1
|
Tyagi S, Singh A, Sharma N, Chaturvedi R, Kushwaha HR. Insights into existing and futuristic treatment approach for chronic myeloid leukaemia. Indian J Med Res 2024; 159:455-467. [PMID: 39382408 PMCID: PMC11463244 DOI: 10.25259/ijmr_1716_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Indexed: 10/10/2024] Open
Abstract
Oncogenes play a crucial part in human cancer development, and when particular drugs obstruct the proteins produced by these oncogenes, the tumoural process can be ceased. For instance, in chronic myeloid leukaemia (CML), all pathological traits are associated with a single oncogene, BCR-ABL1. CML is a triphasic cancerous disorder of haematopoietic stem cells, marked by a balanced translocation between chromosomes 9 and 22, leading to the genesis of a Philadelphia chromosome encompassing the BCR-ABL1 fusion gene. This fusion oncogene further produces a constitutive active tyrosine kinase protein, enhancing the downstream signalling pathways and constitutes cancer. The treatment for CML has been entirely altered from chemotherapy and immunotherapy to targeted therapy with the emergence of tyrosine kinase inhibitors (TKIs) which inhibit BCR-ABL1 kinase activity. However, the inhibitory mechanism of TKIs is constrained by BCR-ABL1 dependent and independent resistance mechanisms, prompting the exploration of novel therapeutics through extensive clinical trials to develop next-generation drugs with enhanced potency. The persistent challenges posed by CML have motivated researchers to seek innovative strategies for its eradication, such as the application of the genome editing tool CRISPR/Cas9. This review provides insights into existing CML diagnoses, treatment modalities, resistance mechanisms, drugs under trial phases and new potential therapeutic drugs. Furthermore, the review looks ahead to a visionary perspective wherein the CRISPR/Cas9 approach holds the potential to evolve into a prospective curative measure for CML.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Protein Kinase Inhibitors/therapeutic use
- Gene Editing
- Drug Resistance, Neoplasm/genetics
- CRISPR-Cas Systems/genetics
Collapse
Affiliation(s)
- Sourabh Tyagi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Anu Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Naveen Sharma
- Biomedical Informatics Division, Indian Council of Medical Research, New Delhi, India
| | - Rupesh Chaturvedi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Hemant Ritturaj Kushwaha
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
2
|
Tezcanli Kaymaz B, Gumus N, Celik B, Alcitepe İ, Biray Avci C, Aktan C. Ponatinib and STAT5 Inhibitor Pimozide Combined Synergistic Treatment Applications Potentially Overcome Drug Resistance via Regulating the Cytokine Expressional Network in Chronic Myeloid Leukemia Cells. J Interferon Cytokine Res 2024; 44:178-189. [PMID: 38579140 DOI: 10.1089/jir.2023.0170] [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] [Indexed: 04/07/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative hematological disease characterized by the chimeric breakpoint-cluster region/Abelson kinase1 (BCR::ABL1) oncoprotein; playing a pivotal role in CML molecular pathology, diagnosis, treatment, and possible resistance arising from the success and tolerance of tyrosine kinase inhibitor (TKI)-based therapy. The transcription factor STAT5 constitutive signaling, which is influenced by the cytokine signaling network, triggers BCR::ABL1-based CML pathogenesis and is also relevant to acquired TKI resistance. The unsuccessful therapeutic approaches targeting BCR::ABL1, in particular third-line therapy with ponatinib, still need to be further developed with alternative combination strategies to overcome drug resistance. As treatment with the STAT5 inhibitor pimozide in combination with ponatinib resulted in an efficient and synergistic therapeutic approach in TKI-resistant CML cells, this study focused on identifying the underlying amplification of ponatinib response mechanisms by determining different cytokine expression profiles in parental and ponatinib-resistant CML cells, in vitro. The results showed that expression of interleukin (IL) 1B, IL9, and IL12A-B was increased by 2-fold, while IL18 was downregulated by 2-fold in the ponatinib-resistant cells compared to sensitive ones. Importantly, ponatinib treatment upregulated the expression of 21 of the 23 interferon and IL genes in the ponatinib-resistant cells, while treatment with pimozide or a combination dose resulted in a reduction in the expression of 19 different cytokine genes, such as for example, inflammatory cytokines, IL1A-B and IL6 or cytokine genes associated with supporting tumor progression, leukemia stem cell growth or poor survival, such as IL3, IL8, IL9, IL10, IL12, or IL15. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis results showed that the genes were mainly enriched in the regulation of receptor signaling through the Janus kinase/signal transducer and activator of transcription pathway, cytokine-cytokine receptor interaction, and hematopoietic cell lineage. Protein-protein interaction analysis showed that IL2, IL6, IL15, IFNG, and others appeared in the top lists of pathways, indicating their high centrality and importance in the network. Therefore, pimozide could be a promising agent to support TKI therapies in ponatinib resistance. This research would help to clarify the role of cytokines in ponatinib resistance and advance the development of new therapeutics to utilize the STAT5 inhibitor pimozide in combination with TKIs.
Collapse
MESH Headings
- Humans
- Pimozide/pharmacology
- Pimozide/therapeutic use
- Cytokines/metabolism
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/metabolism
- Interleukin-15/metabolism
- Interleukin-15/therapeutic use
- Interleukin-6/metabolism
- Interleukin-9/metabolism
- Interleukin-9/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Imidazoles
- Pyridazines
Collapse
Affiliation(s)
| | - Nurcan Gumus
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Besne Celik
- Department of Medical Biology, Ege University Medical School, Izmir, Turkiye
| | - İlayda Alcitepe
- Department of Medical Biology, Ege University Medical School, Izmir, Turkiye
| | - Cigir Biray Avci
- Department of Medical Biology, Ege University Medical School, Izmir, Turkiye
| | - Cagdas Aktan
- Department of Medical Biology, Beykent University Medical School, Istanbul, Turkiye
- Department of Medical Biology, Bandirma Onyedi Eylul University Medical School, Balikesir, Turkiye
| |
Collapse
|
3
|
Cuesta-Casanovas L, Delgado-Martínez J, Cornet-Masana JM, Carbó JM, Banús-Mulet A, Guijarro F, Esteve J, Risueño RM. Prolactin receptor signaling induces acquisition of chemoresistance and reduces clonogenicity in acute myeloid leukemia. Cancer Cell Int 2023; 23:97. [PMID: 37208719 DOI: 10.1186/s12935-023-02944-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/11/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Development of precision medicine requires the identification of easily detectable and druggable biomarkers. Despite recent targeted drug approvals, prognosis of acute myeloid leukemia (AML) patients needs to be greatly improved, as relapse and refractory disease are still difficult to manage. Thus, new therapeutic approaches are needed. Based on in silico-generated preliminary data and the literature, the role of the prolactin (PRL)-mediated signaling was interrogated in AML. METHODS Protein expression and cell viability were determined by flow cytometry. Repopulation capacity was studied in murine xenotransplantation assays. Gene expression was measured by qPCR and luciferase-reporters. SA-β-Gal staining was used as a senescence marker. RESULTS The prolactin receptor (PRLR) was upregulated in AML cells, as compared to their healthy counterpart. The genetic and molecular inhibition of this receptor reduced the colony-forming potential. Disruption of the PRLR signaling, either using a mutant PRL or a dominant-negative isoform of PRLR, reduced the leukemia burden in vivo, in xenotransplantation assays. The expression levels of PRLR directly correlated with resistance to cytarabine. Indeed, acquired cytarabine resistance was accompanied with the induction of PRLR surface expression. The signaling associated to PRLR in AML was mainly mediated by Stat5, in contrast to the residual function of Stat3. In concordance, Stat5 mRNA was significantly overexpressed at mRNA levels in relapse AML samples. A senescence-like phenotype, measured by SA-β-gal staining, was induced upon enforced expression of PRLR in AML cells, partially dependent on ATR. Similar to the previously described chemoresistance-induced senescence in AML, no cell cycle arrest was observed. Additionally, the therapeutic potential of PRLR in AML was genetically validated. CONCLUSIONS These results support the role of PRLR as a therapeutic target for AML and the further development of drug discovery programs searching for specific PRLR inhibitors.
Collapse
Affiliation(s)
- Laia Cuesta-Casanovas
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
- Faculty of Biosciences, Autonomous University of Barcelona, Barcelona, Spain
| | - Jennifer Delgado-Martínez
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
- Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Josep M Cornet-Masana
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
| | - José M Carbó
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
| | - Antònia Banús-Mulet
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
| | - Francesca Guijarro
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
- Department of Hematology, Hospital Clínic, Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Esteve
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
- Department of Hematology, Hospital Clínic, Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ruth M Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-GTP, Crta Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
4
|
Standing D, Feess E, Kodiyalam S, Kuehn M, Hamel Z, Johnson J, Thomas SM, Anant S. The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy. Cancers (Basel) 2023; 15:cancers15092485. [PMID: 37173951 PMCID: PMC10177275 DOI: 10.3390/cancers15092485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Ovarian cancer (OvCa) is a deadly gynecologic malignancy that presents many clinical challenges due to late-stage diagnoses and the development of acquired resistance to standard-of-care treatment protocols. There is an increasing body of evidence suggesting that STATs may play a critical role in OvCa progression, resistance, and disease recurrence, and thus we sought to compile a comprehensive review to summarize the current state of knowledge on the topic. We have examined peer reviewed literature to delineate the role of STATs in both cancer cells and cells within the tumor microenvironment. In addition to summarizing the current knowledge of STAT biology in OvCa, we have also examined the capacity of small molecule inhibitor development to target specific STATs and progress toward clinical applications. From our research, the best studied and targeted factors are STAT3 and STAT5, which has resulted in the development of several inhibitors that are under current evaluation in clinical trials. There remain gaps in understanding the role of STAT1, STAT2, STAT4, and STAT6, due to limited reports in the current literature; as such, further studies to establish their implications in OvCa are necessitated. Moreover, due to the deficiency in our understanding of these STATs, selective inhibitors also remain elusive, and therefore present opportunities for discovery.
Collapse
Affiliation(s)
- David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Emma Feess
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Satvik Kodiyalam
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Michael Kuehn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Zachary Hamel
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Jaimie Johnson
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
5
|
Tan Y, Zhang L, Zhu G, Yang Y, Guo W, Chen L, Chang J, Xu Y, Muyey DM, Wang H. BCR/ABL1ΔE7-8-9 isoform contributes to tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Hematol Oncol 2022; 40:1067-1075. [PMID: 35686657 DOI: 10.1002/hon.3040] [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: 01/25/2022] [Revised: 05/10/2022] [Accepted: 06/04/2022] [Indexed: 12/13/2022]
Abstract
In chronic myeloid leukemia (CML) patients, the involvement of the BCR/ABL1 isoform in tyrosine kinase inhibitors (TKIs) resistance has attracted lots of attention. In this work, a novel isoform that encoded truncated protein due to the deletion of ABL1 exon7, 8, and 9 was reported and named BCR/ABL1ΔE7-8-9 here. This isoform was detected only in 10.2% of CML patients with inadequate responses to TKIs. BCR/ABL1Δexon7-8-9 isoform promoted S phase cell proliferation and reduced the expression of fusion gene and ABL1 phosphorylation level more slowly than that of control cells after TKIs treatment. The novel isoform has the qualities of a functional tyrosine kinase, localized in the cytoplasm, and could not be imported into the nucleus by TKIs. These results indicated that BCR/ABL1Δexon7-8-9 showed poorer sensitivity to imatinib and nilotinib than wild-type BCR/ABL1. According to molecular docking studies, nilotinib and imatinib present different binding sites and have a lower binding capacity with BCR/ABL1ΔE7-8-9 protein than the wild type. Our findings suggested that the novel isoform BCR/ABL1ΔE7-8-9 may contribute to TKIs resistance in CML due to its weakened TKIs binding ability. It enriched the mechanism of spliceosome involved in TKIs resistance. Monitoring the expression of BCR/ABL1ΔE7-8-9 helps guide the treatment of CML patients in the clinic.
