1
|
Pamuk GE, Ehrlich LA. An Overview of Myeloid Blast-Phase Chronic Myeloid Leukemia. Cancers (Basel) 2024; 16:3615. [PMID: 39518058 PMCID: PMC11545322 DOI: 10.3390/cancers16213615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Myeloid blast-phase chronic myeloid leukemia (MBP-CML) is a rare disease with a dismal prognosis. It is twice as common as lymphoid blast-phase CML, and its prognosis is poorer. Despite the success with tyrosine kinase inhibitors in the treatment of chronic-phase CML, the same does not hold true for MBP-CML. In addition to the Philadelphia chromosome, other chromosomal and molecular changes characterize rapid progression. Although some progress in elucidating the biology of MBP-CML has been made, there is need to discover more in order to develop more satisfactory treatment options. Currently, most common treatment options include tyrosine kinase inhibitors (TKIs) as monotherapy or in combination with acute myeloid leukemia-based intensive chemotherapy regimens. Some patients may develop resistance to TKIs via BCR-ABL1-dependent or BCR-ABL1-independent mechanisms. In this paper, we provide an overview of the biology of MBP-CML, the current treatment approaches, and mechanisms of resistance to TKIs. In order to improve treatment responses in these patients, more emphasis should be placed on understanding the biology of myeloid blastic transformation in CML and mechanisms of resistance to TKIs. Although patient numbers are small, randomized clinical trials should be considered.
Collapse
Affiliation(s)
- Gulsum E. Pamuk
- Office of Oncologic Diseases, Center for Drug Evaluation and Research—CDER, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA;
| | | |
Collapse
|
2
|
Wei X, Xiong X, Wang P, Zhang S, Peng D. SIRT1-mediated deacetylation of FOXO3 enhances mitophagy and drives hormone resistance in endometrial cancer. Mol Med 2024; 30:147. [PMID: 39266959 PMCID: PMC11391609 DOI: 10.1186/s10020-024-00915-7] [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: 03/28/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND The complex interplay between Sirtuin 1 (SIRT1) and FOXO3 in endometrial cancer (EC) remains understudied. This research aims to unravel the interactions of deacetylase SIRT1 and transcription factor FOXO3 in EC, focusing on their impact on mitophagy and hormone resistance. METHODS High-throughput sequencing, cell experiments, and bioinformatics tools were employed to investigate the roles and interactions of SIRT1 and FOXO3 in EC. Co-immunoprecipitation (Co-IP) assay was used to assess the interaction between SIRT1 and FOXO3 in RL95-2 cells. Functional assays were used to assess cell viability, proliferation, migration, invasion, apoptosis, and the expression of related genes and proteins. A mouse model of EC was established to evaluate tumor growth and hormone resistance under different interventions. Immunohistochemistry and TUNEL assays were used to assess protein expression and apoptosis in tumor tissues. RESULTS High-throughput transcriptome sequencing revealed a close association between SIRT1, FOXO3, and EC development. Co-IP showed a protein-protein interaction between SIRT1 and FOXO3. Overexpression of SIRT1 enhanced FOXO3 deacetylation and activity, promoting BNIP3 transcription and PINK1/Parkin-mediated mitophagy, which in turn promoted cell proliferation, migration, invasion, and inhibited apoptosis in vitro, as well as increased tumor growth and hormone resistance in vivo. These findings highlighted SIRT1 as an upstream regulator and potential therapeutic target in EC. CONCLUSION This study reveals a novel molecular mechanism underlying the functional relevance of SIRT1 in regulating mitophagy and hormone resistance through the deacetylation of FOXO3 in EC, thereby providing valuable insights for new therapeutic strategies.
Collapse
Affiliation(s)
- Xuehua Wei
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, No. 253, Industry Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Xiangpeng Xiong
- Department of Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang, 336000, China
| | - Pingping Wang
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, No. 253, Industry Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Shufang Zhang
- Department of Gynecology, Southern University of Science and Technology Hospital, Shenzhen, 518000, China
| | - Dongxian Peng
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, No. 253, Industry Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
3
|
Li X, Li W, Zhang Y, Xu L, Song Y. Exploiting the potential of the ubiquitin-proteasome system in overcoming tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Genes Dis 2024; 11:101150. [PMID: 38947742 PMCID: PMC11214299 DOI: 10.1016/j.gendis.2023.101150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/15/2023] [Accepted: 09/01/2023] [Indexed: 07/02/2024] Open
Abstract
The advent of tyrosine kinase inhibitors (TKI) targeting BCR-ABL has drastically changed the treatment approach of chronic myeloid leukemia (CML), greatly prolonged the life of CML patients, and improved their prognosis. However, TKI resistance is still a major problem with CML patients, reducing the efficacy of treatment and their quality of life. TKI resistance is mainly divided into BCR-ABL-dependent and BCR-ABL-independent resistance. Now, the main clinical strategy addressing TKI resistance is to switch to newly developed TKIs. However, data have shown that these new drugs may cause serious adverse reactions and intolerance and cannot address all resistance mutations. Therefore, finding new therapeutic targets to overcome TKI resistance is crucial and the ubiquitin-proteasome system (UPS) has emerged as a focus. The UPS mediates the degradation of most proteins in organisms and controls a wide range of physiological processes. In recent years, the study of UPS in hematological malignant tumors has resulted in effective treatments, such as bortezomib in the treatment of multiple myeloma and mantle cell lymphoma. In CML, the components of UPS cooperate or antagonize the efficacy of TKI by directly or indirectly affecting the ubiquitination of BCR-ABL, interfering with CML-related signaling pathways, and negatively or positively affecting leukemia stem cells. Some of these molecules may help overcome TKI resistance and treat CML. In this review, the mechanism of TKI resistance is briefly described, the components of UPS are introduced, existing studies on UPS participating in TKI resistance are listed, and UPS as the therapeutic target and strategies are discussed.
Collapse
Affiliation(s)
- Xudong Li
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yanli Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Linping Xu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
4
|
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
|
5
|
Heo SJ, Enriquez LD, Federman S, Chang AY, Mace R, Shevade K, Nguyen P, Litterman AJ, Shafer S, Przybyla L, Chow ED. Compact CRISPR genetic screens enabled by improved guide RNA library cloning. Genome Biol 2024; 25:25. [PMID: 38243310 PMCID: PMC10797759 DOI: 10.1186/s13059-023-03132-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/29/2023] [Indexed: 01/21/2024] Open
Abstract
CRISPR genome editing approaches theoretically enable researchers to define the function of each human gene in specific cell types, but challenges remain to efficiently perform genetic perturbations in relevant models. In this work, we develop a library cloning protocol that increases sgRNA uniformity and greatly reduces bias in existing genome-wide libraries. We demonstrate that our libraries can achieve equivalent or better statistical power compared to previously reported screens using an order of magnitude fewer cells. This improved cloning protocol enables genome-scale CRISPR screens in technically challenging cell models and screen formats.
