8/19 inhibition attenuates G1 arrest induced by BCR-ABL antagonists and accelerates death of chronic myelogenous leukemia cells. Cell Death Discov 2025; 11:62. [PMID: 39955308 PMCID: PMC11830074 DOI: 10.1038/s41420-025-02339-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
Imatinib mesylate (IM) and other BCR-ABL tyrosine kinase inhibitors (BCR-ABLi) are the mainstay of chronic myelogenous leukemia (CML) treatment. However, activation of circumventing signaling pathways and quiescence may limit BCR-ABLi efficacy. CDK8/19 Mediator kinases have been implicated in the emergence of non-genetic drug resistance. Dissecting the effects of pharmacological CDK8/19 inhibition on CML survival in response to BCR-ABLi, we found that a selective, non-toxic CDK8/19 inhibitor (CDK8/19i) Senexin B (SenB) and other CDK8/19i sensitized K562 cells to different BCR-ABLi via attenuation of cell cycle arrest. In particular, SenB prevented IM-induced upregulation of genes that negatively regulate cell cycle progression. SenB also antagonized IM-activated p27Kip1 elevation thereby diminishing the population of G1-arrested cells. After transient G1 arrest, cells treated with IM + SenB re-entered the S phase, where they were halted and underwent replicative stress. Consequently, the combination of IM and SenB intensified apoptotic cell death, measured by activation of caspase 9 and 3, subsequent cleavage of poly(ADPriboso)polymerase 1, positive Annexin V staining and increase of subG1 fraction. In contrast, IM-treated BCR-ABL-positive KU812 CML cells, which did not induce p27Kip1, readily died regardless of SenB treatment. Thus, CDK8/19i prevent the quiescence-mediated escape from BCR-ABLi-induced apoptosis, suggesting a strategy for avoiding the CML relapse.
Collapse
Affiliation(s)
- Alvina I Khamidullina
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia.
| | - Margarita A Yastrebova
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Alexandra V Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Julia V Nuzhina
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Nadezhda E Vorobyeva
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Anastasia M Khrustaleva
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Ekaterina A Varlamova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Alexander V Tyakht
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Iaroslav E Abramenko
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Ekaterina S Ivanova
- Blokhin National Medical Research Center of Oncology, 24 Kashirskoye shosse, 115522, Moscow, Russia
| | - Maria A Zamkova
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
- Blokhin National Medical Research Center of Oncology, 24 Kashirskoye shosse, 115522, Moscow, Russia
| | - Jing Li
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, SC, 29208, USA
| | - Chang-Uk Lim
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, SC, 29208, USA
| | - Mengqian Chen
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, SC, 29208, USA
| | - Eugenia V Broude
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, SC, 29208, USA
| | - Igor B Roninson
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, SC, 29208, USA
| | - Alexander A Shtil
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
- Blokhin National Medical Research Center of Oncology, 24 Kashirskoye shosse, 115522, Moscow, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia.
| |
Collapse
|
3
|
Selzer AM, Gerlach G, Gonzalez-Areizaga G, Wales TE, Cui SY, Iyer P, Engen JR, Camacho C, Ishima R, Smithgall TE. An SH3-binding allosteric modulator stabilizes the global conformation of the AML-associated Src-family kinase, Hck. J Biol Chem 2025; 301:108088. [PMID: 39675702 PMCID: PMC11786751 DOI: 10.1016/j.jbc.2024.108088] [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: 08/24/2024] [Revised: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
While ATP-site inhibitors for protein-tyrosine kinases are often effective drugs, their clinical utility can be limited by off-target activity and acquired resistance mutations due to the conserved nature of the ATP-binding site. However, combining ATP-site and allosteric kinase inhibitors can overcome these shortcomings in a double-drugging framework. Here we explored the allosteric effects of two pyrimidine diamines, PDA1 and PDA2, on the conformational dynamics and activity of the Src-family tyrosine kinase Hck, a promising drug target for acute myeloid leukemia. Using 1H-15N HSQC NMR, we mapped the binding site for both analogs to the SH3 domain. Despite the shared binding site, PDA1 and PDA2 had opposing effects on near-full-length Hck dynamics by hydrogen-deuterium exchange mass spectrometry, with PDA1 stabilizing and PDA2 disrupting the overall kinase conformation. Kinase activity assays were consistent with these observations, with PDA2 enhancing kinase activity while PDA1 was without effect. Molecular dynamics simulations predicted selective bridging of the kinase domain N-lobe and SH3 domain by PDA1, a mechanism of allosteric stabilization supported by site-directed mutagenesis of N-lobe contact sites. Cellular thermal shift assays confirmed SH3 domain-dependent interaction of PDA1 with WT Hck in myeloid leukemia cells and with a kinase domain gatekeeper mutant (T338M). These results identify PDA1 as a starting point for Src-family kinase allosteric inhibitor development that may work in concert with ATP-site inhibitors to suppress the evolution of resistance.