Collapse
Affiliation(s)
- Yanhong Tan
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lingli Zhang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Guiyang Zhu
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuchao Yang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenzheng Guo
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lanhui Chen
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianmei Chang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yang Xu
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Daniel Muteb Muyey
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongwei Wang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
6
|
Assanto GM, Scalzulli E, Carmosino I, Martelli M, Breccia M. From bench to bedside: bridging the gaps in best practices for real-world chronic myeloid leukemia care. Expert Rev Hematol 2022; 15:963-971. [PMID: 36305791 DOI: 10.1080/17474086.2022.2142112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Although tyrosine kinase inhibitors (TKIs) determined an improvement of responses and overall survival (OS) in chronic phase chronic myeloid leukemia (CP-CML) patients, some patients still fail the achievement of important milestones. AREAS COVERED In this review, we focus on the need of appropriate molecular and mutational monitoring during TKI treatment with new laboratory tools and on new compounds developed to counteract the unmet clinical need in CP-CML. EXPERT OPINION The appropriate identification of BCR::ABL1 dependent and independent mechanisms of resistance with Next Generation Sequencing (NGS) and digital droplet PCR (ddPCR) can allow to improve the therapeutic strategies and prevent the onset of a failure to treatment. New compounds have been recently approved or are still in investigational trials to improve the response in some critical forms of resistance and/or intolerance to available TKIs.
Collapse
Affiliation(s)
- Giovanni Manfredi Assanto
- Department Cellular Biotechnol & Hematol, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Emilia Scalzulli
- Department Cellular Biotechnol & Hematol, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Ida Carmosino
- Department Cellular Biotechnol & Hematol, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Maurizio Martelli
- Department Cellular Biotechnol & Hematol, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Massimo Breccia
- Department Cellular Biotechnol & Hematol, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| |
Collapse
|
7
|
Genomic Mutations of the STAT5 Transcription Factor Are Associated with Human Cancer and Immune Diseases. Int J Mol Sci 2022; 23:ijms231911297. [PMID: 36232600 PMCID: PMC9569778 DOI: 10.3390/ijms231911297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Signal transducer and activation of transcription 5 (STAT5) is a key transcription factor that regulates various biological processes in mammalian development. Aberrant regulation of STAT5 has also been causally linked to many diseases, including cancers and immune-related diseases. Although persistent activation of STAT5 due to dysregulation of the signaling cascade has been reported to be associated with the progression of solid tumors and leukemia, various genomic mutations of STAT5 have also been found to cause a wide range of diseases. The present review comprehensively summarizes results of recent studies evaluating the intrinsic function of STAT5 and the link between STAT5 mutations and human diseases. This review also describes the types of disease models useful for investigating the mechanism underlying STAT5-driven disease progression. These findings provide basic knowledge for understanding the regulatory mechanisms of STAT5 and the progression of various diseases resulting from aberrant regulation of STAT5. Moreover, this review may provide insights needed to create optimal disease models that reflect human disease associated STAT5 mutations and to design gene therapies to correct STAT5 mutations.
Collapse
|
8
|
Poudel G, Tolland MG, Hughes TP, Pagani IS. Mechanisms of Resistance and Implications for Treatment Strategies in Chronic Myeloid Leukaemia. Cancers (Basel) 2022; 14:cancers14143300. [PMID: 35884363 PMCID: PMC9317051 DOI: 10.3390/cancers14143300] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Chronic myeloid leukaemia (CML) is a type of blood cancer that is currently well-managed with drugs that target cancer-causing proteins. However, a significant proportion of CML patients do not respond to those drug treatments or relapse when they stop those drugs because the cancer cells in those patients stop relying on that protein and instead develop a new way to survive. Therefore, new treatment strategies may be necessary for those patients. In this review, we discuss those additional survival pathways and outline combination treatment strategies to increase responses and clinical outcomes, improving the lives of CML patients. Abstract Tyrosine kinase inhibitors (TKIs) have revolutionised the management of chronic myeloid leukaemia (CML), with the disease now having a five-year survival rate over 80%. The primary focus in the treatment of CML has been on improving the specificity and potency of TKIs to inhibit the activation of the BCR::ABL1 kinase and/or overcoming resistance driven by mutations in the BCR::ABL1 oncogene. However, this approach may be limited in a significant proportion of patients who develop TKI resistance despite the effective inhibition of BCR::ABL1. These patients may require novel therapeutic strategies that target both BCR::ABL1-dependent and BCR::ABL1-independent mechanisms of resistance. The combination treatment strategies that target alternative survival signalling, which may contribute towards BCR::ABL1-independent resistance, could be a successful strategy for eradicating residual leukaemic cells and consequently increasing the response rate in CML patients.
Collapse
Affiliation(s)
- Govinda Poudel
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
| | - Molly G. Tolland
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Timothy P. Hughes
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital and SA Pathology, Adelaide, SA 5000, Australia
| | - Ilaria S. Pagani
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
- Correspondence:
| |
Collapse
|
9
|
Ozel B, Kipcak S, Biray Avci C, Sabour Takanlou M, Sabour Takanlou L, Tezcanli Kaymaz B, Karatekin I, Gunduz C, Selvi Gunel N. Targeting UPR signaling pathway by dasatinib as a promising therapeutic approach in chronic myeloid leukemia. Med Oncol 2022; 39:126. [PMID: 35716222 DOI: 10.1007/s12032-022-01714-y] [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: 03/01/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease that mediated by BCR/ABL oncogenic signaling. CML can be targeted with the imatinib, dasatinib, and nilotinib TKI inhibitors, the latter two of them have been approved for imatinib-resistant or -intolerant CML patients. The TKIs resistance occurs by different molecular mechanisms, including overexpression of BCR-ABL, mutations in the TKI binding site of BCR/ABL, and ER-stress. Unfolded protein responses (UPR) is a cytoprotective mechanism which is activated by ER-stress. The IRE1, PERK, and ATF6 are three main arms of the UPR mechanism and are activated by a common mechanism involving the dissociation of the ER-chaperone BiP/GP78. There is a correlation between ER-stress, CML progression, and response to TKI treatment. In the present study, we aimed to determine alterations of the expression levels of genes related to UPR pathway signaling after treatment with dasatinib in K562 chronic myeloid leukemia cell line by quantitative RT-PCR relatively. The array-data revealed that treatment with dasatinib significantly decreased the UPR mechanism-related genes (including HSPA1B, HSPA2, HSPA4L, ATF6, ATF6B, CEBPB, PERK, TRIB3, DNAJB, ERN1, and UHRF1) in K562 cells. In conclusion, the results showed that dasatinib regulates the UPR mechanism that plays a significant role in cancer progression and therapy resistance in CML. Thus, dasatinib-induced dysfunction of the UPR mechanism may promise encouraging therapy for CML.
Collapse
Affiliation(s)
- Buket Ozel
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Sezgi Kipcak
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Cigir Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey.
| | - Maryam Sabour Takanlou
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Leila Sabour Takanlou
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Burcin Tezcanli Kaymaz
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Ilknur Karatekin
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| | - Nur Selvi Gunel
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, 35100, Izmir, Turkey
| |
Collapse
|
10
|
Tang Y, Xiao S, Wang Z, Liang Y, Xing Y, Wu J, Lu M. A Prognostic Model for Acute Myeloid Leukemia Based on IL-2/STAT5 Pathway-Related Genes. Front Oncol 2022; 12:785899. [PMID: 35186733 PMCID: PMC8847395 DOI: 10.3389/fonc.2022.785899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Accurate prognostic stratification of patients can provide guidance for personalized therapy. Many prognostic models for acute myeloid leukemia (AML) have been reported, but most have considerable inaccuracies due to contained variables with insufficient capacity of predicting survival and lack of adequate verification. Here, 235 genes strongly related to survival in AML were systematically identified through univariate Cox regression analysis of eight independent AML datasets. Pathway enrichment analysis of these 235 genes revealed that the IL-2/STAT5 signaling pathway was the most highly enriched. Through Cox proportional-hazards regression model and stepwise algorithm, we constructed a six-gene STAT5-associated signature based on the most robustly survival-related genes related to the IL-2/STAT5 signaling pathway. Good prognostic performance was observed in the training cohort (GSE37642-GPL96), and the signature was validated in seven other validation cohorts. As an independent prognostic factor, the STAT5-associated signature was positively correlated with patient age and ELN2017 risk levels. An integrated score based on these three prognostic factors had higher prognostic accuracy than the ELN2017 risk category. Characterization of immune cell infiltration indicated that impaired B-cell adaptive immunity, immunosuppressive effects, serious infection, and weakened anti-inflammatory function tended to accompany high-risk patients. Analysis of in-house clinical samples revealed that the STAT5-assocaited signature risk scores of AML patients were significantly higher than those of healthy people. Five chemotherapeutic drugs that were effective in these high-risk patients were screened in silico. Among the five drugs, MS.275, a known HDAC inhibitor, selectively suppressed the proliferation of cancer cells with high STAT5 phosphorylation levels in vitro. Taken together, the data indicate that the STAT5-associated signature is a reliable prognostic model that can be used to optimize prognostic stratification and guide personalized AML treatments.
Collapse
Affiliation(s)
- Yigang Tang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujun Xiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengyuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Liang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangfei Xing
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiale Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Lewuillon C, Laguillaumie MO, Quesnel B, Idziorek T, Touil Y, Lemonnier L. Put in a “Ca2+ll” to Acute Myeloid Leukemia. Cells 2022; 11:cells11030543. [PMID: 35159351 PMCID: PMC8834247 DOI: 10.3390/cells11030543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder characterized by genetic aberrations in myeloid primitive cells (blasts) which lead to their defective maturation/function and their proliferation in the bone marrow (BM) and blood of affected individuals. Current intensive chemotherapy protocols result in complete remission in 50% to 80% of AML patients depending on their age and the AML type involved. While alterations in calcium signaling have been extensively studied in solid tumors, little is known about the role of calcium in most hematologic malignancies, including AML. Our purpose with this review is to raise awareness about this issue and to present (i) the role of calcium signaling in AML cell proliferation and differentiation and in the quiescence of hematopoietic stem cells; (ii) the interplay between mitochondria, metabolism, and oxidative stress; (iii) the effect of the BM microenvironment on AML cell fate; and finally (iv) the mechanism by which chemotherapeutic treatments modify calcium homeostasis in AML cells.
Collapse
Affiliation(s)
- Clara Lewuillon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Marie-Océane Laguillaumie
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Thierry Idziorek
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Yasmine Touil
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Loïc Lemonnier
- Univ. Lille, Inserm, U1003—PHYCEL—Physiologie Cellulaire, F-59000 Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, F-59655 Villeneuve d’Ascq, France
- Correspondence:
| |
Collapse
|
12
|
Bakhshayesh M, Gohari LH, Barati M, Safa M. Combination therapy using TGF-β1 and STI-571 can induce apoptosis in BCR-ABL oncogene-expressing cells. Biomol Concepts 2021; 12:144-155. [PMID: 34700368 DOI: 10.1515/bmc-2021-0016] [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: 08/02/2021] [Accepted: 09/24/2021] [Indexed: 11/15/2022] Open
Abstract
The BCR-ABL oncogene is a tyrosine kinase gene that is over-expressed in CML. It inhibits the TGF-β1 signaling pathway. Due to resistance of cells to the tyrosine kinase inhibitor, STI-571, the combined effect of STI-571 and TGF-β1 on K562 cells was studied in the present research. Results revealed that the TGF-β1 cell signaling pathway, which is activated in K562 cells treated with TGF-β1, activates collective cell signaling pathways involved in survival and apoptosis. It is noteworthy that treating K562 cells with STI-571 triggered apoptotic pathways, accompanied by a reduction in proteins such as Bcl-xL, Bcl-2, p-AKT, p-Stat5, p-FOXO3, and Mcl-1 and an increase in the pro-apoptotic proteins PARP cleavage, and p27, leading to an increase in sub-G1 phase-arrested and Annexin-positive cells. Interestingly, the proliferation behavior of TGF-β1-induced cells was changed with the combination therapy, and STI-571-induced apoptosis was also prompted by this combination. Thus, combination treatment appears to promote sub-G1 cell cycle arrest compared to individually treated cells. Furthermore, it strongly triggered apoptotic signaling. In conclusion, TGF-β1 did not negatively impact the effect of STI-571, based on positive annexin cells, and AKT protein phosphorylation remains effective in apoptosis.