Collapse
Affiliation(s)
- Seok-Jin Heo
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Lauren D Enriquez
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Scot Federman
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Amy Y Chang
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Rachel Mace
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Kaivalya Shevade
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Phuong Nguyen
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Adam J Litterman
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Shawn Shafer
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- GSK, San Francisco, CA, 94158, USA
| | - Laralynne Przybyla
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Eric D Chow
- Laboratory for Genomics Research, San Francisco, CA, 94158, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
6
|
Rajamani BM, Illangeswaran RSS, Benjamin ESB, Balakrishnan B, Jebanesan DZP, Das S, Pai AA, Vidhyadharan RT, Mohan A, Karathedath S, Abraham A, Mathews V, Velayudhan SR, Balasubramanian P. Modulating retinoid-X-receptor alpha (RXRA) expression sensitizes chronic myeloid leukemia cells to imatinib in vitro and reduces disease burden in vivo. Front Pharmacol 2023; 14:1187066. [PMID: 37324449 PMCID: PMC10264673 DOI: 10.3389/fphar.2023.1187066] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: The ligand-activated transcription factors, nuclear hormone receptors (NHRs), remain unexplored in hematological malignancies except for retinoic acid receptor alpha (RARA). Methods: Here we profiled the expression of various NHRs and their coregulators in Chronic myeloid leukemia (CML) cell lines and identified a significant differential expression pattern between inherently imatinib mesylate (IM)-sensitive and resistant cell lines. Results: Retinoid-X-receptor alpha (RXRA) was downregulated in CML cell lines inherently resistant to IM and in primary CML CD34+ cells. Pre-treatment with clinically relevant RXRA ligands improved sensitivity to IM in-vitro in both CML cell lines and primary CML cells. This combination effectively reduced the viability and colony-forming capacity of CML CD34+ cells in-vitro. In-vivo, this combination reduced leukemic burden and prolonged survival. Overexpression (OE) of RXRA inhibited proliferation and improved sensitivity to IM in-vitro. In-vivo, RXRA OE cells showed reduced engraftment of cells in the bone marrow, improved sensitivity to IM, and prolonged survival. Both RXRA OE and ligand treatment markedly reduced BCR::ABL1 downstream kinase activation, activating apoptotic cascades and improving sensitivity to IM. Importantly, RXRA OE also led to the disruption of the oxidative capacity of these cells. Conclusion: Combining IM with clinically available RXRA ligands could form an alternative treatment strategy in CML patients with suboptimal response to IM.
Collapse
Affiliation(s)
- Bharathi M. Rajamani
- Department of Haematology, Christian Medical College, Vellore, India
- Department of Biotechnology, Thiruvalluvar University, Vellore, India
| | | | - Esther Sathya Bama Benjamin
- Department of Haematology, Christian Medical College, Vellore, India
- Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Balaji Balakrishnan
- Department of Haematology, Christian Medical College, Vellore, India
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | | | - Saswati Das
- Department of Haematology, Christian Medical College, Vellore, India
- Department of Biotechnology, Thiruvalluvar University, Vellore, India
| | - Aswin Anand Pai
- Department of Haematology, Christian Medical College, Vellore, India
- Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | | | - Ajith Mohan
- Department of Haematology, Christian Medical College, Vellore, India
| | | | - Aby Abraham
- Department of Haematology, Christian Medical College, Vellore, India
| | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | - Shaji R. Velayudhan
- Department of Haematology, Christian Medical College, Vellore, India
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, India
| | | |
Collapse
|
7
|
Yin L, Zhang Q, Xie S, Cheng Z, Li R, Zhu H, Yu Q, Yuan H, Wang C, Peng H, Zhang G. HDAC inhibitor chidamide overcomes drug resistance in chronic myeloid leukemia with the T315i mutation through the Akt-autophagy pathway. Hum Cell 2023:10.1007/s13577-023-00919-1. [PMID: 37222919 DOI: 10.1007/s13577-023-00919-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/16/2023] [Indexed: 05/25/2023]
Abstract
Currently, therapy for Chronic Myeloid Leukemia (CML) patients with the T315I mutation is a major challenge in clinical practice due to its high degree of resistance to first- and second-generation Tyrosine Kinase Inhibitors (TKIs). Chidamide, a Histone Deacetylase Inhibitor (HDACi) drug, is currently used to treat peripheral T-cell lymphoma. In this study, we investigated the anti-leukemia effects of chidamide on the CML cell lines Ba/F3 P210 and Ba/F3 T315I and primary tumor cells from CML patients with the T315I mutation. The underlying mechanism was investigated, and we found that chidamide could inhibit Ba/F3 T315I cells at G0/G1 phase. Signaling pathway analysis showed that chidamide induced H3 acetylation, downregulated pAKT expression and upregulated pSTAT5 expression in Ba/F3 T315I cells. Additionally, we found that the antitumor effect of chidamide could be exerted by regulating the crosstalk between apoptosis and autophagy. When chidamide was used in combination with imatinib or nilotinib, the antitumor effects were enhanced compared with chidamide alone in Ba/F3 T315I and Ba/F3 P210 cells. Therefore, we conclude that chidamide may overcome T315I mutation-related drug resistance in CML patients and works efficiently if used in combination with TKIs.
Collapse
Affiliation(s)
- Le Yin
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Qingyang Zhang
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Sisi Xie
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Zhao Cheng
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Ruijuan Li
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Hongkai Zhu
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Qian Yu
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Huan Yuan
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Canfei Wang
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China.
| | - Hongling Peng
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, 410011, Hunan, China.
- Institute of Molecular Hematology, Central South University, Changsha, China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China.
| | - Guangsen Zhang
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| |
Collapse
|
8
|
Li T, Yao L, Hua Y, Wu Q. Comprehensive analysis of prognosis of cuproptosis-related oxidative stress genes in multiple myeloma. Front Genet 2023; 14:1100170. [PMID: 37065484 PMCID: PMC10102368 DOI: 10.3389/fgene.2023.1100170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: Multiple myeloma (MM) is a highly heterogeneous hematologic malignancy. The patients’ survival outcomes vary widely. Establishing a more accurate prognostic model is necessary to improve prognostic precision and guide clinical therapy.Methods: We developed an eight-gene model to assess the prognostic outcome of MM patients. Univariate Cox analysis, Least absolute shrinkage and selection operator (LASSO) regression, and multivariate Cox regression analyses were used to identify the significant genes and construct the model. Other independent databases were used to validate the model.Results: The results showed that the overall survival of patients in the high-risk group was signifificantly shorter compared with that of those in the low-risk group. The eight-gene model demonstrated high accuracy and reliability in predicting the prognosis of MM patients.Discussion: Our study provides a novel prognostic model for MM patients based on cuproptosis and oxidative stress. The eight-gene model can provide valid predictions for prognosis and guide personalized clinical treatment. Further studies are needed to validate the clinical utility of the model and explore potential therapeutic targets.