Collapse
Affiliation(s)
- Ari M Selzer
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gabriella Gerlach
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Giancarlo Gonzalez-Areizaga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas E Wales
- Department of Chemistry and Chemical Biology, College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Stephanie Y Cui
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Prema Iyer
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - John R Engen
- Department of Chemistry and Chemical Biology, College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Carlos Camacho
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rieko Ishima
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Thomas E Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
4
|
Warfvinge R, Geironson Ulfsson L, Dhapola P, Safi F, Sommarin M, Soneji S, Hjorth-Hansen H, Mustjoki S, Richter J, Thakur RK, Karlsson G. Single-cell multiomics analysis of chronic myeloid leukemia links cellular heterogeneity to therapy response. eLife 2024; 12:RP92074. [PMID: 39503729 PMCID: PMC11540304 DOI: 10.7554/elife.92074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
The advent of tyrosine kinase inhibitors (TKIs) as treatment of chronic myeloid leukemia (CML) is a paradigm in molecularly targeted cancer therapy. Nonetheless, TKI-insensitive leukemia stem cells (LSCs) persist in most patients even after years of treatment and are imperative for disease progression as well as recurrence during treatment-free remission (TFR). Here, we have generated high-resolution single-cell multiomics maps from CML patients at diagnosis, retrospectively stratified by BCR::ABL1IS (%) following 12 months of TKI therapy. Simultaneous measurement of global gene expression profiles together with >40 surface markers from the same cells revealed that each patient harbored a unique composition of stem and progenitor cells at diagnosis. The patients with treatment failure after 12 months of therapy had a markedly higher abundance of molecularly defined primitive cells at diagnosis compared to the optimal responders. The multiomic feature landscape enabled visualization of the primitive fraction as a mixture of molecularly distinct BCR::ABL1+ LSCs and BCR::ABL1-hematopoietic stem cells (HSCs) in variable ratio across patients, and guided their prospective isolation by a combination of CD26 and CD35 cell surface markers. We for the first time show that BCR::ABL1+ LSCs and BCR::ABL1- HSCs can be distinctly separated as CD26+CD35- and CD26-CD35+, respectively. In addition, we found the ratio of LSC/HSC to be higher in patients with prospective treatment failure compared to optimal responders, at diagnosis as well as following 3 months of TKI therapy. Collectively, this data builds a framework for understanding therapy response and adapting treatment by devising strategies to extinguish or suppress TKI-insensitive LSCs.