Collapse
Affiliation(s)
- Masoome Bakhshayesh
- Genetics department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ladan Hosseini Gohari
- Cellular & Molecular Research Center, Medical Laboratory Science Department, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Safa
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Krishnan V, Kim DDH, Hughes TP, Branford S, Ong ST. Integrating genetic and epigenetic factors in chronic myeloid leukemia risk assessment: toward gene expression-based biomarkers. Haematologica 2021; 107:358-370. [PMID: 34615339 PMCID: PMC8804571 DOI: 10.3324/haematol.2021.279317] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 11/17/2022] Open
Abstract
Cancer treatment is constantly evolving from a one-size-fits-all towards bespoke approaches for each patient. In certain solid cancers, including breast and lung, tumor genome profiling has been incorporated into therapeutic decision-making. For chronic phase chronic myeloid leukemia (CML), while tyrosine kinase inhibitor therapy is the standard treatment, current clinical scoring systems cannot accurately predict the heterogeneous treatment outcomes observed in patients. Biomarkers capable of segregating patients according to outcome at diagnosis are needed to improve management, and facilitate enrollment in clinical trials seeking to prevent blast crisis transformation and improve the depth of molecular responses. To this end, gene expression (GE) profiling studies have evaluated whether GE signatures at diagnosis are clinically informative. Patient material from a variety of sources has been profiled using microarrays, RNA sequencing and, more recently, single-cell RNA sequencing. However, differences in the cell types profiled, the technologies used, and the inherent complexities associated with the interpretation of genomic data pose challenges in distilling GE datasets into biomarkers with clinical utility. The goal of this paper is to review previous studies evaluating GE profiling in CML, and explore their potential as risk assessment tools for individualized CML treatment. We also review the contribution that acquired mutations, including those seen in clonal hematopoiesis, make to GE profiles, and how a model integrating contributions of genetic and epigenetic factors in resistance to tyrosine kinase inhibitors and blast crisis transformation can define a route to GE-based biomarkers. Finally, we outline a four-stage approach for the development of GE-based biomarkers in CML.
Collapse
Affiliation(s)
- Vaidehi Krishnan
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore; International Chronic Myeloid Leukemia Foundation
| | - Dennis Dong Hwan Kim
- International Chronic Myeloid Leukemia Foundation; Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto
| | - Timothy P Hughes
- International Chronic Myeloid Leukemia Foundation; School of Medicine, University of Adelaide, Adelaide, Australia; South Australian Health and Medical Research Institute, Adelaide, Australia; Department of Haematology, Royal Adelaide Hospital, Adelaide
| | - Susan Branford
- International Chronic Myeloid Leukemia Foundation; School of Medicine, University of Adelaide, Adelaide, Australia; Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; School of Pharmacy and Medical Science, University of South Australia, Adelaide
| | - S Tiong Ong
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore; International Chronic Myeloid Leukemia Foundation; Department of Haematology, Singapore General Hospital, Singapore, Singapore; Department of Medical Oncology, National Cancer Centre Singapore; Department of Medicine, Duke University Medical Center, Durham, NC.
| |
Collapse
|
14
|
Narasimhan M, Khamkar V, Tilwani S, Dalal SN, Shetty D, Subramanian PG, Gupta S, Govekar R. Atypical activation of signaling downstream of inactivated Bcr-Abl mediates chemoresistance in chronic myeloid leukemia. J Cell Commun Signal 2021; 16:207-222. [PMID: 34596797 DOI: 10.1007/s12079-021-00647-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/20/2021] [Indexed: 11/28/2022] Open
Abstract
Chronic myeloid leukemia (CML) epitomises successful targeted therapy, where inhibition of tyrosine kinase activity of oncoprotein Bcr-Abl1 by imatinib, induces remission in 86% patients in initial chronic phase (CP). However, in acute phase of blast crisis, 80% patients show resistance, 40% among them despite inhibition of Bcr-Abl1 activity. This implies activation of either Bcr-Abl1- independent signalling pathways or restoration of signalling downstream of inactive Bcr-Abl1. In the present study, mass spectrometry and subsequent in silico pathway analysis of differentiators in resistant CML-CP cells identified key differentiators, 14-3-3ε and p38 MAPK, which belong to Bcr-Abl1 pathway. Their levels and activity respectively, indicated active Bcr-Abl1 pathway in CML-BC resistant cells, though Bcr-Abl1 is inhibited by imatinib. Further, contribution of these components to resistance was demonstrated by inhibition of Bcr-Abl1 down-stream signalling by knocking-out of 14-3-3ε and inhibition of p38 MAPK activity. The observations merit clinical validation to explore their translational potential.
Collapse
Affiliation(s)
- Mythreyi Narasimhan
- Rukmini Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.,Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Vaishnavi Khamkar
- Rukmini Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Sarika Tilwani
- Sorab Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.,Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Sorab N Dalal
- Sorab Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.,Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Dhanlaxmi Shetty
- Department of Cancer Cytogenetics, , ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - P G Subramanian
- Hematopathology Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.,Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Sanjay Gupta
- Gupta Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.,Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Rukmini Govekar
- Rukmini Lab, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India. .,Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Mumbai, 400094, India.
| |
Collapse
|
15
|
Rinaldi I, Putri A, Louisa M, Koesnoe S. High STAT5A Expression is Associated with Major Molecular Response Achievement Failure of Chronic Phase Chronic Myeloid Leukemia Patients Receiving Hydroxyurea before Imatinib: A Cross-sectional Study. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: STAT5 is a transcriptional factor which when highly expressed in chronic myeloid leukemia (CML) cells stimulate proliferation and mediate resistance from tyrosine kinase inhibitors, resulting in major molecular response (MMR) failure. STAT5 has two isoforms, STAT5A and STAT5B. However, prolonged use of imatinib appears to only upregulate STAT5A pathway. In addition, the resistance conferred by STAT5A does not extend to other drugs such as hydroxyurea. Hence, STAT5A and STAT5B might have different functions in CML cells.
AIM: The objective of the study was to determine the association of STAT5A and STAT5B expression with MMR failure in CML patients.
METHODS: This was a cross-sectional study of CML patients in chronic phase with age ≥ 18 years old, receiving IM therapy ≥ 12 months, and previously given hydroxyurea. MMR status was evaluated and patients were categorized as achieved or failed to achieve MMR. Expression levels of STAT5A and STAT5B were conducted using RT-PCR methods. Associations between STAT5A expression, STAT5B expression, hydroxyurea duration, and imatinib duration with MMR achievement were calculated using logistic regression.
RESULTS: A total of 118 patients were analyzed; 71.1% failed to achieve MMR. Multivariate logistic regression analysis showed statistically significant association between high STAT5A expression (odds ratio [OR]: 3.852; 95% confidence interval [CI]: 1.420–10.452; p = 0.008), STAT5A/STAT5B interaction (OR: 0.150; 95% CI: 0.038–0.593; p = 0.007), longer hydroxyurea administration (OR: 3.882; 95% CI: 1.023–14.733; p = 0.046), and shorter imatinib administration (OR: 0.333; 95% CI: 0.132–0.840; p = 0.020) with MMR achievement failure. After adjusting STAT5A expression with STAT5A/STAT5B interaction, high STAT5A expression independently increased the likelihood of MMR achievement failure only in high expression STAT5B patients (OR: 3.852; 95% CI: 1.420–10.452; p = 0.008).
CONCLUSION: High STAT5A expression which is induced by high STAT5B is associated with MMR achievement failure of chronic phase CML patients who received hydroxyurea before imatinib. Longer duration of hydroxyurea and shorter duration of IM confound of STAT5A expression to MMR achievement failure.
Collapse
|
16
|
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: 66] [Impact Index Per Article: 22.0] [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
|
17
|
Pons M, Zeyn Y, Zahn S, Mahendrarajah N, Page BDG, Gunning PT, Moriggl R, Brenner W, Butter F, Krämer OH. Oncogenic Kinase Cascades Induce Molecular Mechanisms That Protect Leukemic Cell Models from Lethal Effects of De Novo dNTP Synthesis Inhibition. Cancers (Basel) 2021; 13:3464. [PMID: 34298678 PMCID: PMC8304262 DOI: 10.3390/cancers13143464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/15/2023] Open
Abstract
The ribonucleotide reductase inhibitor hydroxyurea suppresses de novo dNTP synthesis and attenuates the hyperproliferation of leukemic blasts. Mechanisms that determine whether cells undergo apoptosis in response to hydroxyurea are ill-defined. We used unbiased proteomics to uncover which pathways control the transition of the hydroxyurea-induced replication stress into an apoptotic program in chronic and acute myeloid leukemia cells. We noted a decrease in the serine/threonine kinase RAF1/c-RAF in cells that undergo apoptosis in response to clinically relevant doses of hydroxyurea. Using the RAF inhibitor LY3009120, we show that RAF activity determines the sensitivity of leukemic cells toward hydroxyurea. We further disclose that pharmacological inhibition of the RAF downstream target BCL-XL with the drug navitoclax and RNAi combine favorably with hydroxyurea against leukemic cells. BCR-ABL1 and hyperactive FLT3 are tyrosine kinases that causally contribute to the development of leukemia and induce RAF1 and BCL-XL. Accordingly, the ABL inhibitor imatinib and the FLT3 inhibitor quizartinib sensitize leukemic cells to pro-apoptotic effects of hydroxyurea. Moreover, hydroxyurea and navitoclax kill leukemic cells with mutant FLT3 that are resistant to quizartinib. These data reveal cellular susceptibility factors toward hydroxyurea and how they can be exploited to eliminate difficult-to-treat leukemic cells with clinically relevant drug combinations.
Collapse
Affiliation(s)
- Miriam Pons
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (S.Z.); (N.M.)
| | - Yanira Zeyn
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (S.Z.); (N.M.)
| | - Stella Zahn
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (S.Z.); (N.M.)
| | - Nisintha Mahendrarajah
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (S.Z.); (N.M.)
| | - Brent D. G. Page
- Faculty of Pharmaceutical Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
| | - Patrick T. Gunning
- Department of Chemical & Physical Sciences, University of Toronto, Mississauga, ON L5L 1C6, Canada;
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Walburgis Brenner
- Clinic for Obstetrics and Women’s Health, University Medical Center, 55131 Mainz, Germany;
| | - Falk Butter
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany;
| | - Oliver H. Krämer
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (S.Z.); (N.M.)
| |
Collapse
|
18
|
Therapeutic delivery of siRNA with polymeric carriers to down-regulate STAT5A expression in high-risk B-cell acute lymphoblastic leukemia (B-ALL). PLoS One 2021; 16:e0251719. [PMID: 34157051 PMCID: PMC8219370 DOI: 10.1371/journal.pone.0251719] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/02/2021] [Indexed: 11/19/2022] Open
Abstract
Overexpression and persistent activation of STAT5 play an important role in the development and progression of acute lymphoblastic leukemia (ALL), the most common pediatric cancer. Small interfering RNA (siRNA)-mediated downregulation of STAT5 represents a promising therapeutic approach for ALL to overcome the limitations of current treatment modalities such as high relapse rates and poor prognosis. However, to effectively transport siRNA molecules to target cells, development of potent carriers is of utmost importance to surpass hurdles of delivery. In this study, we investigated the use of lipopolymers as non-viral delivery systems derived from low molecular weight polyethylenimines (PEI) substituted with lauric acid (Lau), linoleic acid (LA) and stearic acid (StA) to deliver siRNA molecules to ALL cell lines and primary samples. Among the lipid-substituted polymers explored, Lau- and LA-substituted PEI displayed excellent siRNA delivery to SUP-B15 and RS4;11 cells. STAT5A gene expression was downregulated (36-92%) in SUP-B15 and (32%) in RS4;11 cells using the polymeric delivery systems, which consequently reduced cell growth and inhibited the formation of colonies in ALL cells. With regard to ALL primary cells, siRNA-mediated STAT5A gene silencing was observed in four of eight patient cells using our leading polymeric delivery system, 1.2PEI-Lau8, accompanied by the significant reduction in colony formation in three of eight patients. In both BCR-ABL positive and negative groups, three of five patients demonstrated marked cell growth inhibition in both MTT and trypan blue exclusion assays using 1.2PEI-Lau8/siRNA complexes in comparison with their control siRNA groups. Three patient samples did not show any positive results with our delivery systems. Differential therapeutic responses to siRNA therapy observed in different patients could result from variable genetic profiles and patient-to-patient variability in delivery. This study supports the potential of siRNA therapy and the designed lipopolymers as a delivery system in ALL therapy.