Collapse
|
9
|
Tsubaki M, Takeda T, Koumoto Y, Usami T, Matsuda T, Seki S, Sakai K, Nishio K, Nishida S. Activation of ERK1/2 by MOS and TPL2 leads to dasatinib resistance in chronic myeloid leukaemia cells. Cell Prolif 2023:e13420. [PMID: 36847709 DOI: 10.1111/cpr.13420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 03/01/2023] Open
Abstract
The development of BCR::ABL1 tyrosine kinase inhibitors (TKIs), such as dasatinib, has dramatically improved survival in cases of chronic myeloid leukaemia (CML). However, the development of resistance to BCR::ABL1 TKIs is a clinical problem. BCR::ABL1 TKI resistance is known to have BCR::ABL1-dependent or BCR::ABL1-independent mechanisms, but the mechanism of BCR::ABL1 independence is not well understood. In the present study, we investigated the mechanism of BCR::ABL1-independent dasatinib resistance. The expression and activation level of genes or proteins were evaluated using array CGH, real time PCR, or western blot analysis. Gene expression was modulated using siRNA-mediated knockdown. Cell survival was assessed by using trypan blue dye method. We found that dasatinib-resistant K562/DR and KU812/DR cells did not harbour a BCR::ABL1 mutation but had elevated expression and/or activation of MOS, TPL2 and ERK1/2. In addition, MOS siRNA, TPL2 siRNA and trametinib resensitized dasatinib-resistant cells to dasatinib. Moreover, expression levels of MOS in dasatinib non-responder patients with CML were higher than those in dasatinib responders, and the expression of TPL2 tended to increase in dasatinib non-responder patients compared with that in responder patients. Our results indicate that activation of ERK1/2 by elevated MOS and TPL2 expression is involved in dasatinib resistance, and inhibition of these proteins overcomes dasatinib resistance. Therefore, MOS, TPL2 and ERK1/2 inhibitors may be therapeutically useful for treating BCR::ABL1-independent dasatinib-resistant CML.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Yuuichi Koumoto
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Takehiro Usami
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| |
Collapse
|
10
|
Prognostic impact of ASXL1 mutations in chronic phase chronic myeloid leukemia. Blood Cancer J 2022; 12:144. [PMID: 36307398 PMCID: PMC9616867 DOI: 10.1038/s41408-022-00742-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/22/2022] Open
Abstract
While the clinical impact of mutations in the ABL1 gene on response to therapy in chronic phase chronic myeloid leukemia (CP-CML) is well established, less is known about how other mutations affect prognosis. In a retrospective analysis, we identified 115 patients with CML (71 chronic, 15 accelerated and 29 blast phase) where targeted next-generation sequencing of genes recurrently mutated in myeloid leukemias was performed. ASXL1 was the most frequently mutated gene in the chronic (14%) and accelerated phase (40%) CML patients, whereas RUNX1 (20%) was the most common mutation in blast phase. Compared with wild-type ASXL1, CP-CML with mutant ASXL1 was associated with worse event-free survival (EFS) (median of 32.8 vs 88.3 months; P = 0.002) and failure-free survival (median of 13.8 vs 57.8 months; P = 0.04). In a multivariate analysis, ASXL1 mutation was the only independent risk factor associated with worse EFS in chronic phase CML with a hazard ratio of 4.25 (95% CI 1.59–11.35, P = 0.004). In conclusion, mutations in ASXL1 are associated with worse outcomes when detected in chronic phase CML.
Collapse
|
11
|
Stukan I, Gryzik M, Hoser G, Want A, Grabowska-Pyrzewicz W, Zdioruk M, Napiórkowska M, Cieślak M, Królewska-Golińska K, Nawrot B, Basak G, Wojda U. Novel Dicarboximide BK124.1 Breaks Multidrug Resistance and Shows Anticancer Efficacy in Chronic Myeloid Leukemia Preclinical Models and Patients' CD34 +/CD38 - Leukemia Stem Cells. Cancers (Basel) 2022; 14:cancers14153641. [PMID: 35892900 PMCID: PMC9332833 DOI: 10.3390/cancers14153641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/20/2022] [Accepted: 07/23/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Chemotherapy is a first line treatment in many cancer types, but the constant exposition to chemotherapeutics often leads to therapy resistance. An example is chronic myeloid leukemia that, due to the use of tyrosine kinase inhibitors such as imatinib, remains manageable, however incurable. Overall, 20–25% of imatinib responders develop secondary resistance, and among them, 20–40% is due to mechanisms such as expression of P-glycoprotein (MDR1) or leukemia stem cells’ mechanisms of survival and cancer regrowth. This study provides the first evidence from animal and cellular models that this resistance can be overcome with the novel dicarboximide BK124.1. The compound causes no visible toxicity in mice, and has proper pharmacokinetics for therapeutic applications. It was efficient against both multidrug resistant CML blasts and CD34+/CD38− leukemia stem cells coming from CML patients. Future development of BK124.1 could offer curative treatment of CML and of other cancers resistant or intolerant to current chemotherapy. Abstract The search is ongoing for new anticancer therapeutics that would overcome resistance to chemotherapy. This includes chronic myeloid leukemia, particularly suitable for the studies of novel anticancer compounds due to its homogenous and well-known genetic background. Here we show anticancer efficacy of novel dicarboximide denoted BK124.1 (C31H37ClN2O4) in a mouse CML xenograft model and in vitro in two types of chemoresistant CML cells: MDR1 blasts and in CD34+ patients’ stem cells (N = 8) using immunoblotting and flow cytometry. Intraperitoneal administration of BK124.1 showed anti-CML efficacy in the xenograft mouse model (N = 6) comparable to the commonly used imatinib and hydroxyurea. In K562 blasts, BK124.1 decreased the protein levels of BCR-ABL1 kinase and its downstream effectors, resulting in G2/M cell cycle arrest and apoptosis associated with FOXO3a/p21waf1/cip1 upregulation in the nucleus. Additionally, BK124.1 evoked massive apoptosis in multidrug resistant K562-MDR1 cells (IC50 = 2.16 μM), in CD34+ cells from CML patients (IC50 = 1.5 µM), and in the CD34+/CD38− subpopulation consisting of rare, drug-resistant cancer initiating stem cells. Given the advantages of BK124.1 as a potential chemotherapeutic and its unique ability to overcome BCR-ABL1 dependent and independent multidrug resistance mechanisms, future development of BK124.1 could offer a cure for CML and other cancers resistant to present drugs.
Collapse
Affiliation(s)
- Iga Stukan
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Marek Gryzik
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Grażyna Hoser
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
- Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Andrew Want
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Wioleta Grabowska-Pyrzewicz
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Mikolaj Zdioruk
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Mariola Napiórkowska
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Marcin Cieślak
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-363 Lodz, Poland; (M.C.); (K.K.-G.); (B.N.)
| | - Karolina Królewska-Golińska
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-363 Lodz, Poland; (M.C.); (K.K.-G.); (B.N.)
| | - Barbara Nawrot
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-363 Lodz, Poland; (M.C.); (K.K.-G.); (B.N.)
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
- Correspondence: ; Tel.: +48-22-5892-578
| |
Collapse
|
12
|
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
|
13
|
BCR-ABL1 Tyrosine Kinase Complex Signaling Transduction: Challenges to Overcome Resistance in Chronic Myeloid Leukemia. Pharmaceutics 2022; 14:pharmaceutics14010215. [PMID: 35057108 PMCID: PMC8780254 DOI: 10.3390/pharmaceutics14010215] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/27/2023] Open
Abstract
The constitutively active BCR-ABL1 tyrosine kinase, found in t(9;22)(q34;q11) chromosomal translocation-derived leukemia, initiates an extremely complex signaling transduction cascade that induces a strong state of resistance to chemotherapy. Targeted therapies based on tyrosine kinase inhibitors (TKIs), such as imatinib, dasatinib, nilotinib, bosutinib, and ponatinib, have revolutionized the treatment of BCR-ABL1-driven leukemia, particularly chronic myeloid leukemia (CML). However, TKIs do not cure CML patients, as some develop TKI resistance and the majority relapse upon withdrawal from treatment. Importantly, although BCR-ABL1 tyrosine kinase is necessary to initiate and establish the malignant phenotype of Ph-related leukemia, in the later advanced phase of the disease, BCR-ABL1-independent mechanisms are also in place. Here, we present an overview of the signaling pathways initiated by BCR-ABL1 and discuss the major challenges regarding immunologic/pharmacologic combined therapies.