Collapse
Affiliation(s)
- Rebecca Warfvinge
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | | | - Parashar Dhapola
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Fatemeh Safi
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Mikael Sommarin
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Shamit Soneji
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Henrik Hjorth-Hansen
- Department of Hematology, St Olavs HospitalTrondheimNorway
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Satu Mustjoki
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of HelsinkiHelsinkiFinland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer CenterTrondheimNorway
- iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Johan Richter
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund UniversityLundSweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University HospitalLundSweden
| | - Ram Krishna Thakur
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Göran Karlsson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| |
Collapse
|
5
|
Cruz-Rodriguez N, Tang H, Bateman B, Tang W, Deininger M. BCR::ABL1 Proteolysis-targeting chimeras (PROTACs): The new frontier in the treatment of Ph + leukemias? Leukemia 2024; 38:1885-1893. [PMID: 39098922 PMCID: PMC11569815 DOI: 10.1038/s41375-024-02365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
BCR::ABL1 tyrosine kinase inhibitors (TKIs) have turned chronic myeloid leukemia (CML) from a lethal condition into a chronic ailment. With optimal management, the survival of CML patients diagnosed in the chronic phase is approaching that of age-matched controls. However, only one-third of patients can discontinue TKIs and enter a state of functional cure termed treatment-free remission (TFR), while the remainder require life-long TKI therapy to avoid the recurrence of active leukemia. Approximately 10% of patients exhibit primary or acquired TKI resistance and eventually progress to the blast phase. It is thought that recurrence after attempted TFR originates from CML stem cells (LSCs) surviving despite continued suppression of BCR::ABL1 kinase. Although kinase activity is indispensable for induction of overt CML, kinase-independent scaffold functions of BCR::ABL1 are known to contribute to leukemogenesis, raising the intriguing but as yet hypothetical possibility, that degradation of BCR::ABL1 protein may accomplish what TKIs fail to achieve - eliminate residual LSCs to turn functional into real cures. The advent of BCR::ABL1 proteolysis targeting chimeras (PROTACs), heterobifunctional molecules linking a TKI-based warhead to an E3 ligase recruiter, has moved clinical protein degradation into the realm of the possible. Here we examine the molecular rationale as well as pros and cons of degrading BCR::ABL1 protein. We review reported BCR::ABL1 PROTACs, point out limitations of available data and compounds and suggest directions for future research. Ultimately, clinical testing of a potent and specific BCR::ABL1 degrader will be required to determine the efficacy and tolerability of this approach.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Proteolysis
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Drug Resistance, Neoplasm
- Proto-Oncogene Proteins c-abl/metabolism
- Animals
- Proteolysis Targeting Chimera
Collapse
Affiliation(s)
| | - Hua Tang
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Weiping Tang
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Deininger
- Versiti Blood Research Institute, Milwaukee, WI, USA.
- Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
6
|
Hochhaus A, Réa D, Boquimpani C, Minami Y, Cortes JE, Hughes TP, Apperley JF, Lomaia E, Voloshin S, Turkina A, Kim DW, Abdo A, Fogliatto LM, le Coutre P, Sasaki K, Kim DDH, Saussele S, Annunziata M, Chaudhri N, Chee L, García-Gutiérrez V, Kapoor S, Allepuz A, Quenet S, Bédoucha V, Mauro MJ. Asciminib vs bosutinib in chronic-phase chronic myeloid leukemia previously treated with at least two tyrosine kinase inhibitors: longer-term follow-up of ASCEMBL. Leukemia 2023; 37:617-626. [PMID: 36717654 PMCID: PMC9991909 DOI: 10.1038/s41375-023-01829-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023]
Abstract
Asciminib, the first BCR::ABL1 inhibitor that Specifically Targets the ABL Myristoyl Pocket (STAMP), is approved worldwide for the treatment of adults with Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase (CML-CP) treated with ≥2 prior tyrosine kinase inhibitors (TKIs). In ASCEMBL, patients with CML-CP treated with ≥2 prior TKIs were randomized (stratified by baseline major cytogenetic response [MCyR]) 2:1 to asciminib 40 mg twice daily or bosutinib 500 mg once daily. Consistent with previously published primary analysis results, after a median follow-up of 2.3 years, asciminib continued to demonstrate superior efficacy and better safety and tolerability than bosutinib. The major molecular response (MMR) rate at week 96 (key secondary endpoint) was 37.6% with asciminib vs 15.8% with bosutinib; the MMR rate difference between the arms, after adjusting for baseline MCyR, was 21.7% (95% CI, 10.53-32.95; two-sided p = 0.001). Fewer grade ≥3 adverse events (AEs) (56.4% vs 68.4%) and AEs leading to treatment discontinuation (7.7% vs 26.3%) occurred with asciminib than with bosutinib. A higher proportion of patients on asciminib than bosutinib remained on treatment and continued to derive benefit over time, supporting asciminib as a standard of care for patients with CML-CP previously treated with ≥2 TKIs.