Collapse
|
19
|
Zhang H, Wang P, Song T, Bonnette UL, Zhang Z. Screening and identification of key genes in imatinib-resistant chronic myelogenous leukemia cells: a bioinformatics study. ACTA ACUST UNITED AC 2021; 26:408-414. [PMID: 34053416 DOI: 10.1080/16078454.2021.1931740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Chronic myelogenous leukemia (CML) is one of the most common cancers in the world. Imatinib is one of the most effective therapeutic strategies to inhibit the BCR-ABL tyrosine Kinase in patients with CML, but resistance is increasingly encountered. MATERIAL AND METHODS Microarray data GSE7114, GSE92624 and GSE97562 were downloaded and analyzed from Gene Expression Omnibus (GEO) to identify the candidate genes in the imatinib-resistant CML cells. The differentially expressed genes (DEGs) were appraised, and the protein-protein interaction (PPI) network was created by using STRING and Cytoscape. RESULTS We screened a total of 217 DEGs, including 151 upregulated genes and 66 downregulated genes. The enriched functions and pathways of genes include insulin-like growth factor I binding, cysteine-type endopeptidase inhibitor activity involved in apoptotic process, cell adhesion, positive regulation of nitric oxide biosynthetic process and hematopoietic cell lineage. Nine hub genes were appraised and Gene Ontology enrichment analysis revealed that these genes are mainly enriched in cell cycle, peptidase inhibitor activity and cell division. Several genes such as BIRC5, CCNE2 and MCM4 were identified in survival analysis and these genes alteration are significantly associated with worse overall survival and disease-free survival. CONCLUSIONS These genes have the potential to become surrogate markers for a clinical evaluation of imatinib-resistant CML patients. Our results provide potential target genes for diagnosis and treatment of imatinib-resistant CML patients.
Collapse
Affiliation(s)
- Hong Zhang
- State Key Laboratory of Fine Chemicals, School of Life Science and Technology, Dalian University of Technology, Dalian, People's Republic of China
| | - Peiran Wang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| | - Ting Song
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| | - Uwituze Laura Bonnette
- State Key Laboratory of Fine Chemicals, School of Life Science and Technology, Dalian University of Technology, Dalian, People's Republic of China
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, School of Life Science and Technology, Dalian University of Technology, Dalian, People's Republic of China
| |
Collapse
|
20
|
Nehme R, Hallal R, El Dor M, Kobeissy F, Gouilleux F, Mazurier F, Zibara K. Repurposing of Acriflavine to Target Chronic Myeloid Leukemia Treatment. Curr Med Chem 2021; 28:2218-2233. [PMID: 32900342 DOI: 10.2174/0929867327666200908114411] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 11/22/2022]
Abstract
Drug repurposing has lately received increasing interest in several diseases especially in cancers, due to its advantages in facilitating the development of new therapeutic strategies, by adopting a cost-friendly approach and avoiding the strict Food and Drug Administration (FDA) regulations. Acriflavine (ACF) is an FDA approved molecule that has been extensively studied since 1912 with antiseptic, trypanocidal, anti-viral, anti-bacterial and anti-cancer effects. ACF has been shown to block the growth of solid and hematopoietic tumor cells. Indeed, ACF acts as an inhibitor of various proteins, including DNA-dependent protein kinases C (DNA-PKcs), topoisomerase I and II, hypoxia-inducible factor 1α (HIF-1α), in addition to its recent discovery as an inhibitor of the signal transducer and activator of transcription (STAT). Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder characterized by the expression of the constitutively active tyrosine kinase BCR-ABL. This protein allows the activation of several signaling pathways known for their role in cell proliferation and survival, such as the JAK/STAT pathway. CML therapy, based on tyrosine kinase inhibitors (TKIs), such as imatinib (IM), is highly effective. However, 15% of patients are refractory to IM, where in some cases, 20-30% of patients become resistant. Thus, we suggest the repurposing of ACF in CML after IM failure or in combination with IM to improve the anti-tumor effects of IM. In this review, we present the different pharmacological properties of ACF along with its anti-leukemic effects in the hope of its repurposing in CML therapy.
Collapse
Affiliation(s)
- Rawan Nehme
- Universite de Tours, EA7501 GICC, Tours, France
| | | | - Maya El Dor
- Universite de Tours, EA7501 GICC, Tours, France
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | |
Collapse
|
21
|
Mu H, Zhu X, Jia H, Zhou L, Liu H. Combination Therapies in Chronic Myeloid Leukemia for Potential Treatment-Free Remission: Focus on Leukemia Stem Cells and Immune Modulation. Front Oncol 2021; 11:643382. [PMID: 34055612 PMCID: PMC8155539 DOI: 10.3389/fonc.2021.643382] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Although tyrosine Kinase Inhibitors (TKI) has revolutionized the treatment of chronic myeloid leukemia (CML), patients are not cured with the current therapy modalities. Also, the more recent goal of CML treatment is to induce successful treatment-free remission (TFR) among patients achieving durable deep molecular response (DMR). Together, it is necessary to develop novel, curative treatment strategies. With advancements in understanding the biology of CML, such as dormant Leukemic Stem Cells (LSCs) and impaired immune modulation, a number of agents are now under investigation. This review updates such agents that target LSCs, and together with TKIs, have the potential to eradicate CML. Moreover, we describe the developing immunotherapy for controlling CML.
Collapse
Affiliation(s)
- Hui Mu
- Medical School, Nantong University, Nantong, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Jia
- Medical School, Nantong University, Nantong, China
| | - Lu Zhou
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
22
|
Chung SS, Ng JCF, Laddach A, Thomas NSB, Fraternali F. Short loop functional commonality identified in leukaemia proteome highlights crucial protein sub-networks. NAR Genom Bioinform 2021; 3:lqab010. [PMID: 33709075 PMCID: PMC7936661 DOI: 10.1093/nargab/lqab010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/19/2020] [Accepted: 01/26/2021] [Indexed: 11/13/2022] Open
Abstract
Direct drug targeting of mutated proteins in cancer is not always possible and efficacy can be nullified by compensating protein-protein interactions (PPIs). Here, we establish an in silico pipeline to identify specific PPI sub-networks containing mutated proteins as potential targets, which we apply to mutation data of four different leukaemias. Our method is based on extracting cyclic interactions of a small number of proteins topologically and functionally linked in the Protein-Protein Interaction Network (PPIN), which we call short loop network motifs (SLM). We uncover a new property of PPINs named 'short loop commonality' to measure indirect PPIs occurring via common SLM interactions. This detects 'modules' of PPI networks enriched with annotated biological functions of proteins containing mutation hotspots, exemplified by FLT3 and other receptor tyrosine kinase proteins. We further identify functional dependency or mutual exclusivity of short loop commonality pairs in large-scale cellular CRISPR-Cas9 knockout screening data. Our pipeline provides a new strategy for identifying new therapeutic targets for drug discovery.
Collapse
Affiliation(s)
- Sun Sook Chung
- Department of Haematological Medicine, King's College London, London, SE5 9NU, UK
| | - Joseph C F Ng
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
| | - Anna Laddach
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
| | - N Shaun B Thomas
- Department of Haematological Medicine, King's College London, London, SE5 9NU, UK
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
| |
Collapse
|
23
|
Schoepf AM, Salcher S, Obexer P, Gust R. Tackling resistance in chronic myeloid leukemia: Novel cell death modulators with improved efficacy. Eur J Med Chem 2021; 216:113285. [PMID: 33662676 DOI: 10.1016/j.ejmech.2021.113285] [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: 10/10/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 10/22/2022]
Abstract
The development of resistance poses a serious problem in the therapy of cancer due to the necessity of a multiple-drug and unlimited treatment of affected patients. In chronic myeloid leukemia (CML), the introduction of imatinib has revolutionized the therapy. The persistence of an untreatable cancer stem cell pool and other resistance-causing factors, however, also impede the cure of this malignancy. New therapeutic approaches are therefore essential to overcome current treatment drawbacks. In this regard, an intervention in the STAT5 signaling pathway can significantly improve drug response, as this central signaling node induces the formation of highly resistant CML cells. In the present study, we continued the design of efficient chemosensitizers derived from the partial PPARγ agonist telmisartan. The developed 2-carbonitriles or 2-carboxymethyl esters showed improved potency in sensitizing K562-resistant cells to imatinib treatment, even at concentrations, which are considered patient-relevant. At 5 μM, for instance, 2d sensitized the cells in such a manner that the resistance was fully overcome and the recovered efficacy of imatinib resulted in >76% cell death. Importantly, all compounds were non-cytotoxic per se. A transactivation experiment showed that only the carbonitriles are partial agonists of PPARγ, which does not seem to be involved in the mode of action. Yet, immunoassays revealed a suppression of the STAT5 phosphorylation status by co-application of the most active derivatives with imatinib. This mechanism consequently resulted in reduced cell proliferation and induction of cell death in resistant CML cells.
Collapse
Affiliation(s)
- Anna M Schoepf
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI - Center for Molecular Biosciences Innsbruck, University of Innsbruck, CCB - Centrum for Chemistry and Biomedicine, Innrain 80-82, 6020, Innsbruck, Austria; Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, United Kingdom
| | - Stefan Salcher
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria; Department of Internal Medicine V, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Petra Obexer
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria; Department of Pediatrics II, Medical University Innsbruck, Innrain 66, 6020, Innsbruck, Austria
| | - Ronald Gust
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI - Center for Molecular Biosciences Innsbruck, University of Innsbruck, CCB - Centrum for Chemistry and Biomedicine, Innrain 80-82, 6020, Innsbruck, Austria.
| |
Collapse
|
24
|
Zhang C, Shen L, Zhu Y, Xu R, Deng Z, Liu X, Ding Y, Wang C, Shi Y, Bei L, Wei D, Thorne RF, Zhang XD, Yu L, Chen S. KDM6A promotes imatinib resistance through YY1-mediated transcriptional upregulation of TRKA independently of its demethylase activity in chronic myelogenous leukemia. Am J Cancer Res 2021; 11:2691-2705. [PMID: 33456567 PMCID: PMC7806474 DOI: 10.7150/thno.50571] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Despite landmark therapy of chronic myelogenous leukemia (CML) with tyrosine kinase inhibitors (TKIs), drug resistance remains problematic. Cancer pathogenesis involves epigenetic dysregulation and in particular, histone lysine demethylases (KDMs) have been implicated in TKI resistance. We sought to identify KDMs with altered expression in CML and define their contribution to imatinib resistance. Methods: Bioinformatics screening compared KDM expression in CML versus normal bone marrow with shRNA knockdown and flow cytometry used to measure effects on imatinib-induced apoptosis in K562 cells. Transcriptomic analyses were performed against KDM6A CRISPR knockout/shRNA knockdown K562 cells along with gene rescue experiments using wildtype and mutant demethylase-dead KDM6A constructs. Co-immunoprecipitation, luciferase reporter and ChIP were employed to elucidate mechanisms of KDM6A-dependent resistance. Results: Amongst five KDMs upregulated in CML, only KDM6A depletion sensitized CML cells to imatinib-induced apoptosis. Re-introduction of demethylase-dead KDM6A as well as wild-type KDM6A restored imatinib resistance. RNA-seq identified NTRK1 gene downregulation after depletion of KDM6A. Moreover, NTRK1 expression positively correlated with KDM6A in a subset of clinical CML samples and KDM6A knockdown in fresh CML isolates decreased NTRK1 encoded protein (TRKA) expression. Mechanistically, KDM6A was recruited to the NTRK1 promoter by the transcription factor YY1 with subsequent TRKA upregulation activating down-stream survival pathways to invoke imatinib resistance. Conclusion: Contrary to its reported role as a tumor suppressor and independent of its demethylase function, KDM6A promotes imatinib-resistance in CML cells. The identification of the KDM6A/YY1/TRKA axis as a novel imatinib-resistance mechanism represents an unexplored avenue to overcome TKI resistance in CML.