Collapse
|
14
|
De Santis S, Monaldi C, Mancini M, Bruno S, Cavo M, Soverini S. Overcoming Resistance to Kinase Inhibitors: The Paradigm of Chronic Myeloid Leukemia. Onco Targets Ther 2022; 15:103-116. [PMID: 35115784 PMCID: PMC8800859 DOI: 10.2147/ott.s289306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Abstract
Protein kinases (PKs) play crucial roles in cellular proliferation and survival, hence their deregulation is a common event in the pathogenesis of solid and hematologic malignancies. Targeting PKs has been a promising strategy in cancer treatment, and there are now a variety of approved anticancer drugs targeting PKs. However, the phenomenon of resistance remains an obstacle to be addressed and overcoming resistance is a goal to be achieved. Chronic myeloid leukemia (CML) is the first as well as one of the best examples of a cancer that can be targeted by molecular therapy; hence, it can be used as a model disease for other cancers. This review aims to summarize up-to-date knowledge on the main mechanisms implicated in resistance to PK inhibitory therapies and to outline the main strategies that are being explored to overcome resistance. The importance of molecular diagnostics and disease monitoring in counteracting resistance will also be discussed.
Collapse
Affiliation(s)
- Sara De Santis
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
- Correspondence: Sara De Santis Insitute of Hematology “Lorenzo e Ariosto Seràgnoli”, Via Massarenti 9, Bologna, 40138, ItalyTel +39 051 2143791Fax +39 051 2144037 Email
| | - Cecilia Monaldi
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
| | - Manuela Mancini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Samantha Bruno
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
| | - Michele Cavo
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Simona Soverini
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
| |
Collapse
|
15
|
Ismail MA, Nasrallah GK, Monne M, AlSayab A, Yassin MA, Varadharaj G, Younes S, Sorio C, Cook R, Modjtahedi H, Al-Dewik NI. Description of PTPRG genetic variants identified in a cohort of Chronic Myeloid Leukemia patients and their ability to influence response to Tyrosine kinase Inhibitors. Gene 2021; 813:146101. [PMID: 34906644 DOI: 10.1016/j.gene.2021.146101] [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: 07/09/2021] [Revised: 10/07/2021] [Accepted: 11/16/2021] [Indexed: 12/25/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) have remarkably transformed Ph+ chronic myeloid leukemia (CML) management; however, TKI resistance remains a major clinical challenge. Mutations in BCR-ABL1 are well studied but fail to explain 20-40% of resistant cases, suggesting the activation of alternative, BCR-ABL1-independent pathways. Protein Tyrosine Phosphatase Receptor Gamma (PTPRG), a tumor suppressor, was found to be well expressed in CML patients responsive to TKIs and down-regulated in resistant patients. In this study, we aimed to identify genetic variants in PTPRG that could potentially modulate TKIs response in CML patients. DNA was extracted from peripheral blood samples collected from two CML cohorts (Qatar and Italy) and targeted exome sequencing was performed. Among 31 CML patients, six were TKI-responders and 25 were TKI-resistant. Sequencing identified ten variants, seven were annotated and three were novel SNPs (c.1602_1603insC, c.85+86delC, and c.2289-129delA). Among them, five variants were identified in 15 resistant cases. Of these, one novel exon variant (c.1602_1603insC), c.841-29C>T (rs199917960) and c.1378-224A>G (rs2063204) were found to be significantly different between the resistant cases compared to responders. Our findings suggest that PTPRG variants may act as an indirect resistance mechanism of BCR-ABL1 to affect TKI treatment.
Collapse
Affiliation(s)
- Mohamed A Ismail
- School of Life Science, Pharmacy and Chemistry, Faculty of science, engineering & computing-Kingston University London, United Kingdom; Interim Translational Research Institute (iTRI), Hamad Medical Corporation (HMC), Doha, Qatar
| | - Gheyath K Nasrallah
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Maria Monne
- Centro di Diagnostica Biomolecolare e Citogenetica Emato-Oncologica, "San Francesco" Hospital, Nuoro, Italy
| | - Ali AlSayab
- Interim Translational Research Institute (iTRI), Hamad Medical Corporation (HMC), Doha, Qatar
| | - Mohamed A Yassin
- Department of Medical Oncology, National Centre for Cancer Care and Research, Hamad Medical Corporation (HMC), Doha, Qatar
| | | | - Salma Younes
- Department of Research, Women's Wellness and Research Center, Hamad Medical Corporation, Qatar
| | - Claudio Sorio
- Department of Medicine, University of Verona, Verona, Italy
| | - Richard Cook
- School of Life Science, Pharmacy and Chemistry, Faculty of science, engineering & computing-Kingston University London, United Kingdom
| | - Helmout Modjtahedi
- School of Life Science, Pharmacy and Chemistry, Faculty of science, engineering & computing-Kingston University London, United Kingdom
| | - Nader I Al-Dewik
- Interim Translational Research Institute (iTRI), Hamad Medical Corporation (HMC), Doha, Qatar; Faculty of Health and Social Care Sciences, Kingston University, St. George's University of London, UK; Clinical and Metabolic Genetics, Department of Pediatrics, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Science (CHLS), Hamad Bin Khalifa University (HBKU), Doha, Qatar.
| |
Collapse
|
16
|
Chang LL, Xu XQ, Liu XL, Guo QQ, Fan YN, He BX, Zhang WZ. Emerging role of m6A methylation modification in ovarian cancer. Cancer Cell Int 2021; 21:663. [PMID: 34895230 PMCID: PMC8666073 DOI: 10.1186/s12935-021-02371-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
m6A (N6-methyladenosine) methylation, a well-known modification in tumour epigenetics, dynamically and reversibly fine tunes the entire process of RNA metabolism. Aberrant levels of m6A and its regulators, which can predict the survival and outcomes of cancer patients, are involved in tumorigenesis, metastasis and resistance. Ovarian cancer (OC) ranks first among gynaecological tumours in the causes of death. At first diagnosis, patients with OC are usually at advanced stages owing to a lack of early biomarkers and effective targets. After treatment, patients with OC often develop drug resistance. This article reviews the recent experimental advances in understanding the role of m6A modification in OC, raising the possibility to treat m6A modification and its regulators as promising diagnostic markers and therapeutic targets for OC. ![]()
Collapse
Affiliation(s)
- Lin-Lin Chang
- Department of Pharmacy, Affiliated Tumour Hospital of Zhengzhou University, Henan Cancer Hospital, 127# Dongming Rd, Zhengzhou, 450008, Henan, China.