Collapse
Affiliation(s)
| | - Delphine Réa
- Adult Hematology and INSERM CIC1427, Hôpital Saint-Louis, Paris, France
| | - Carla Boquimpani
- HEMORIO, State Institute of Hematology Arthur de Siquiera Cavalcanti, Rio de Janeiro, Brazil.,Oncoclínica Centro de Tratamento Oncológico, Rio de Janeiro, RJ, Brazil
| | - Yosuke Minami
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Timothy P Hughes
- South Australian Health and Medical Research Institute and University of Adelaide, Adelaide, SA, Australia
| | | | - Elza Lomaia
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Sergey Voloshin
- Russian Research Institute of Hematology and Transfusiology, St. Petersburg, Russia
| | - Anna Turkina
- National Medical Research Center for Hematology, Moscow, Russia
| | - Dong-Wook Kim
- Uijeongbu Eulji Medical Center, Geumo-dong, Uijeongbu-si, South Korea
| | - Andre Abdo
- Instituto do Câncer do Estado de São Paulo (ICESPSP), São Paulo, Brazil
| | | | | | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dennis Dong Hwan Kim
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Susanne Saussele
- III. Medizinische Klinik, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany
| | | | - Naeem Chaudhri
- King Faisal Specialist Hospital & Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Lynette Chee
- Peter MacCallum Cancer Center and The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Valentin García-Gutiérrez
- Servicio de Hematología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Shruti Kapoor
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | | | | | |
Collapse
|
7
|
Rudich A, Garzon R, Dorrance A. Non-Coding RNAs Are Implicit in Chronic Myeloid Leukemia Therapy Resistance. Int J Mol Sci 2022; 23:ijms232012271. [PMID: 36293127 PMCID: PMC9603161 DOI: 10.3390/ijms232012271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm initiated by the presence of the fusion gene BCR::ABL1. The development of tyrosine kinase inhibitors (TKIs) highly specific to p210BCR-ABL1, the constitutively active tyrosine kinase encoded by BCR::ABL1, has greatly improved the prognosis for CML patients. Now, the survival rate of CML nearly parallels that of age matched controls. However, therapy resistance remains a persistent problem in the pursuit of a cure. TKI resistance can be attributed to both BCR::ABL1 dependent and independent mechanisms. Recently, the role of non-coding RNAs (ncRNAs) has been increasingly explored due to their frequent dysregulation in a variety of malignancies. Specifically, microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs) have been shown to contribute to the development and progression of therapy resistance in CML. Since each ncRNA exhibits multiple functions and is capable of controlling gene expression, they exert their effect on CML resistance through a diverse set of mechanisms and pathways. In most cases ncRNAs with tumor suppressing functions are silenced in CML, while those with oncogenic properties are overexpressed. Here, we discuss the relevance of many aberrantly expressed ncRNAs and their effect on therapy resistance in CML.
Collapse
MESH Headings
- Humans
- Fusion Proteins, bcr-abl
- RNA, Circular
- RNA, Long Noncoding/genetics
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- MicroRNAs/genetics
- MicroRNAs/pharmacology
Collapse
|
8
|
Dey S, Basu S, Shah S, Bhattacharyya D, Gupta PP, Acharjee M, Roychoudhury S, Nath S. Deep sequencing reveals the spectrum of BCR-ABL1 mutations upon front-line therapy resistance in chronic myeloid leukemia: An Eastern-Indian cohort study. Cancer Treat Res Commun 2022; 33:100635. [PMID: 36155130 DOI: 10.1016/j.ctarc.2022.100635] [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: 08/17/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
The course of clinical management in chronic myeloid leukemia (CML) often faces a road-block in the form of front-line (imatinib) therapy resistance. Subsequently, several hotspot mutations were clinically validated in the kinase domain (KD) of BCR-ABL1, in deterring imatinib sensitivity and further, made targeted by next-generation tyrosine-kinase-inhibitor (TKI) drugs. Identifying KD mutations, occurring even at low frequencies, became pertinent here. Globally, cohorts from different origins were tested and the mutational spectra were mapped to categorize clinical management as well as related pathological features of CML. Moreover, targeted deep sequencing could reveal the mutational landscape more efficiently than the less sensitive Sanger sequencing method. However, no such efforts were reported from Eastern Indian cohorts of imatinib-resistant CML-sufferers. This study assessed a prospective study cohort of imatinib-resistant CML cases from Eastern India. Following dissecting the molecular and clinical parameters, the mutational spectrum was comparatively examined using conventional Sanger and next-generation deep sequencing method. This cohort showed a prevalence of e14a2-p210 variant of BCR-ABL1 and acquired resistance against imatinib, while the disease was mostly confined in its chronic phase. Together with a few common hotspot mutations identified in this cohort, deep sequencing revealed cases with a candidate mutation, otherwise undetermined by Sanger method. Also, cases with a second low frequency mutation were identified upon applying deep sequencing. Along with highlighting a few aspects of CML biology employing an Eastern-Indian cohort, this data could mark the immense importance of deep sequencing to contribute in the clinical management of CML upon front-line therapy resistance.