Collapse
|
25
|
Sobhia ME, Kumar GS, Mallick A, Singh H, Kumar K, Chaurasiya M, Singh M, Gera N, Deverakonda S, Baghel V. Computational and Biological Investigations on Abl1 Tyrosine Kinase: A Review. Curr Drug Targets 2020; 22:38-51. [PMID: 33050861 DOI: 10.2174/1389450121999201013152513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/21/2020] [Accepted: 09/10/2020] [Indexed: 11/22/2022]
Abstract
Abl1 tyrosine kinase is a validated target for the treatment of chronic myeloid leukemia. It is a form of cancer that is difficult to treat and much research is being done to identify new molecular entities and to tackle drug resistance issues. In recent years, drug resistance of Abl1 tyrosine kinase has become a major healthcare concern. Second and third-generation TKI reported better responses against the resistant forms; still they had no impact on long-term survival prolongation. New compounds derived from natural products and organic small molecule inhibitors can lay the foundation for better clinical therapies in the future. Computational methods, experimental and biological studies can help us understand the mechanism of drug resistance and identify novel molecule inhibitors. ADMET parameters analysis of reported drugs and novel small molecule inhibitors can also provide valuable insights. In this review, available therapies, point mutations, structure-activity relationship and ADMET parameters of reported series of Abl1 tyrosine kinase inhibitors and drugs are summarised. We summarise in detail recent computational and molecular biology studies that focus on designing drug molecules, investigation of natural product compounds and organic new chemical entities. Current ongoing research suggests that selective targeting of Abl1 tyrosine kinase at the molecular level to combat drug resistance in chronic myeloid leukemia is promising.
Collapse
Affiliation(s)
- Masilamani Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - G Siva Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Antara Mallick
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Harmanpreet Singh
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Kranthi Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Meenakshi Chaurasiya
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Monica Singh
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Narendra Gera
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Sindhuja Deverakonda
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| | - Vinay Baghel
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, India
| |
Collapse
|
26
|
Prospects for Clinical Development of Stat5 Inhibitor IST5-002: High Transcriptomic Specificity in Prostate Cancer and Low Toxicity In Vivo. Cancers (Basel) 2020; 12:cancers12113412. [PMID: 33217941 PMCID: PMC7724566 DOI: 10.3390/cancers12113412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary There is an unmet medical need for new and potent pharmacological inhibitor drugs for the protein Stat5 that would be orally bioavailable for treatment of several different cancers. Previous work has established a critical role for Stat5 in molecular and clinical progression of prostate cancer to metastatic disease and in the pathogenesis of several leukemias and blood-based disorders. Our group has developed a potent pharmacological inhibitor for Stat5, IST5-002, which targets two critical steps in the activation process of Stat5 in cancer cells. In the present work, we evaluated the characteristics of IST5-002 for further development into a cancer drug. We evaluated whether IST5-002 affects the Stat5 targets genes in prostate cancer, defined more closely its mechanisms of action, and investigated its initial toxicity as the basis for further development in order to enable its entrance into clinical testing in patients. Our study supports optimization of IST5-002 compound for oral bioavailability and for clinical development. Abstract Stat5 is of significant interest in the search for new therapeutics for prostate cancer (PC) and hematopoietic disorders. We evaluated the transcriptomic specificity of the Stat5a/b inhibitor IST5-002 (IST5) in PC, defined more closely its mechanisms of action, and investigated the in vivo toxicity of IST5 for further optimization for clinical development. The transcriptomic specificity of IST5 vs. genetic Stat5 knockdown was evaluated by RNA-seq analysis, which showed high similarity with the Pearson correlation coefficient ranging from 0.98–0.99. The potency of IST5 vs. its derivative lacking the phosphate group in suppressing Stat5 was evaluated in two separate but complementary assays. The inhibitory activity of IST5 against kinases was investigated in cell-free assays followed by more focused evaluation in a cell-based assay. IST5 has no specific inhibitory activity against 54 kinases, while suppressing Stat5 phosphorylation and subsequent dimerization in PC cells. The phosphate group was not critical for the biological activity of IST5 in cells. The acute, sub-chronic and chronic toxicity studies of IST5 were carried out in mice. IST5 did not cause any significant toxic effects or changes in the blood profiles. The present work supports further optimization of IST5 for oral bioavailability for clinical development for therapies for solid tumors, hematological and myeloproliferative disorders.
Collapse
|
27
|
Involvement of STAT5 in Oncogenesis. Biomedicines 2020; 8:biomedicines8090316. [PMID: 32872372 PMCID: PMC7555335 DOI: 10.3390/biomedicines8090316] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins, and in particular STAT3, have been established as heavily implicated in cancer. Recently, the involvement of STAT5 signalling in the pathology of cancer has been shown to be of increasing importance. STAT5 plays a crucial role in the development of the mammary gland and the homeostasis of the immune system. However, in various cancers, aberrant STAT5 signalling promotes the expression of target genes, such as cyclin D, Bcl-2 and MMP-2, that result in increased cell proliferation, survival and metastasis. To target constitutive STAT5 signalling in cancers, there are several STAT5 inhibitors that can prevent STAT5 phosphorylation, dimerisation, or its transcriptional activity. Tyrosine kinase inhibitors (TKIs) that target molecules upstream of STAT5 could also be utilised. Consequently, since STAT5 contributes to tumour aggressiveness and cancer progression, inhibiting STAT5 constitutive activation in cancers that rely on its signalling makes for a promising targeted treatment option.
Collapse
|
28
|
Hallal R, Nehme R, Brachet-Botineau M, Nehme A, Dakik H, Deynoux M, Dello Sbarba P, Levern Y, Zibara K, Gouilleux F, Mazurier F. Acriflavine targets oncogenic STAT5 signaling in myeloid leukemia cells. J Cell Mol Med 2020; 24:10052-10062. [PMID: 32667731 PMCID: PMC7520299 DOI: 10.1111/jcmm.15612] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Acriflavine (ACF) is an antiseptic with anticancer properties, blocking the growth of solid and haematopoietic tumour cells. Moreover, this compound has been also shown to overcome the resistance of cancer cells to chemotherapeutic agents. ACF has been shown to target hypoxia‐inducible factors (HIFs) activity, which are key effectors of hypoxia‐mediated chemoresistance. In this study, we showed that ACF inhibits the growth and survival of chronic myeloid leukaemia (CML) and acute myeloid leukaemia (AML) cell lines in normoxic conditions. We further demonstrated that ACF down‐regulates STAT5 expression in CML and AML cells but activates STAT3 in CML cells in a HIF‐independent manner. In addition, we demonstrated that ACF suppresses the resistance of CML cells to tyrosine kinase inhibitors, such as imatinib. Our data suggest that the dual effect of ACF might be exploited to eradicate de novo or acquired resistance of myeloid leukaemia cells to chemotherapy.
Collapse
Affiliation(s)
- Rawan Hallal
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France.,PRASE, Lebanese University, Beirut, Lebanon
| | - Rawan Nehme
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France.,PRASE, Lebanese University, Beirut, Lebanon
| | | | - Ali Nehme
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France
| | - Hassan Dakik
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France
| | - Margaux Deynoux
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France
| | - Persio Dello Sbarba
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Yves Levern
- INRAE, Imagerie en Infectiologie, UMR Infectiologie et Santé Publique, Université de Tours, Nouzilly, France
| | - Kazem Zibara
- PRASE, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Fabrice Gouilleux
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France
| | - Frédéric Mazurier
- Université de Tours, EA7501 GICC, Tours, France.,CNRS ERL7001 LNOx, Tours, France
| |
Collapse
|
29
|
Chen XX, Zhu JH, Li ZP, Xiao HT, Zhou H. Comprehensive Characterization of the Prognosis Value of Alternative Splicing Events in Acute Myeloid Leukemia. DNA Cell Biol 2020; 39:1243-1255. [PMID: 32543226 DOI: 10.1089/dna.2020.5534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence have demonstrated that dysregulated alternative splicing (AS) events promoted tumor development and was correlated with worse prognosis in the context of certain malignancies. Nevertheless, a comprehensive examination of the prognosis role of AS events in acute myeloid leukemia (AML) has not yet been illuminated. In this study, univariate and multivariate Cox regression analysis were used to identify survival-related AS events and independent prognostic predictors. The interaction between splicing factors (SFs) and AS events was visualized by Cytoscape. A total of 3013 survival-associated AS events in 1977 genes were screened in 151 AML patients. Interestingly, the majority (2031 events) were revealed to be protective factors. Furthermore, the prediction models were constructed for each type of AS and all of them displayed good performance in predicting prognosis, considering their area under curve values of the receiver operating characteristic were all above 0.7. Notably, the splicing regulatory network displayed the underlying interaction networks between SFs and AS events. Taken together, our study demonstrated the survival-related AS events in AML and uncovered the possible association between SFs and prognostic AS events, which provide new prognostic biomarkers and aid to develop novel targets for AML therapy.
Collapse
Affiliation(s)
- Xue-Xing Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Hua Zhu
- Laboratory of Clinical Immunology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Ping Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Tao Xiao
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Braun TP, Eide CA, Druker BJ. Response and Resistance to BCR-ABL1-Targeted Therapies. Cancer Cell 2020; 37:530-542. [PMID: 32289275 PMCID: PMC7722523 DOI: 10.1016/j.ccell.2020.03.006] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Chronic myeloid leukemia (CML), caused by constitutively active BCR-ABL1 fusion tyrosine kinase, has served as a paradigm for successful application of molecularly targeted cancer therapy. The development of the tyrosine kinase inhibitor (TKI) imatinib allows patients with CML to experience near-normal life expectancy. Specific point mutations that decrease drug binding affinity can produce TKI resistance, and second- and third-generation TKIs largely mitigate this problem. Some patients develop TKI resistance without known resistance mutations, with significant heterogeneity in the underlying mechanism, but this is relatively uncommon, with the majority of patients with chronic phase CML achieving long-term disease control. In contrast, responses to TKI treatment are short lived in advanced phases of the disease or in BCR-ABL1-positive acute lymphoblastic leukemia, with relapse driven by both BCR-ABL1 kinase-dependent and -independent mechanisms. Additionally, the frontline CML treatment with second-generation TKIs produces deeper molecular responses, driving disease burden below the detection limit for a greater number of patients. For patients with deep molecular responses, up to half have been able to discontinue therapy. Current efforts are focused on identifying therapeutic strategies to drive deeper molecular responses, enabling more patients to attempt TKI discontinuation.
Collapse
MESH Headings
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy
- Protein Kinase Inhibitors/therapeutic use
Collapse
Affiliation(s)
- Theodore P Braun
- Division of Hematology/Medical Oncology, Knight Cancer Insitute, Oregon Health & Science University, Portland, OR, USA.
| | - Christopher A Eide
- Division of Hematology/Medical Oncology, Knight Cancer Insitute, Oregon Health & Science University, Portland, OR, USA
| | - Brian J Druker
- Division of Hematology/Medical Oncology, Knight Cancer Insitute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Wang JX, Zhang L, Huang ZW, Zhang XN, Jiang YY, Liu FJ, Long L, Xue MJ, Lu G, Liu Q, Long ZJ. Aurora kinase inhibitor restrains STAT5-activated leukemic cell proliferation by inducing mitochondrial impairment. J Cell Physiol 2020; 235:8358-8370. [PMID: 32239704 DOI: 10.1002/jcp.29680] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
Current chemotherapy regimens on acute myeloid leukemia (AML) still have some drawbacks, such as intolerance and drug resistance, which calls need for the development of targeted therapy. Signal transducer and activator of transcription 5 (STAT5) is often overexpressed or abnormally activated in leukemia and involved in cell self-renewal, proliferation, and stress adaptation. Overexpressed Aurora A (AURKA) is associated with poor prognosis in tumors, and inhibitors against AURKA are already in clinical trials. However, it has rarely been reported whether AURKA inhibitors restrain STAT5-activated leukemia cells. In this study, we constructed STAT5 constitutively activated (cS5) cells and found that STAT5 promoted cell proliferation and colony formation. Moreover, cS5 cells showed elevated reactive oxygen species (ROS) and adenosine triphosphate (ATP) levels, which indicated higher mitochondrial metabolism in cS5 cells. A novel AURKA inhibitor AKI604 was synthesized and showed significant inhibitory effects to the proliferation and colony formation in both STAT5 constitutively activated and nonactivated AML cells. AKI604 induced mitochondrial impairment, leading to the disruption of mitochondrial membrane potential and the elevation of ROS as well as cellular calcium (Ca2+ ) levels. AKI604 could also decline basal oxygen consumption rate and ATP biosynthesis, indicating the damage of oxidative phosphorylation. Furthermore, AKI604 exhibited significant antitumor effect in the HL-60 cS5 xenograft model of the BALB/c nude mice without an obvious influence on mice body weight and other healthy indicators. This study suggested that AKI604 was a potential strategy to overcome STAT5-induced leukemic proliferation in AML treatment by inducing mitochondrial impairment.