| | - Xia-Qing Xu
- Department of Clinical Pharmacy, Zhengzhou Central Hospital Affiliated To Zhengzhou University, Zhengzhou, China
| | - Xue-Ling Liu
- Department of Pharmacy, Affiliated Tumour Hospital of Zhengzhou University, Henan Cancer Hospital, 127# Dongming Rd, Zhengzhou, 450008, Henan, China
| | - Qian-Qian Guo
- Department of Pharmacy, Affiliated Tumour Hospital of Zhengzhou University, Henan Cancer Hospital, 127# Dongming Rd, Zhengzhou, 450008, Henan, China
| | - Yan-Nan Fan
- Department of Pharmacy, Affiliated Tumour Hospital of Zhengzhou University, Henan Cancer Hospital, 127# Dongming Rd, Zhengzhou, 450008, Henan, China
| | - Bao-Xia He
- Department of Pharmacy, Affiliated Tumour Hospital of Zhengzhou University, Henan Cancer Hospital, 127# Dongming Rd, Zhengzhou, 450008, Henan, China
| | - Wen-Zhou Zhang
- Department of Pharmacy, Affiliated Tumour Hospital of Zhengzhou University, Henan Cancer Hospital, 127# Dongming Rd, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
17
|
Leukemia Stem Cells as a Potential Target to Achieve Therapy-Free Remission in Chronic Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13225822. [PMID: 34830976 PMCID: PMC8616035 DOI: 10.3390/cancers13225822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Leukemia stem cells (LSCs, also known as leukemia-initiating cells) not only drive leukemia initiation and progression, but also contribute to drug resistance and/or disease relapse. Therefore, eradication of every last LSC is critical for a patient's long-term cure. Chronic myeloid leukemia (CML) is a myeloproliferative disorder that arises from multipotent hematopoietic stem and progenitor cells. Tyrosine kinase inhibitors (TKIs) have dramatically improved long-term outcomes and quality of life for patients with CML in the chronic phase. Point mutations of the kinase domain of BCR-ABL1 lead to TKI resistance through a reduction in drug binding, and as a result, several new generations of TKIs have been introduced to the clinic. Some patients develop TKI resistance without known mutations, however, and the presence of LSCs is believed to be at least partially associated with resistance development and CML relapse. We previously proposed targeting quiescent LSCs as a therapeutic approach to CML, and a number of potential strategies for targeting insensitive LSCs have been presented over the last decade. The identification of specific markers distinguishing CML-LSCs from healthy HSCs, and the potential contributions of the bone marrow microenvironment to CML pathogenesis, have also been explored. Nonetheless, 25% of CML patients are still expected to switch TKIs at least once, and various TKI discontinuation studies have shown a wide range in the incidence of molecular relapse (from 30% to 60%). In this review, we revisit the current knowledge regarding the role(s) of LSCs in CML leukemogenesis and response to pharmacological treatment and explore how durable treatment-free remission may be achieved and maintained after discontinuing TKI treatment.
Collapse
|
18
|
Xu X, Yin S, Ren Y, Hu C, Zhang A, Lin Y. Proteomics analysis reveals the correlation of programmed ROS-autophagy loop and dysregulated G1/S checkpoint with imatinib resistance in chronic myeloid leukemia cells. Proteomics 2021; 22:e2100094. [PMID: 34564948 DOI: 10.1002/pmic.202100094] [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] [Received: 04/07/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 11/07/2022]
Abstract
Although tyrosine kinase inhibitors (TKIs), including imatinib, have greatly improved clinical treatment of patients with chronic myeloid leukemia (CML), drug resistance remains a major obstacle. Studies on the mechanisms underlying imatinib resistance and other alternative drugs are urgently needed. Liquid chromatography tandem mass spectrometry was applied to investigate the differences in proteomics and phosphoproteomics between K562 and K562/G (imatinib resistant K562). Multiple bioinformatics analyses were performed to unveil the differential signal pathways. CCK-8 was used to detect cell proliferation. Flow cytometry was performed to analyze reactive oxygen species (ROS), cell cycle, and cell apoptosis. Western blotting and quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) were used to observe the changes of ROS and autophagy associated with imatinib resistance in CML. Our results indicated that ROS-autophagy formed one negative feedback loop and was associated with imatinib resistance. Additionally, the limited-rate enzymes of serine synthesis pathway were escalated in K562/G, which could contribute to the increased cyclin-dependent kinases and cell proliferation index. According to phosphoproteomics data, K562/G cells exhibited abnormal phosphorylation of splicing signals. These results revealed that it could be one useful strategy to correct metabolism shift and oxidative stress, or moderately regulate autophagy. Future research should focus on the discovery of potential targets in ROS-autophagy loop.
Collapse
Affiliation(s)
- Xiucai Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Shihong Yin
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yingli Ren
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chaojie Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Aimei Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Ya Lin
- Wannan Medical College, Wuhu, Anhui, People's Republic of China
| |
Collapse
|
19
|
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
|
20
|
Wardhani SO, Susanti H, Rahayu P, Yueniwati Y, Fajar J. The Levels of FoxO3a Predict the Failure of Imatinib Mesylate Therapy among Chronic Myeloid Leukemia Patients. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION: Forkhead Transcription Factor 3a (FoxO3a) has been proposed to have a high efficacy to predict the failure of imatinib mesylate (IM) therapy among Chronic Myeloid Leukemia (CML) patients. However, the limited evidence had made this marker remained controversy.
OBJECTIVES: We aimed to investigate the correlation between the levels of FoxO3a and the risk of treatment failure of IM therapy in CML patients.
METHODS: A prospective cohort study was carried out between February 2019 and February 2020 in Saiful Anwar Hospital, Malang, Indonesia. All CML patients treated with IM on our hospital during the study period were included. The levels of FoxO3a was determined using the Enzyme-linked immunosorbent assay (ELISA) using Cusabio Biotech Kit (Cusabio Biotech Co., New York, USA). The treatment response was assessed using the European Leukemia criteria. The correlation and effect estimate between the levels of FoxO3a and treatment response of CML patients was assessed using multiple logistic regression.
RESULTS: 53 CML patients receiving IM in our hospital were included, consisting of 29 patients with good response and 24 patients with non-response. Our study found that CML patients with lower levels of FoxO3a was associated with increased risk to develop treatment failure when treated with IM. Moreover, we also found that higher risk of treatment failure of IM therapy was also found in patients with increased levels of thrombocytes, basophils, and leukocytes, and lower levels of hemoglobin.
CONCLUSION: We reveal that FoxO3a is the prominent marker to predict the treatment response of CML patients treated with IM.