Collapse
MESH Headings
- Humans
- Cohort Studies
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- High-Throughput Nucleotide Sequencing/methods
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mutation
- Prospective Studies
- India
Collapse
Affiliation(s)
- Samya Dey
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India
| | - Soumi Basu
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India; Department of Molecular Genetics and Cell Biology, University of Nebraska Medical Center, Omaha, USA
| | - Shahena Shah
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India
| | - Debmalya Bhattacharyya
- Department of Hemato-oncology, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India
| | - Partha Pratim Gupta
- Department of Hemato-oncology, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India
| | - Mahasweta Acharjee
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India
| | - Susanta Roychoudhury
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India; CSIR-Indian Institute of Chemical Biology, CN-06, CN Block, Sector V, Kolkata 700091, India
| | - Somsubhra Nath
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700063, India; Institute of Health Sciences, Presidency University, Plot No. DG/02/02, Premises No. 14-0358, Action Area-ID, New Town, Kolkata, 700156, India.
| |
Collapse
|
9
|
Boni C, Bonifacio M, Vezzalini M, Scaffidi L, Tomasello L, Parker LL, Boscarino D, Paladin D, Krampera M, Sorio C. Successful Preservation of Native BCR::ABL1 in Chronic Myeloid Leukemia Primary Leukocytes Reveals a Reduced Kinase Activity. Front Oncol 2022; 12:904510. [PMID: 35756686 PMCID: PMC9216732 DOI: 10.3389/fonc.2022.904510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by the acquisition of t(9;22) generating the fusion tyrosine kinase BCR::ABL1. However, despite the crucial role of this protein in the dysregulation of numerous signal transduction pathways, a direct measure of BCR::ABL1 kinase activity in chronic phase (CP) CML was never accomplished due to intense degradative activity present in mature leukocytes. Therefore, we developed a procedure suitable to preserve BCR::ABL1 protein under non-denaturing, neutral pH conditions in primary, chronic phase (CP)-CML samples. As a result, specific kinase activity was detected utilizing a biotinylated peptide substrate highly selective for c-ABL1. Furthermore, through this approach, BCR::ABL1 kinase activity was barely detectable in CP-CML compared to Ph+ acute lymphoblastic leukemia primary samples, where kinase activity is comparable to those measured in Ph+ cell lines. These in vitro findings provide the first direct measure of BCR::ABL1 kinase activity in primary CP-CML and reveal the presence of a still uncharacterized inhibitory mechanism that maintains BCR::ABL1 in a low activity state in CP-CML despite its overexpression.