Collapse
Affiliation(s)
- Jin-Xing Wang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Ling Zhang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Ze-Wei Huang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Xue-Ning Zhang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Yan-Yan Jiang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Fang-Jie Liu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liang Long
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Man-Jie Xue
- Medical Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gui Lu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Zi-Jie Long
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Identification and development of non-cytotoxic cell death modulators: Impact of sartans and derivatives on PPARγ activation and on growth of imatinib-resistant chronic myelogenous leukemia cells. Eur J Med Chem 2020; 195:112258. [PMID: 32272420 DOI: 10.1016/j.ejmech.2020.112258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022]
Abstract
4'-((2-Propyl-1H-benzo[d]imidazol-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid derived from telmisartan was identified as lead for the design of cell death modulators. In this study, we evaluated the efficacy of telmisartan itself and other sartans in combination with imatinib against K562-resistant cells. The findings were directly used to further optimize the lead structure. Telmisartan and candesartan cilexetil represented the most effective sartans, thus the influence of carboxyl/methyl carboxylate groups at positions 7 (compounds 6, 7) or 4 (compounds 12-14) at the benzimidazole core was studied. Additionally, according to the results of a former structure-activity study, telmisartan was transformed to the related amide (1). Telmisartan amide 1, as well as the esters 6 and 12 markedly sensitized the resistant CML cells to imatinib treatment. Correlation with their potency to activate PPARγ is not given. Candesartan cilexetil, telmisartan and 1 showed the profile of partial agonists at PPARγ with EC50 values of 4.2, 4.3 and 9.1 μM, respectively, while 6 and 12 caused only marginal intrinsic activation at 10 μM (Amax = 22% and 13%). However, the repression of the STAT5 phosphorylation relates with the possibility to sensitize K562-resistant CML cells to imatinib treatment. It is worth mentioning that all compounds were per se non-cytotoxic at relevant concentrations.
Collapse
|
33
|
Brachet-Botineau M, Polomski M, Neubauer HA, Juen L, Hédou D, Viaud-Massuard MC, Prié G, Gouilleux F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers (Basel) 2020; 12:E240. [PMID: 31963765 PMCID: PMC7016966 DOI: 10.3390/cancers12010240] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 and 5 are important effectors of cellular transformation, and aberrant STAT3 and STAT5 signaling have been demonstrated in hematopoietic cancers. STAT3 and STAT5 are common targets for different tyrosine kinase oncogenes (TKOs). In addition, STAT3 and STAT5 proteins were shown to contain activating mutations in some rare but aggressive leukemias/lymphomas. Both proteins also contribute to drug resistance in hematopoietic malignancies and are now well recognized as major targets in cancer treatment. The development of inhibitors targeting STAT3 and STAT5 has been the subject of intense investigations during the last decade. This review summarizes the current knowledge of oncogenic STAT3 and STAT5 functions in hematopoietic cancers as well as advances in preclinical and clinical development of pharmacological inhibitors.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| | - Marion Polomski
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria;
| | - Ludovic Juen
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Damien Hédou
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Marie-Claude Viaud-Massuard
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Gildas Prié
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| |
Collapse
|
34
|
Nie ZY, Yao M, Yang Z, Yang L, Liu XJ, Yu J, Ma Y, Zhang N, Zhang XY, Liu MH, Jiang LL, Luo JM. De-regulated STAT5A/miR-202-5p/USP15/Caspase-6 regulatory axis suppresses CML cell apoptosis and contributes to Imatinib resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:17. [PMID: 31952546 PMCID: PMC6969434 DOI: 10.1186/s13046-019-1502-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/12/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND STAT5 plays an important role in the transformation of hematopoietic cells by BCR-ABL. However, the downstream target genes activated by STAT5 in chronic myeloid leukemia (CML) cells remain largely unclear. Here, we investigated the mechanistic functional relationship between STAT5A-regulated microRNA and CML cell apoptosis. METHODS The expression of USP15, Caspase-6, STAT5A-regulated miR-202-5p and STAT5A was detected by qRT-PCR and Western blotting in CML cell lines and PBMCs of CML patients. Cell apoptosis was evaluated by flow cytometry. Both gain- and loss-of-function experiments were used to investigate the roles of USP15, miR-202-5p and STAT5A in CML. Luciferase reporter assay detected the effect of miR-202-5p on USP15 expression. Xenograft animal model was used to test the effect of anti-miR-202-5p and pimozide on K562 cell xenograft growth. RESULTS USP15 expression was significantly downregulated in CML cell lines and PBMCs of CML patients. Depletion of USP15 increased, whereas overexpression of USP15 reduced the resistance of CML cells to Imatinib. Further, decreased deubiquitinating activity of USP15 by USP15 downregulation led to reduced caspase-6 level, thus attenuating CML cell apoptosis. Mechanistically, miR-202-5p was upregulated in K562G cells and negatively regulated USP15 expression by directly targeting USP15 3'-UTR. Correspondingly, upregulation of miR-202-5p enhanced the resistance of CML cells to Imatinib by inhibiting cell apoptosis. Importantly, STAT5A was upregulated in CML cells and directly activated miR-202-5p transcription by binding to the pre-miR-202 promoter. Pimozide induced CML cell apoptosis and significantly reduced K562 cell xenograft growth in vivo by blocking STAT5A/miR-202-5p/USP15/Caspase-6 regulatory axis. CONCLUSIONS we provide the first evidence that de-regulated STAT5A/miR-202-5p/USP15/Caspase-6 regulatory axis suppresses the apoptosis of CML cells, targeting this pathway might be a promising therapeutic approach for the treatment of CML.
Collapse
Affiliation(s)
- Zi-Yuan Nie
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China
| | - Min Yao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, No. 361 Zhongshan E Rd, Shijiazhuang, 050017, China
| | - Zhan Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, No. 361 Zhongshan E Rd, Shijiazhuang, 050017, China.,Department of Urology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China
| | - Lin Yang
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China
| | - Xiao-Jun Liu
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China
| | - Jing Yu
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, No. 361 Zhongshan E Rd, Shijiazhuang, 050017, China
| | - Ying Ma
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, No. 361 Zhongshan E Rd, Shijiazhuang, 050017, China
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, No. 361 Zhongshan E Rd, Shijiazhuang, 050017, China
| | - Xiao-Yan Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China
| | - Meng-Han Liu
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China
| | - Ling-Ling Jiang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, No. 361 Zhongshan E Rd, Shijiazhuang, 050017, China.
| | - Jian-Min Luo
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping W Rd, Shijiazhuang, 050000, China.
| |
Collapse
|
35
|
Brachet-Botineau M, Deynoux M, Vallet N, Polomski M, Juen L, Hérault O, Mazurier F, Viaud-Massuard MC, Prié G, Gouilleux F. A Novel Inhibitor of STAT5 Signaling Overcomes Chemotherapy Resistance in Myeloid Leukemia Cells. Cancers (Basel) 2019; 11:cancers11122043. [PMID: 31861239 PMCID: PMC6966442 DOI: 10.3390/cancers11122043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/11/2019] [Accepted: 12/14/2019] [Indexed: 01/08/2023] Open
Abstract
Signal transducers and activators of transcription 5A and 5B (STAT5A and STAT5B) are crucial downstream effectors of tyrosine kinase oncogenes (TKO) such as BCR-ABL in chronic myeloid leukemia (CML) and FLT3-ITD in acute myeloid leukemia (AML). Both proteins have been shown to promote the resistance of CML cells to tyrosine kinase inhibitors (TKI) such as imatinib mesylate (IM). We recently synthesized and discovered a new inhibitor (17f) with promising antileukemic activity. 17f selectively inhibits STAT5 signaling in CML and AML cells by interfering with the phosphorylation and transcriptional activity of these proteins. In this study, the effects of 17f were evaluated on CML and AML cell lines that respectively acquired resistance to IM and cytarabine (Ara-C), a conventional therapeutic agent used in AML treatment. We showed that 17f strongly inhibits the growth and survival of resistant CML and AML cells when associated with IM or Ara-C. We also obtained evidence that 17f inhibits STAT5B but not STAT5A protein expression in resistant CML and AML cells. Furthermore, we demonstrated that 17f also targets oncogenic STAT5B N642H mutant in transformed hematopoietic cells.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
| | - Margaux Deynoux
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
| | - Nicolas Vallet
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
- Service d’Hématologie et Thérapie Cellulaire, CHRU de Tours, 37000 Tours, France
| | - Marion Polomski
- IMT, GICC, EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (M.-C.V.-M.); (G.P.)
| | - Ludovic Juen
- IMT, GICC, EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (M.-C.V.-M.); (G.P.)
| | - Olivier Hérault
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
- Service d’Hematologie Biologique, CHRU de Tours, 37000 Tours, France
| | - Frédéric Mazurier
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
| | | | - Gildas Prié
- IMT, GICC, EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
- Correspondence: ; Tel.: +33-(2)-47-36-62-91
| |
Collapse
|
36
|
de Araujo ED, Orlova A, Neubauer HA, Bajusz D, Seo HS, Dhe-Paganon S, Keserű GM, Moriggl R, Gunning PT. Structural Implications of STAT3 and STAT5 SH2 Domain Mutations. Cancers (Basel) 2019; 11:E1757. [PMID: 31717342 PMCID: PMC6895964 DOI: 10.3390/cancers11111757] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 01/13/2023] Open
Abstract
Src Homology 2 (SH2) domains arose within metazoan signaling pathways and are involved in protein regulation of multiple pleiotropic cascades. In signal transducer and activator of transcription (STAT) proteins, SH2 domain interactions are critical for molecular activation and nuclear accumulation of phosphorylated STAT dimers to drive transcription. Sequencing analysis of patient samples has revealed the SH2 domain as a hotspot in the mutational landscape of STAT proteins although the functional impact for the vast majority of these mutations remains poorly characterized. Despite several well resolved structures for SH2 domain-containing proteins, structural data regarding the distinctive STAT-type SH2 domain is limited. Here, we review the unique features of STAT-type SH2 domains in the context of all currently reported STAT3 and STAT5 SH2 domain clinical mutations. The genetic volatility of specific regions in the SH2 domain can result in either activating or deactivating mutations at the same site in the domain, underscoring the delicate evolutionary balance of wild type STAT structural motifs in maintaining precise levels of cellular activity. Understanding the molecular and biophysical impact of these disease-associated mutations can uncover convergent mechanisms of action for mutations localized within the STAT SH2 domain to facilitate the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Elvin D. de Araujo
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada;
- Department of Chemical & Physical Sciences, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria; (A.O.); (H.A.N.); (R.M.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria; (A.O.); (H.A.N.); (R.M.)
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Center for Natural Sciences, 1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; (H.-S.S.); (S.D.-P.)
- Department of Biological Chemistry, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; (H.-S.S.); (S.D.-P.)
- Department of Biological Chemistry, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - György M. Keserű
- Medicinal Chemistry Research Group, Research Center for Natural Sciences, 1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria; (A.O.); (H.A.N.); (R.M.)
| | - Patrick T. Gunning
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada;
- Department of Chemical & Physical Sciences, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
37
|
Maurer B, Kollmann S, Pickem J, Hoelbl-Kovacic A, Sexl V. STAT5A and STAT5B-Twins with Different Personalities in Hematopoiesis and Leukemia. Cancers (Basel) 2019; 11:E1726. [PMID: 31690038 PMCID: PMC6895831 DOI: 10.3390/cancers11111726] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factors STAT5A and STAT5B have essential roles in survival and proliferation of hematopoietic cells-which have been considered largely redundant. Mutations of upstream kinases, copy number gains, or activating mutations in STAT5A, or more frequently in STAT5B, cause altered hematopoiesis and cancer. Interfering with their activity by pharmacological intervention is an up-and-coming therapeutic avenue. Precision medicine requests detailed knowledge of STAT5A's and STAT5B's individual functions. Recent evidence highlights the privileged role for STAT5B over STAT5A in normal and malignant hematopoiesis. Here, we provide an overview on their individual functions within the hematopoietic system.