Collapse
|
21
|
Zizioli D, Bernardi S, Varinelli M, Farina M, Mignani L, Bosio K, Finazzi D, Monti E, Polverelli N, Malagola M, Borsani E, Borsani G, Russo D. Development of BCR-ABL1 Transgenic Zebrafish Model Reproducing Chronic Myeloid Leukemia (CML) Like-Disease and Providing a New Insight into CML Mechanisms. Cells 2021; 10:cells10020445. [PMID: 33669758 PMCID: PMC7922348 DOI: 10.3390/cells10020445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 12/27/2022] Open
Abstract
Zebrafish has proven to be a versatile and reliable experimental in vivo tool to study human hematopoiesis and model hematological malignancies. Transgenic technologies enable the generation of specific leukemia types by the expression of human oncogenes under specific promoters. Using this technology, a variety of myeloid and lymphoid malignancies zebrafish models have been described. Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasia characterized by the BCR-ABL1 fusion gene, derived from the t (9;22) translocation causing the Philadelphia Chromosome (Ph). The BCR-ABL1 protein is a constitutively activated tyrosine kinas inducing the leukemogenesis and resulting in an accumulation of immature leukemic cells into bone marrow and peripheral blood. To model Ph+ CML, a transgenic zebrafish line expressing the human BCR-ABL1 was generated by the Gal4/UAS system, and then crossed with the hsp70-Gal4 transgenic line. The new line named (BCR-ABL1pUAS:CFP/hsp70-Gal4), presented altered expression of hematopoietic markers during embryonic development compared to controls and transgenic larvae showed proliferating hematopoietic cells in the caudal hematopoietic tissue (CHT). The present transgenic zebrafish would be a robust CML model and a high-throughput drug screening tool.
Collapse
Affiliation(s)
- Daniela Zizioli
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.V.); (L.M.); (D.F.); (E.M.)
- Correspondence: daniela.zizioli@unibs; Tel.: +39-(03)-03717546
| | - Simona Bernardi
- Unit of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy; (S.B.); (M.F.); (K.B.); (N.P.); (M.M.); (D.R.)
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| | - Marco Varinelli
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.V.); (L.M.); (D.F.); (E.M.)
| | - Mirko Farina
- Unit of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy; (S.B.); (M.F.); (K.B.); (N.P.); (M.M.); (D.R.)
| | - Luca Mignani
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.V.); (L.M.); (D.F.); (E.M.)
| | - Katia Bosio
- Unit of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy; (S.B.); (M.F.); (K.B.); (N.P.); (M.M.); (D.R.)
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| | - Dario Finazzi
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.V.); (L.M.); (D.F.); (E.M.)
- Laboratorio Centrale Analisi Chimico-Cliniche, ASST Spedali Civili, 25123 Brescia, Italy
| | - Eugenio Monti
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.V.); (L.M.); (D.F.); (E.M.)
| | - Nicola Polverelli
- Unit of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy; (S.B.); (M.F.); (K.B.); (N.P.); (M.M.); (D.R.)
| | - Michele Malagola
- Unit of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy; (S.B.); (M.F.); (K.B.); (N.P.); (M.M.); (D.R.)
| | - Elisa Borsani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy;
| | - Giuseppe Borsani
- Unit of Biology and Genetic, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Domenico Russo
- Unit of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy; (S.B.); (M.F.); (K.B.); (N.P.); (M.M.); (D.R.)
| |
Collapse
|
22
|
Lei H, Xu HZ, Shan HZ, Liu M, Lu Y, Fang ZX, Jin J, Jing B, Xiao XH, Gao SM, Gao FH, Xia L, Yang L, Liu LG, Wang WW, Liu CX, Tong Y, Wu YZ, Zheng JK, Chen GQ, Zhou L, Wu YL. Targeting USP47 overcomes tyrosine kinase inhibitor resistance and eradicates leukemia stem/progenitor cells in chronic myelogenous leukemia. Nat Commun 2021; 12:51. [PMID: 33397955 PMCID: PMC7782553 DOI: 10.1038/s41467-020-20259-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Identifying novel drug targets to overcome resistance to tyrosine kinase inhibitors (TKIs) and eradicating leukemia stem/progenitor cells are required for the treatment of chronic myelogenous leukemia (CML). Here, we show that ubiquitin-specific peptidase 47 (USP47) is a potential target to overcome TKI resistance. Functional analysis shows that USP47 knockdown represses proliferation of CML cells sensitive or resistant to imatinib in vitro and in vivo. The knockout of Usp47 significantly inhibits BCR-ABL and BCR-ABLT315I-induced CML in mice with the reduction of Lin-Sca1+c-Kit+ CML stem/progenitor cells. Mechanistic studies show that stabilizing Y-box binding protein 1 contributes to USP47-mediated DNA damage repair in CML cells. Inhibiting USP47 by P22077 exerts cytotoxicity to CML cells with or without TKI resistance in vitro and in vivo. Moreover, P22077 eliminates leukemia stem/progenitor cells in CML mice. Together, targeting USP47 is a promising strategy to overcome TKI resistance and eradicate leukemia stem/progenitor cells in CML.
Collapse
MESH Headings
- Animals
- Cell Proliferation/drug effects
- DNA Damage
- DNA Repair/drug effects
- Drug Resistance, Neoplasm/drug effects
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Fusion Proteins, bcr-abl
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice, Knockout
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Proteasome Endopeptidase Complex/metabolism
- Protein Binding/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein Stability/drug effects
- Proteolysis/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- STAT5 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Thiophenes/pharmacology
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin-Specific Proteases/metabolism
- Xenograft Model Antitumor Assays
- Y-Box-Binding Protein 1/metabolism
- ras Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Han-Zhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Hui-Zhuang Shan
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Meng Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Ying Lu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Zhi-Xiao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jin Jin
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Bo Jing
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Xin-Hua Xiao
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Shen-Meng Gao
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Feng-Hou Gao
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, 200011, Shanghai, China
| | - Li Xia
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Li Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Li-Gen Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wei-Wei Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Chuan-Xu Liu
- Department of Hematology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Yin Tong
- Department of Hematology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 200081, Shanghai, China
| | - Yun-Zhao Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jun-Ke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education and Chinese Academy of Medical Sciences Research Unit (NO.2019RU043), Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Li Zhou
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Ying-Li Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
23
|
Involvement of Oxidative Stress in Resistance to Tyrosine-Kinase Inhibitors Therapy in Chronic Myeloid Leukemia. CURRENT HEALTH SCIENCES JOURNAL 2020; 46:420-432. [PMID: 33717518 PMCID: PMC7948018 DOI: 10.12865/chsj.46.04.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/12/2020] [Indexed: 11/29/2022]
Abstract
Oxidative stress involves disruption of the cellular redox status through excessive production of reactive oxygen species or through deficiency in the cellular antioxidant capacity. It is involved in the pathogeny of multiple entities (hematological diseases, metabolic disorders, cardiovascular and renal pathology etc.), as well as in the pharmacokinetics of specific treatments for these pathologies. Chronic myeloid leukemia is a chronic myeloproliferative disease for which current standard treatment is BCR-ABL tyrosine kinase inhibitors. The innovation of this therapy has significantly improved life expectancy for patients with chronic myeloid leukemia, but in some cases, this treatment becomes ineffective, installing the resistance to tyrosine kinase inhibitors therapy. There were described two types of tyrosin kinase inhibitors resistance: primary and secondary resistance. In the present paper we proposed to evaluate the involvement of oxidative in the resistance to tyrosine kinase inhibitors therapy, in the clonal instability in chronic myeloid leukemia and in the progression of the disease to an advanced stage. We concluded that oxidative stress can play a dual role in the evolution of chronic myeloid leukemia: on the one hand it can promote genomic instability and accelerate the progression of the disease to advanced stages associated with tyrosin kinase inhibitors resistance and, on the other hand, it can contribute to leukemic cell apoptosis. It seems to be outlined a fragile balance between the pro- and anti-apoptotic effects of the reactive oxygen species, closely related to their level in the leukemic cells.