Collapse
Affiliation(s)
- Christian Boni
- Department of Medicine, General Pathology Section, University of Verona, Verona, Italy
| | | | - Marzia Vezzalini
- Department of Medicine, General Pathology Section, University of Verona, Verona, Italy
| | - Luigi Scaffidi
- Department of Medicine, Hematology Section, University of Verona, Verona, Italy
| | - Luisa Tomasello
- Department of Medicine, General Pathology Section, University of Verona, Verona, Italy
| | - Laurie L Parker
- Department of Biochemistry, Molecular Biology and Biophysics, Masonic Cancer Center, University of Minnesota, Minneapolis, United States
| | | | | | - Mauro Krampera
- Department of Medicine, Hematology Section, University of Verona, Verona, Italy
| | - Claudio Sorio
- Department of Medicine, General Pathology Section, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Secondary chronic myeloid leukemia in a patient with CALR and ASXL1-mutated primary myelofibrosis. Int J Hematol 2022; 116:442-445. [DOI: 10.1007/s12185-022-03331-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 10/18/2022]
|
11
|
Benchikh S, Bousfiha A, El Hamouchi A, Soro SGC, Malki A, Nassereddine S. Chronic myeloid leukemia: cytogenetics and molecular biology’s part in the comprehension and management of the pathology and treatment evolution. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00248-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Background
Chronic myelogenous leukemia (CML) is a type of blood cancer that affects hematopoietic stem cells and is often characterized by the presence of the Philadelphia chromosome. The Philadelphia chromosome encodes for a protein with high tyrosine kinase activity which acts as a tumorigenic factor.
Main body
This review article reports an update on the pathophysiology of CML and highlights the role of cytogenetic and molecular biology in screening, diagnosis, therapeutic monitoring as well as evaluating patients’ response to treatment. Additionally, these genetic tests allow identifying additional chromosomal abnormalities (ACA) and BCR-ABL tyrosine kinase domain mutations in intolerant or resistant patients. Thus, therapeutic advances have enabled this pathology to become manageable and almost curable in its clinical course. The scientific literature search used in the synthesis of this paper was carried out in the PubMed database, and the figures were generated using online software named BioRender.
Conclusion
The role of cytogenetic and molecular biology is crucial for the diagnosis and medical monitoring of patients. In-depth knowledge of molecular mechanisms of the BCR-ABL kinase facilitated the development of new targeted therapies that have improved the vital prognosis in patients. However, the emergence of ACA and new mutations resistant to tyrosine kinase inhibitors constitutes a real challenge in the quest for adequate therapy.
Collapse
|
12
|
Zhao H, Pomicter AD, Eiring AM, Franzini A, Ahmann J, Hwang JY, Senina A, Helton B, Iyer S, Yan D, Khorashad JS, Zabriskie MS, Agarwal A, Redwine HM, Bowler AD, Clair PM, McWeeney SK, Druker BJ, Tyner JW, Stirewalt DL, Oehler VG, Varambally S, Berrett KC, Vahrenkamp JM, Gertz J, Varley KE, Radich JP, Deininger MW. MS4A3 promotes differentiation in chronic myeloid leukemia by enhancing common β-chain cytokine receptor endocytosis. Blood 2022; 139:761-778. [PMID: 34780648 PMCID: PMC8814676 DOI: 10.1182/blood.2021011802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 10/27/2021] [Indexed: 02/05/2023] Open
Abstract
The chronic phase of chronic myeloid leukemia (CP-CML) is characterized by the excessive production of maturating myeloid cells. As CML stem/progenitor cells (LSPCs) are poised to cycle and differentiate, LSPCs must balance conservation and differentiation to avoid exhaustion, similar to normal hematopoiesis under stress. Since BCR-ABL1 tyrosine kinase inhibitors (TKIs) eliminate differentiating cells but spare BCR-ABL1-independent LSPCs, understanding the mechanisms that regulate LSPC differentiation may inform strategies to eliminate LSPCs. Upon performing a meta-analysis of published CML transcriptomes, we discovered that low expression of the MS4A3 transmembrane protein is a universal characteristic of LSPC quiescence, BCR-ABL1 independence, and transformation to blast phase (BP). Several mechanisms are involved in suppressing MS4A3, including aberrant methylation and a MECOM-C/EBPε axis. Contrary to previous reports, we find that MS4A3 does not function as a G1/S phase inhibitor but promotes endocytosis of common β-chain (βc) cytokine receptors upon GM-CSF/IL-3 stimulation, enhancing downstream signaling and cellular differentiation. This suggests that LSPCs downregulate MS4A3 to evade βc cytokine-induced differentiation and maintain a more primitive, TKI-insensitive state. Accordingly, knockdown (KD) or deletion of MS4A3/Ms4a3 promotes TKI resistance and survival of CML cells ex vivo and enhances leukemogenesis in vivo, while targeted delivery of exogenous MS4A3 protein promotes differentiation. These data support a model in which MS4A3 governs response to differentiating myeloid cytokines, providing a unifying mechanism for the differentiation block characteristic of CML quiescence and BP-CML. Promoting MS4A3 reexpression or delivery of ectopic MS4A3 may help eliminate LSPCs in vivo.