Collapse
Affiliation(s)
- Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Judith Pickem
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
38
|
Tolomeo M, Meli M, Grimaudo S. STAT5 and STAT5 Inhibitors in Hematological Malignancies. Anticancer Agents Med Chem 2019; 19:2036-2046. [PMID: 31490767 DOI: 10.2174/1871520619666190906160848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/09/2019] [Accepted: 07/18/2019] [Indexed: 11/22/2022]
Abstract
The JAK-STAT pathway is an important physiologic regulator of different cellular functions including proliferation, apoptosis, differentiation, and immunological responses. Out of six different STAT proteins, STAT5 plays its main role in hematopoiesis and constitutive STAT5 activation seems to be a key event in the pathogenesis of several hematological malignancies. This has led many researchers to develop compounds capable of inhibiting STAT5 activation or interfering with its functions. Several anti-STAT5 molecules have shown potent STAT5 inhibitory activity in vitro. However, compared to the large amount of clinical studies with JAK inhibitors that are currently widely used in the clinics to treat myeloproliferative disorders, the clinical trials with STAT5 inhibitors are very limited. At present, a few STAT5 inhibitors are in phase I or II clinical trials for the treatment of leukemias and graft vs host disease. These studies seem to indicate that such compounds could be well tolerated and useful in reducing the occurrence of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia. Of interest, STAT5 seems to play an important role in the regulation of hematopoietic stem cell self-renewal suggesting that combination therapies including STAT5 inhibitors can erode the cancer stem cell pool and possibly open the way for the complete cancer eradication. In this review, we discuss the implication of STAT5 in hematological malignancies and the results obtained with the novel STAT5 inhibitors.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Maria Meli
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Stefania Grimaudo
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
39
|
Memarzadeh K, Savage DJ, Bean AJ. Low UBE4B expression increases sensitivity of chemoresistant neuroblastoma cells to EGFR and STAT5 inhibition. Cancer Biol Ther 2019; 20:1416-1429. [PMID: 31475882 PMCID: PMC6804809 DOI: 10.1080/15384047.2019.1647049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 05/24/2019] [Accepted: 06/23/2019] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma is the most common malignancy in infants. Overexpression of the epidermal growth factor receptor (EGFR) in neuroblastoma tumors underlies resistance to chemotherapeutics. UBE4B, an E3/E4 ubiquitin ligase involved in EGFR degradation, is located on chromosome 1p36, a region in which loss of heterozygosity is observed in approximately one-third of neuroblastoma tumors and is correlated with poor prognosis. In chemoresistant neuroblastoma cells, depletion of UBE4B yielded significantly reduced cell proliferation and migration, and enhanced apoptosis in response to EGFR inhibitor, Cetuximab. We have previously shown that UBE4B levels are inversely correlated with EGFR levels in neuroblastoma tumors. We searched for additional targets of UBE4B that mediate cellular alterations associated with tumorogenesis in chemoresistant neuroblastoma cells depleted of UBE4B using reverse phase protein arrays. The expression of STAT5a, an effector protein downstream of EGFR, doubled in the absence of UBE4B, and verified by quantitative immunoblotting. Chemoresistant neuroblastoma cells were treated with SH-4-54, a STAT5 inhibitor, and observed insignificant effects on cell proliferation, migration, and apoptosis. However, SH-4-54 significantly enhanced the anti-proliferative and anti-migratory effects of Cetuximab in naïve SK-N-AS neuroblastoma cells. Interestingly, in UBE4B depleted SK-N-AS cells, SH-4-54 significantly potentiated the effect of Cetuximab rendering cells increasingly sensitive an otherwise minimally effective Cetuximab concentration. Thus, neuroblastoma cells with low UBE4B levels were significantly more sensitive to combined EGFR and STAT5 inhibition than parental cells. These findings may have potential therapeutic implications for patients with 1p36 chromosome LOH and low tumor UBE4B expression.
Collapse
Affiliation(s)
- Kimiya Memarzadeh
- Program in Neuroscience, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - David J. Savage
- Program in Neuroscience, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Andrew J. Bean
- Program in Neuroscience, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX, USA
- Program in Neuroscience, Cell Biology and Biochemistry, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
40
|
Kollmann S, Grundschober E, Maurer B, Warsch W, Grausenburger R, Edlinger L, Huuhtanen J, Lagger S, Hennighausen L, Valent P, Decker T, Strobl B, Mueller M, Mustjoki S, Hoelbl-Kovacic A, Sexl V. Twins with different personalities: STAT5B-but not STAT5A-has a key role in BCR/ABL-induced leukemia. Leukemia 2019; 33:1583-1597. [PMID: 30679796 PMCID: PMC6755975 DOI: 10.1038/s41375-018-0369-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 12/03/2018] [Indexed: 01/12/2023]
Abstract
Deregulation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway is found in cancer with STAT5A/B controlling leukemic cell survival and disease progression. As mutations in STAT5B, but not STAT5A, have been frequently described in hematopoietic tumors, we used BCR/ABL as model systems to investigate the contribution of STAT5A or STAT5B for leukemogenesis. The absence of STAT5A decreased cell survival and colony formation. Even more drastic effects were observed in the absence of STAT5B. STAT5B-deficient cells formed BCR/ABL+ colonies or stable cell lines at low frequency. The rarely evolving Stat5b-/- cell lines expressed enhanced levels of BCR/ABL oncoprotein compared to wild-type cells. In line, Stat5b-/- leukemic cells induced leukemia with a significantly prolonged disease onset, whereas Stat5a-/- cells rapidly caused a fatal disease superimposable to wild-type cells. RNA-sequencing (RNA-seq) profiling revealed a marked enhancement of interferon (IFN)-α and IFN-γ signatures in Stat5b-/- cells. Inhibition of IFN responses rescued BCR/ABL+ colony formation of Stat5b-/--deficient cells. A downregulated IFN response was also observed in patients suffering from leukemia carrying STAT5B mutations. Our data define STAT5B as major STAT5 isoform driving BCR/ABL+ leukemia. STAT5B enables transformation by suppressing IFN-α/γ, thereby facilitating leukemogenesis. Our findings might help explain the high frequency of STAT5B mutations in hematopoietic tumors.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Cell Proliferation
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Interferons/pharmacology
- Leukemia, Large Granular Lymphocytic/drug therapy
- Leukemia, Large Granular Lymphocytic/metabolism
- Leukemia, Large Granular Lymphocytic/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Mutation
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/metabolism
- Survival Rate
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Eva Grundschober
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Wolfgang Warsch
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Leo Edlinger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, P.O.Box 700, 00290, Helsinki, Finland
| | - Sabine Lagger
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Decker
- Max F. Perutz Laboratories (MFPL), University of Vienna, 1030, Vienna, Austria
| | - Birgit Strobl
- Department for Biomedical Sciences Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Mathias Mueller
- Department for Biomedical Sciences Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, P.O.Box 700, 00290, Helsinki, Finland
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria.
| |
Collapse
|
41
|
Arrington J, Xue L, Wang WH, Geahlen RL, Tao WA. Identification of the Direct Substrates of the ABL Kinase via Kinase Assay Linked Phosphoproteomics with Multiple Drug Treatments. J Proteome Res 2019; 18:1679-1690. [PMID: 30869898 DOI: 10.1021/acs.jproteome.8b00942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ableson tyrosine kinase (ABL) plays essential roles in cell differentiation, division, adhesion, and stress response. However, fusion of the breakpoint cluster region (BCR) to ABL produces constitutive kinase activity that causes chronic myelogenous leukemia (CML). Small molecule tyrosine kinase inhibitors (TKIs) such as imatinib revolutionized the treatment of CML and other cancers, but acquired resistance to these inhibitors is rising. Thus, careful dissection of ABL signaling pathways is needed to find novel drug targets. Here we present a refined proteomic approach for elucidation of direct kinase substrates called kinase assay linked phosphoproteomics (KALIP). Our strategy integrates in vitro kinase assays at both the peptide and protein levels with quantitative tyrosine phosphoproteomics in response to treatment by multiple TKIs. Utilizing multiple TKIs permits elimination of off-target effects of these drugs, and overlapping the in vivo and in vitro data sets allows us to define a list of the most probable kinase substrates. Applying our approach produced a list of 60 ABL substrates, including novel and known proteins. We demonstrate that spleen tyrosine kinase (SYK) is a novel direct substrate of ABL, and we predict our proteomic strategy may facilitate identification of substrates in other cancers that have disrupted kinase signaling.
Collapse
|
42
|
Niogret C, Miah SMS, Rota G, Fonta NP, Wang H, Held W, Birchmeier W, Sexl V, Yang W, Vivier E, Ho PC, Brossay L, Guarda G. Shp-2 is critical for ERK and metabolic engagement downstream of IL-15 receptor in NK cells. Nat Commun 2019; 10:1444. [PMID: 30926899 PMCID: PMC6441079 DOI: 10.1038/s41467-019-09431-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
The phosphatase Shp-2 was implicated in NK cell development and functions due to its interaction with NK inhibitory receptors, but its exact role in NK cells is still unclear. Here we show, using mice conditionally deficient for Shp-2 in the NK lineage, that NK cell development and responsiveness are largely unaffected. Instead, we find that Shp-2 serves mainly to enforce NK cell responses to activation by IL-15 and IL-2. Shp-2-deficient NK cells have reduced proliferation and survival when treated with high dose IL-15 or IL-2. Mechanistically, Shp-2 deficiency hampers acute IL-15 stimulation-induced raise in glycolytic and respiration rates, and causes a dramatic defect in ERK activation. Moreover, inhibition of the ERK and mTOR cascades largely phenocopies the defect observed in the absence of Shp-2. Together, our data reveal a critical function of Shp-2 as a molecular nexus bridging acute IL-15 signaling with downstream metabolic burst and NK cell expansion.
Collapse
Affiliation(s)
- Charlène Niogret
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - S M Shahjahan Miah
- Department of Molecular Microbiology and Immunology and Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Giorgia Rota
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - Nicolas P Fonta
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland.,Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500, Bellinzona, Switzerland
| | - Haiping Wang
- Department of Oncology UNIL CHUV, University of Lausanne, 1066, Epalinges, Switzerland.,Department of Fundamental Oncology, University of Lausanne, 1066, Epalinges, Switzerland
| | - Werner Held
- Department of Oncology UNIL CHUV, University of Lausanne, 1066, Epalinges, Switzerland
| | - Walter Birchmeier
- Max-Delbrueck-Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125, Berlin, Germany
| | - Veronica Sexl
- Department for Biomedical Sciences, Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Wentian Yang
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Avenue de Luminy, 13288, Marseille, France.,Service d'Immunologie, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, 13385, Marseille, France.,Innate Pharma Research Labs., Innate Pharma, 117 Avenue de Luminy, 13276, Marseille, France
| | - Ping-Chih Ho
- Department of Oncology UNIL CHUV, University of Lausanne, 1066, Epalinges, Switzerland.,Department of Fundamental Oncology, University of Lausanne, 1066, Epalinges, Switzerland
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology and Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, 02912, USA.
| | - Greta Guarda
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland. .,Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500, Bellinzona, Switzerland.
| |
Collapse
|
43
|
Chan O, Talati C, Sweet K, Pinilla-Ibarz J. Can increased immunogenicity in chronic myeloid leukemia improve outcomes? Expert Rev Hematol 2019; 12:225-233. [PMID: 30855193 DOI: 10.1080/17474086.2019.1588105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Chronic myeloid leukemia (CML) has long been thought to be the model disease for immunotherapy with its characteristic BCR-ABL fusion protein. Although targeted therapy using tyrosine kinase inhibitors (TKIs) is highly effective at inducing remission, most patients require life-long TKI to decrease the risk of relapse. In recent years, much effort has been devoted to finding ways to eliminate CML stem cells (LSCs); the source of disease persistence. Areas covered: In this review, the authors present recent immunologic findings pertinent to CML, vaccinations targeting leukemia antigens, interferon combination therapies, and other emerging strategies aimed at increasing immunogenicity and improving outcomes in patients with CML. Recent publications and abstracts found in Pubmed and hematology/oncology meetings related to these topics were identified and incorporated into this review. Expert commentary: Further understanding of the immune system and antigenic composition of LSCs has allowed for novel therapeutic development. Immunotherapies are effective at the malignant stem cell level and combining these approaches with TKI is a promising option. Despite ongoing challenges, it is increasingly recognized that a cure may be achievable through immunotherapies.