Collapse
|
24
|
Novel strategies to eradicate resistant cells in chronic myeloid leukemia. Future Med Chem 2020; 12:2089-2092. [PMID: 33228384 DOI: 10.4155/fmc-2020-0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
25
|
Komorowski L, Fidyt K, Patkowska E, Firczuk M. Philadelphia Chromosome-Positive Leukemia in the Lymphoid Lineage-Similarities and Differences with the Myeloid Lineage and Specific Vulnerabilities. Int J Mol Sci 2020; 21:E5776. [PMID: 32806528 PMCID: PMC7460962 DOI: 10.3390/ijms21165776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022] Open
Abstract
Philadelphia chromosome (Ph) results from a translocation between the breakpoint cluster region (BCR) gene on chromosome 9 and ABL proto-oncogene 1 (ABL1) gene on chromosome 22. The fusion gene, BCR-ABL1, is a constitutively active tyrosine kinase which promotes development of leukemia. Depending on the breakpoint site within the BCR gene, different isoforms of BCR-ABL1 exist, with p210 and p190 being the most prevalent. P210 isoform is the hallmark of chronic myeloid leukemia (CML), while p190 isoform is expressed in majority of Ph-positive B cell acute lymphoblastic leukemia (Ph+ B-ALL) cases. The crucial component of treatment protocols of CML and Ph+ B-ALL patients are tyrosine kinase inhibitors (TKIs), drugs which target both BCR-ABL1 isoforms. While TKIs therapy is successful in great majority of CML patients, Ph+ B-ALL often relapses as a drug-resistant disease. Recently, the high-throughput genomic and proteomic analyses revealed significant differences between CML and Ph+ B-ALL. In this review we summarize recent discoveries related to differential signaling pathways mediated by different BCR-ABL1 isoforms, lineage-specific genetic lesions, and metabolic reprogramming. In particular, we emphasize the features distinguishing Ph+ B-ALL from CML and focus on potential therapeutic approaches exploiting those characteristics, which could improve the treatment of Ph+ B-ALL.
Collapse
Affiliation(s)
- Lukasz Komorowski
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 St, 02-097 Warsaw, Poland; (L.K.); (K.F.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Trojdena 2a St, 02-091 Warsaw, Poland
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 St, 02-097 Warsaw, Poland; (L.K.); (K.F.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Trojdena 2a St, 02-091 Warsaw, Poland
| | - Elżbieta Patkowska
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indiry Gandhi 14, 02-776 Warsaw, Poland;
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 St, 02-097 Warsaw, Poland; (L.K.); (K.F.)
| |
Collapse
|
26
|
Gazeau N, Derrieux C, Nibourel O, Berthon C, Grardel N, Goursaud L, Boyer T, Dumezy F, Coiteux V, Quesnel B, Preudhomme C, Roche-Lestienne C, Duployez N. Disease escape with the selective loss of the Philadelphia chromosome after tyrosine kinase inhibitor exposure in Ph-positive acute lymphoblastic leukemia. Leukemia 2020; 34:2230-2233. [PMID: 31988437 DOI: 10.1038/s41375-020-0715-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Nicolas Gazeau
- CHU Lille, Hematology Department, Claude Huriez Hospital, F-59000, Lille, France
| | | | - Olivier Nibourel
- CHU Lille, Laboratory of Hematology, F-59000, Lille, France
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France
| | - Céline Berthon
- CHU Lille, Hematology Department, Claude Huriez Hospital, F-59000, Lille, France
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France
| | | | - Laure Goursaud
- CHU Lille, Hematology Department, Claude Huriez Hospital, F-59000, Lille, France
| | - Thomas Boyer
- CHU Lille, Laboratory of Hematology, F-59000, Lille, France
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France
| | - Florent Dumezy
- CHU Lille, Laboratory of Hematology, F-59000, Lille, France
| | - Valérie Coiteux
- CHU Lille, Hematology Department, Claude Huriez Hospital, F-59000, Lille, France
| | - Bruno Quesnel
- CHU Lille, Hematology Department, Claude Huriez Hospital, F-59000, Lille, France
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France
| | - Claude Preudhomme
- CHU Lille, Laboratory of Hematology, F-59000, Lille, France
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France
| | - Catherine Roche-Lestienne
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France
- CHU Lille, Institute of Medical Genetics, Jeanne de Flandre Hospital, F-59000, Lille, France
| | - Nicolas Duployez
- CHU Lille, Laboratory of Hematology, F-59000, Lille, France.
- University of Lille, INSERM, UMR-S 1172, F-59000, Lille, France.
| |
Collapse
|
27
|
Teng YQ, Jin H, Liu ZY, Li DD, Ye XM, Yang LY, Zhou J. The Lyn-SIRT1 signaling pathway is involved in imatinib resistance in chronic myeloid leukaemia. Am J Transl Res 2020; 12:2711-2725. [PMID: 32655803 PMCID: PMC7344074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Imatinib resistance is commonly associated with the activation of BCR-ABL signaling in chronic myeloid leukaemia (CML). The activation of Lyn can result in imatinib resistance by regulating the formation of BCR-ABL protein complexes. SIRT1 is a novel survival pathway activated by BCR-ABL expression in haematopoietic progenitor cells. This study aimed to investigate whether the signaling pathway of Lyn/BCR-ABL/SIRT1 could mediate imatinib resistance in CML. METHODS The MTT assay was used to detect cell viability. Apoptosis was measured by a flow cytometry assay. Protein expression was detected by Western blotting. Knockdown CML cells were constructed by shRNA interference. The CML mouse model was used to investigate the role of SIRT1 in CML in vivo. RESULTS Lyn was overexpressed in K562R cells. BCR-ABL phosphorylation and activation were promoted by Lyn. Imatinib suppressed BCR-ABL phosphorylation in both K562 and K562R cells. BCR-ABL positively regulated SIRT1 and Foxo1 but negatively regulated acetylated Foxo1 (Ac-Foxo1) and p53 expression. Pharmacological inhibition of SIRT1 or knockdown of SIRT1 increased apoptosis and reduced growth in vitro and in vivo. Foxo1 was downregulated by SIRT1 inhibition or knockdown, while Ac-Foxo1 and p53 were upregulated. In vivo experiments showed that imatinib and/or SIRT1 inhibition both prolonged the survival of the CML mouse model and that the effects of imatinib were enhanced in combination with SIRT1 inhibition. CONCLUSION We proposed a novel molecular mechanism of imatinib resistance in CML in which the high expression of Lyn in imatinib-resistant cells inhibited Ac-Foxo1 and p53 expression through the BCR-ABL/SIRT1/Foxo1 signaling pathway, thus reducing apoptosis and mediating imatinib resistance.