Collapse
MESH Headings
- Animals
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Down-Regulation
- Endocytosis
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Receptors, Cytokine/metabolism
- Transcriptome
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Helong Zhao
- Versiti Blood Research Institute, Milwaukee, WI
- Medical College of Wisconsin, Milwaukee, WI
- Division of Hematology and Hematologic Malignancies and
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | | | | | - Anca Franzini
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Jonathan Ahmann
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Jae-Yeon Hwang
- Department of Oncological Sciences, The University of Utah, Salt Lake City, UT
| | - Anna Senina
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Bret Helton
- Department of Chemistry, University of Washington, Seattle, WA
| | - Siddharth Iyer
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Dongqing Yan
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Jamshid S Khorashad
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | | | - Anupriya Agarwal
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Hannah M Redwine
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Amber D Bowler
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Phillip M Clair
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Shannon K McWeeney
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Brian J Druker
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Jeffrey W Tyner
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | | | | | | | | | | | - Jason Gertz
- Department of Oncological Sciences, The University of Utah, Salt Lake City, UT
| | - Katherine E Varley
- Department of Oncological Sciences, The University of Utah, Salt Lake City, UT
| | | | - Michael W Deininger
- Versiti Blood Research Institute, Milwaukee, WI
- Medical College of Wisconsin, Milwaukee, WI
- Division of Hematology and Hematologic Malignancies and
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| |
Collapse
|
13
|
Hochhaus A. TKI discontinuation in CML: how do we make more patients eligible? How do we increase the chances of a successful treatment-free remission? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:106-112. [PMID: 34889388 PMCID: PMC8791110 DOI: 10.1182/hematology.2021000238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Treatment-free remission (TFR) is a new and significant goal of chronic myeloid leukemia management. TFR should be considered for patients in stable deep molecular response (DMR) after careful discussion in the shared decision-making process. Second-generation tyrosine kinase inhibitors (TKIs) improve the speed of response and the incidence of DMR. Treatment may be changed to a more active TKI to improve the depth of response in selected patients who have not reached DMR. Stem cell persistence is associated with active immune surveillance and activation of BCR-ABL1-independent pathways, eg, STAT3, JAK1/2, and BCL2. Ongoing studies aim to prove the efficacy of maintenance therapies targeting these pathways after TKI discontinuation.
Collapse
Affiliation(s)
- Andreas Hochhaus
- Correspondence Andreas Hochhaus, Klinik für Innere Medizin II, Universitätsklinikum Jena, Am Klinikum 1, 07740 Jena, Germany; e-mail:
| |
Collapse
|
14
|
Resistance to Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia-From Molecular Mechanisms to Clinical Relevance. Cancers (Basel) 2021; 13:cancers13194820. [PMID: 34638304 PMCID: PMC8508378 DOI: 10.3390/cancers13194820] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Chronic myeloid leukemia (CML) is a myeloproliferative neoplasia associated with a molecular alteration, the fusion gene BCR-ABL1, that encodes the tyrosine kinase oncoprotein BCR-ABL1. This led to the development of tyrosine kinase inhibitors (TKI), with Imatinib being the first TKI approved. Although the vast majority of CML patients respond to Imatinib, resistance to this targeted therapy contributes to therapeutic failure and relapse. Here we review the molecular mechanisms and other factors (e.g., patient adherence) involved in TKI resistance, the methodologies to access these mechanisms, and the possible therapeutic approaches to circumvent TKI resistance in CML. Abstract Resistance to targeted therapies is a complex and multifactorial process that culminates in the selection of a cancer clone with the ability to evade treatment. Chronic myeloid leukemia (CML) was the first malignancy recognized to be associated with a genetic alteration, the t(9;22)(q34;q11). This translocation originates the BCR-ABL1 fusion gene, encoding the cytoplasmic chimeric BCR-ABL1 protein that displays an abnormally high tyrosine kinase activity. Although the vast majority of patients with CML respond to Imatinib, a tyrosine kinase inhibitor (TKI), resistance might occur either de novo or during treatment. In CML, the TKI resistance mechanisms are usually subdivided into BCR-ABL1-dependent and independent mechanisms. Furthermore, patients’ compliance/adherence to therapy is critical to CML management. Techniques with enhanced sensitivity like NGS and dPCR, the use of artificial intelligence (AI) techniques, and the development of mathematical modeling and computational prediction methods could reveal the underlying mechanisms of drug resistance and facilitate the design of more effective treatment strategies for improving drug efficacy in CML patients. Here we review the molecular mechanisms and other factors involved in resistance to TKIs in CML and the new methodologies to access these mechanisms, and the therapeutic approaches to circumvent TKI resistance.