Collapse
Affiliation(s)
- Onyee Chan
- a Moffitt Cancer Center , University of South Florida , Tampa , FL , USA
| | - Chetasi Talati
- b Division of Malignant Hematology , Moffitt Cancer Center , Tampa , FL , USA
| | - Kendra Sweet
- b Division of Malignant Hematology , Moffitt Cancer Center , Tampa , FL , USA
| | | |
Collapse
|
44
|
Recio C, Guerra B, Guerra-Rodríguez M, Aranda-Tavío H, Martín-Rodríguez P, de Mirecki-Garrido M, Brito-Casillas Y, García-Castellano JM, Estévez-Braun A, Fernández-Pérez L. Signal transducer and activator of transcription (STAT)-5: an opportunity for drug development in oncohematology. Oncogene 2019; 38:4657-4668. [DOI: 10.1038/s41388-019-0752-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/09/2019] [Accepted: 02/03/2019] [Indexed: 02/08/2023]
|
45
|
Martín-Rodríguez P, Guerra B, Hueso-Falcón I, Aranda-Tavío H, Díaz-Chico J, Quintana J, Estévez F, Díaz-Chico B, Amesty A, Estévez-Braun A, Fernández-Pérez L. A Novel Naphthoquinone-Coumarin Hybrid That Inhibits BCR-ABL1-STAT5 Oncogenic Pathway and Reduces Survival in Imatinib-Resistant Chronic Myelogenous Leukemia Cells. Front Pharmacol 2019; 9:1546. [PMID: 30687103 PMCID: PMC6334626 DOI: 10.3389/fphar.2018.01546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 12/18/2018] [Indexed: 01/13/2023] Open
Abstract
BCR-ABL1-STAT5 is an oncogenic signaling pathway in human chronic myelogenous leukemia (CML) and it represents a valid target for anti-CML drug design. Resistance to direct BCR-ABL1 inhibitors is a common clinical issue, so STAT5 inhibition has become an interesting alternative target. In this study, the effects of NPQ-C6, a novel naphtoquinone-coumarin conjugate, were evaluated on human CML-derived K562 cells. Live-Cell Imaging analysis revealed that NPQ-C6 inhibited 2D (IC50AUC = 1.4 ± 0.6 μM) growth of CML cells. NPQ-C6 increased sub-G1 and reduced G0/G1 cell cycle phases in a dose- and time-dependent manner. This effect on cell cycle was related to increased levels of apoptotic nuclei, cleavage of caspase-3, -9, and PARP and annexin V-positive cells. NPQ-C6 increased γH2AX, a double-strand DNA break marker. NPQ-C6 showed a wide range of modulatory effects on cell signaling through an early increased phosphorylation of JNK, P38-MAPK and AKT, and decreased phosphorylation of ERK1/2, BCR-ABL1, and STAT5. NPQ-C6 inhibited expression of c-MYC and PYM-1, two target gene products of BCR-ABL1/STAT5 signaling pathway. Cytokine-induced activation of STAT5/STAT3-dependent transcriptional and DNA binding activities were also inhibited by NPQ-C6. Notably, NPQ-C6 maintained its activity on BCR-ABL1/STAT5/c-MYC/PIM-1 oncogenic pathway in imatinib-resistant cells. Molecular modeling suggested BCR-ABL1 and JAK2 proteins as NPQ-C6 targets. In summary, our data show a novel multikinase modulator that might be therapeutically effective in BCR-ABL1-STAT5-related malignancies.
Collapse
Affiliation(s)
- Patricia Martín-Rodríguez
- Laboratorio de Farmacología Molecular y Traslacional, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Borja Guerra
- Laboratorio de Farmacología Molecular y Traslacional, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Idaira Hueso-Falcón
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Haidee Aranda-Tavío
- Laboratorio de Farmacología Molecular y Traslacional, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Juan Díaz-Chico
- Laboratorio de Farmacología Molecular y Traslacional, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - José Quintana
- Laboratorio de Bioquímica, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Francisco Estévez
- Laboratorio de Bioquímica, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Bonifacio Díaz-Chico
- Laboratorio de Farmacología Molecular y Traslacional, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Angel Amesty
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Ana Estévez-Braun
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Leandro Fernández-Pérez
- Laboratorio de Farmacología Molecular y Traslacional, Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| |
Collapse
|
46
|
Jiang X, Cheng Y, Hu C, Zhang A, Ren Y, Xu X. MicroRNA-221 sensitizes chronic myeloid leukemia cells to imatinib by targeting STAT5. Leuk Lymphoma 2018; 60:1709-1720. [PMID: 30516071 DOI: 10.1080/10428194.2018.1543875] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRNAs) are involved in various processes from the development to drug resistance of tumors, including chronic myeloid leukemia (CML). In this study, we examined the STAT5-related miRNA-expression profile in CML cell lines (K562 and imatinib-resistant K562/G) by quantitative real-time reverse-transcriptase polymerase chain reactions. MiR-221 expression was markedly decreased in K562/G cells and peripheral blood mononuclear cells from patients with treatment failure, when compared to imatinib-sensitive CML cells and patients with optimal responses respectively. We also observed the expression of STAT5 inversely correlated with miR-221 expression in K562 and KBM5 cells. Additionally, STAT5 was validated as a direct target of miR-221 in dual-luciferase reporter vector assays. MiR-221 restoration and STAT5 knockdown in K562/G cells increased the sensitivity of CML cells to imatinib by reducing the Bcl2: Bax ratio. Collectively, our data suggested that miR-221-STAT5 axis played crucial roles in controlling the sensitivity of CML cells to imatinib.
Collapse
Affiliation(s)
- Xiaoxiao Jiang
- a Central Laboratory , Anhui Provincial Hospital, Anhui Medical University , Hefei , China
| | - Yanhong Cheng
- a Central Laboratory , Anhui Provincial Hospital, Anhui Medical University , Hefei , China
| | - Chaojie Hu
- a Central Laboratory , Anhui Provincial Hospital, Anhui Medical University , Hefei , China
| | - Aimei Zhang
- a Central Laboratory , Anhui Provincial Hospital, Anhui Medical University , Hefei , China
| | - Yingli Ren
- a Central Laboratory , Anhui Provincial Hospital, Anhui Medical University , Hefei , China
| | - Xiucai Xu
- a Central Laboratory , Anhui Provincial Hospital, Anhui Medical University , Hefei , China
| |
Collapse
|
47
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
48
|
Predicting Tumor Sensitivity to Chemotherapeutic Drugs in Oral Squamous Cell Carcinoma Patients. Sci Rep 2018; 8:15545. [PMID: 30341378 PMCID: PMC6195614 DOI: 10.1038/s41598-018-33998-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/10/2018] [Indexed: 02/01/2023] Open
Abstract
Oral Squamous Cell Carcinoma (OSCC) patients respond poorly to chemotherapy. We analyzed the expression of 11 drug response-related genes in 31 OSCC biopsies, collected prior to any treatment, using custom-designed PCR array. Further, we investigated the drug response pattern of selected anticancer drugs by BH3 (Bcl2 Homology-3) profiling in the primary cells isolated from OSCC tissues. Then, we correlated the results of drug-response gene expression pattern with apoptotic priming to predict tumor response to chemotherapy. The best performing drug (BPD) and response differences (RD) between the drugs were identified using statistical methods to select the best choice of drug in a personalized manner. Based on the correlation, we classified OSCC tumors as sensitive (13 tumors), moderately responsive (16 tumors) or resistant (2 tumors) to chemotherapy. We found that up-regulation of genes linked with drug resistance facilitates survival of tumor samples, which was revealed by the percentage of apoptotic priming. Moreover, we found that paclitaxel-induced 40–45% apoptotic priming compared to other drugs. Average response difference (RD) analysis showed that 80% of tumors responded well to paclitaxel as compared to other drugs studied. Therefore, gene expression analysis with BH3 profiling reveals drug sensitivity that could be translated for drug selection before treatment.
Collapse
|
49
|
The Cooperative Relationship between STAT5 and Reactive Oxygen Species in Leukemia: Mechanism and Therapeutic Potential. Cancers (Basel) 2018; 10:cancers10100359. [PMID: 30262727 PMCID: PMC6210354 DOI: 10.3390/cancers10100359] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are now recognized as important second messengers with roles in many aspects of signaling during leukemogenesis. They serve as critical cell signaling molecules that regulate the activity of various enzymes including tyrosine phosphatases. ROS can induce inactivation of tyrosine phosphatases, which counteract the effects of tyrosine kinases. ROS increase phosphorylation of many proteins including signal transducer and activator of transcription-5 (STAT5) via Janus kinases (JAKs). STAT5 is aberrantly activated through phosphorylation in many types of cancer and this constitutive activation is associated with cell survival, proliferation, and self-renewal. Such leukemic activation of STAT5 is rarely caused by mutation of the STAT5 gene itself but instead by overactive mutant receptors with tyrosine kinase activity as well as JAK, SRC family protein tyrosine kinases (SFKs), and Abelson murine leukemia viral oncogene homolog (ABL) kinases. Interestingly, STAT5 suppresses transcription of several genes encoding antioxidant enzymes while simultaneously enhancing transcription of NADPH oxidase. By doing so, STAT5 activation promotes an overall elevation of ROS level, which acts as a feed-forward loop, especially in high risk Fms-related tyrosine kinase 3 (FLT3) mutant leukemia. Therefore, efforts have been made recently to target ROS in cancer cells. Drugs that are able to either quench ROS production or inversely augment ROS-related signaling pathways both have potential as cancer therapies and may afford some selectivity by activating feedback inhibition of the ROS-STAT5 kinome. This review summarizes the cooperative relationship between ROS and STAT5 and explores the pros and cons of emerging ROS-targeting therapies that are selective for leukemia characterized by persistent STAT5 phosphorylation.
Collapse
|
50
|
Minieri V, De Dominici M, Porazzi P, Mariani SA, Spinelli O, Rambaldi A, Peterson LF, Porcu P, Nevalainen MT, Calabretta B. Targeting STAT5 or STAT5-Regulated Pathways Suppresses Leukemogenesis of Ph+ Acute Lymphoblastic Leukemia. Cancer Res 2018; 78:5793-5807. [PMID: 30154155 DOI: 10.1158/0008-5472.can-18-0195] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/25/2018] [Accepted: 08/21/2018] [Indexed: 12/16/2022]
Abstract
Combining standard cytotoxic chemotherapy with BCR-ABL1 tyrosine kinase inhibitors (TKI) has greatly improved the upfront treatment of patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). However, due to the development of drug resistance through both BCR-ABL1-dependent and -independent mechanisms, prognosis remains poor. The STAT5 transcription factor is activated by BCR-ABL1 and by JAK2-dependent cytokine signaling; therefore, inhibiting its activity could address both mechanisms of resistance in Ph+ ALL. We show here that genetic and pharmacologic inhibition of STAT5 activity suppresses cell growth, induces apoptosis, and inhibits leukemogenesis of Ph+ cell lines and patient-derived newly diagnosed and relapsed/TKI-resistant Ph+ ALL cells ex vivo and in mouse models. STAT5 silencing decreased expression of the growth-promoting PIM-1 kinase, the apoptosis inhibitors MCL1 and BCL2, and increased expression of proapoptotic BIM protein. The resulting apoptosis of STAT5-silenced Ph+ BV173 cells was rescued by silencing of BIM or restoration of BCL2 expression. Treatment of Ph+ ALL cells, including samples from relapsed/refractory patients, with the PIM kinase inhibitor AZD1208 and/or the BCL2 family antagonist Sabutoclax markedly suppressed cell growth and leukemogenesis ex vivo and in mice. Together, these studies indicate that targeting STAT5 or STAT5-regulated pathways may provide a new approach for therapy development in Ph+ ALL, especially the relapsed/TKI-resistant disease.Significance: Suppression of STAT5 by BCL2 and PIM kinase inhibitors reduces leukemia burden in mice and constitutes a new potential therapeutic approach against Ph+ ALL, especially in tyrosine kinase inhibitor-resistant disease. Cancer Res; 78(20); 5793-807. ©2018 AACR.
Collapse
Affiliation(s)
- Valentina Minieri
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Marco De Dominici
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Patrizia Porazzi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Samanta A Mariani
- The Queen's Medical Research Institute, Centre for Inflammation Research, The University of Edinburgh, Scotland, United Kingdom
| | - Orietta Spinelli
- Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo, Italy.,Universita' Statale Milano, Italy
| | - Luke F Peterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Pierluigi Porcu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Marja T Nevalainen
- Department of Pathology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|