Collapse
|
15
|
Osman AEG, Deininger MW. Chronic Myeloid Leukemia: Modern therapies, current challenges and future directions. Blood Rev 2021; 49:100825. [PMID: 33773846 DOI: 10.1016/j.blre.2021.100825] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm caused by a reciprocal translocation [t(9;22)(q34;q11.2)] that leads to the fusion of ABL1 gene sequences (9q34) downstream of BCR gene sequences (22q11) and is cytogenetically visible as Philadelphia chromosome (Ph). The resulting BCR/ABL1 chimeric protein is a constitutively active tyrosine kinase that activates multiple signaling pathways, which collectively lead to malignant transformation. During the early (chronic) phase of CML (CP-CML), the myeloid cell compartment is expanded, but differentiation is maintained. Without effective therapy, CP-CML invariably progresses to blast phase (BP-CML), an acute leukemia of myeloid or lymphoid phenotype. The development of BCR-AB1 tyrosine kinase inhibitors (TKIs) revolutionized the treatment of CML and ignited the start of a new era in oncology. With three generations of BCR/ABL1 TKIs approved today, the majority of CML patients enjoy long term remissions and near normal life expectancy. However, only a minority of patients maintain remission after TKI discontinuation, a status termed treatment free remission (TFR). Unfortunately, 5-10% of patients fail TKIs due to resistance and are at risk of progression to BP-CML, which is curable only with hematopoietic stem cell transplantation. Overcoming TKI resistance, improving the prognosis of BP-CML and improving the rates of TFR are areas of active research in CML.
Collapse
Affiliation(s)
- Afaf E G Osman
- Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Michael W Deininger
- Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Single-Strand Annealing in Cancer. Int J Mol Sci 2021; 22:ijms22042167. [PMID: 33671579 PMCID: PMC7926775 DOI: 10.3390/ijms22042167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/23/2022] Open
Abstract
DNA double-strand breaks (DSBs) are among the most serious forms of DNA damage. In humans, DSBs are repaired mainly by non-homologous end joining (NHEJ) and homologous recombination repair (HRR). Single-strand annealing (SSA), another DSB repair system, uses homologous repeats flanking a DSB to join DNA ends and is error-prone, as it removes DNA fragments between repeats along with one repeat. Many DNA deletions observed in cancer cells display homology at breakpoint junctions, suggesting the involvement of SSA. When multiple DSBs occur in different chromosomes, SSA may result in chromosomal translocations, essential in the pathogenesis of many cancers. Inhibition of RAD52 (RAD52 Homolog, DNA Repair Protein), the master regulator of SSA, results in decreased proliferation of BRCA1/2 (BRCA1/2 DNA Repair Associated)-deficient cells, occurring in many hereditary breast and ovarian cancer cases. Therefore, RAD52 may be targeted in synthetic lethality in cancer. SSA may modulate the response to platinum-based anticancer drugs and radiation. SSA may increase the efficacy of the CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 (CRISPR associated 9) genome editing and reduce its off-target effect. Several basic problems associated with SSA, including its evolutionary role, interplay with HRR and NHEJ and should be addressed to better understand its role in cancer pathogenesis and therapy.
Collapse
|