Collapse
Affiliation(s)
- Yue-Qiu Teng
- Central Laboratory of Blood Cancer, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang Province, P. R. China
| | - Hua Jin
- Department of Neurosurgery, The Third Affiliated Hospital of Harbin Medical UniversityHarbin 150040, Heilongjiang Province, P. R. China
| | - Zhi-Yu Liu
- Central Laboratory of Blood Cancer, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang Province, P. R. China
| | - Dan-Dan Li
- Department of Hematology, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang Province, P. R. China
| | - Xiang-Mei Ye
- Central Laboratory of Blood Cancer, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang Province, P. R. China
| | - Lu-Yuan Yang
- Central Laboratory of Blood Cancer, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang Province, P. R. China
| | - Jin Zhou
- Department of Hematology, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang Province, P. R. China
| |
Collapse
|
28
|
CML - Not only BCR-ABL1 matters. Best Pract Res Clin Haematol 2020; 33:101194. [PMID: 33038988 DOI: 10.1016/j.beha.2020.101194] [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] [Received: 03/07/2020] [Revised: 04/30/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
BCR-ABL1 is in the center of chronic myeloid leukemia (CML) pathology, diagnosis and treatment, as confirmed by the success of tyrosine kinase inhibitor (TKI) therapy. However, additional mechanisms and events, many of which function independently of BCR-ABL1, play important roles, particularly in terms of leukemic stem cell (LSC) persistence, primary and secondary resistance, and disease progression. Promising therapeutic approaches aim to disrupt pathways which mediate LSC survival during successful TKI treatment, in the hope of improving long-term treatment-free-remission and perhaps provide a functional cure for some patients. Over the years through advances in sequencing technology frequent molecular aberrations in addition to BCR-ABL1 have been identified not only in advanced disease but also in chronic phase CML, often affecting epigenetic regulators such as ASXL1, DNMT3A and TET2. Analyses of serial samples have revealed various patterns of clonal evolution with some mutations preceding the BCR-ABL1 acquisition. Such mutations can be considered to be important co-factors in the pathogenesis of CML and could potentially influence therapeutic strategies in the future.
Collapse
|
29
|
Willig JB, Vianna DRB, Beckenkamp A, Beckenkamp LR, Sévigny J, Wink MR, Buffon A, Pilger DA. Imatinib mesylate affects extracellular ATP catabolism and expression of NTPDases in a chronic myeloid leukemia cell line. Purinergic Signal 2020; 16:29-40. [PMID: 31955347 PMCID: PMC7166234 DOI: 10.1007/s11302-019-09686-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 12/29/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm, characterized by the occurrence of the t(9;22)(q34;q11) translocation. First-line therapy for CML consists of treatment with imatinib mesylate, which selectively inhibits the BCR-ABL protein by competing for its ATP-binding site. Adenine nucleotide signaling is modulated by the ectonucleotidases and this pathway is related to tumorigenic processes. Considering the relationship between ATP and cancer, we aimed to evaluate the influence of imatinib mesylate on the expressions and functions of the NTPDase and ecto-5'-nucleotidase (CD73) enzymes in imatinib-sensitive and -resistant K-562 cell lines. mRNA analysis showed that K-562 cells express all ENTPDs and NT5E. However, when treated with imatinib mesylate for 24 h, the expression of ENTPD1, -2, -3 and -5 increased, leading to a higher nucleotides hydrolysis rate. HPLC analysis identified increased ATP degradation in cells after 24 h of treatment, with consequent ADP and AMP formation, corroborating the increase in gene and protein expression of ectonucleotidases as observed in previous results. On the other hand, we observed that imatinib-resistant K-562 cells presented a decrease in nucleotide hydrolysis and expressions of ENTPD1 and -5. These results suggest an involvement of imatinib in modulating ectonucleotidases in CML that will need further investigation. Since these ectonucleotidases have important catalytic activities in the tumor microenvironment, their modulation in CML cells may represent an important therapeutic approach to regulate levels of extracellular adenine nucleotides.
Collapse
Affiliation(s)
- Julia Biz Willig
- Faculty of Farmacy, Program for Post-graduation in Pharmaceutical Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratory of Biochemical and Cytological Analyses, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, Bairro Santana, Porto Alegre, Rio Grande do Sul, CEP 90610-000, Brazil
| | - Débora Renz Barreto Vianna
- Faculty of Farmacy, Program for Post-graduation in Pharmaceutical Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratory of Biochemical and Cytological Analyses, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, Bairro Santana, Porto Alegre, Rio Grande do Sul, CEP 90610-000, Brazil
| | - Aline Beckenkamp
- Faculty of Farmacy, Program for Post-graduation in Pharmaceutical Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Liziane Raquel Beckenkamp
- Laboratory of Cell Biology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jean Sévigny
- Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, Canada
- Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
| | - Márcia Rosângela Wink
- Laboratory of Cell Biology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Andréia Buffon
- Faculty of Farmacy, Program for Post-graduation in Pharmaceutical Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diogo André Pilger
- Faculty of Farmacy, Program for Post-graduation in Pharmaceutical Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
- Laboratory of Biochemical and Cytological Analyses, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, Bairro Santana, Porto Alegre, Rio Grande do Sul, CEP 90610-000, Brazil.
| |
Collapse
|
30
|
Özgür Yurttaş N, Eşkazan AE. Novel therapeutic approaches in chronic myeloid leukemia. Leuk Res 2020; 91:106337. [PMID: 32200189 DOI: 10.1016/j.leukres.2020.106337] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
The tyrosine kinase inhibitors (TKIs) have revolutionized the management of chronic myeloid leukemia (CML) and BCR-ABL1 inhibitors form the mainstay of CML treatment. Although patients with CML generally do well under TKI therapy, there is a subgroup of patients who are resistant and/or intolerant to TKIs. In these group of patients, there is the need of additional treatment strategies. In this review, we provide an update on the current knowledge of these novel treatment approaches that can be used alone and/or in combination with TKIs.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Clinical Trials as Topic
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Everolimus/therapeutic use
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/immunology
- Gene Expression
- Histone Deacetylase Inhibitors/therapeutic use
- Homoharringtonine/therapeutic use
- Humans
- Immunotherapy/methods
- Interferon-alpha/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy/methods
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Piperidines/therapeutic use
- Polyethylene Glycols/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/therapeutic use
- Pyridines/therapeutic use
- Quinolones/therapeutic use
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Nurgül Özgür Yurttaş
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ahmet Emre Eşkazan
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
31
|
SRSF1 mediates cytokine-induced impaired imatinib sensitivity in chronic myeloid leukemia. Leukemia 2020; 34:1787-1798. [PMID: 32051529 DOI: 10.1038/s41375-020-0732-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 12/10/2019] [Accepted: 01/29/2020] [Indexed: 12/11/2022]
Abstract
Patients with chronic myeloid leukemia (CML) who are treated with tyrosine kinase inhibitors (TKIs) experience significant heterogeneity regarding depth and speed of responses. Factors intrinsic and extrinsic to CML cells contribute to response heterogeneity and TKI resistance. Among extrinsic factors, cytokine-mediated TKI resistance has been demonstrated in CML progenitors, but the underlying mechanisms remain obscure. Using RNA-sequencing, we identified differentially expressed splicing factors in primary CD34+ chronic phase (CP) CML progenitors and controls. We found SRSF1 expression to be increased as a result of both BCR-ABL1- and cytokine-mediated signaling. SRSF1 overexpression conferred cytokine independence to untransformed hematopoietic cells and impaired imatinib sensitivity in CML cells, while SRSF1 depletion in CD34+ CP CML cells prevented the ability of extrinsic cytokines to decrease imatinib sensitivity. Mechanistically, PRKCH and PLCH1 were upregulated by elevated SRSF1 levels, and contributed to impaired imatinib sensitivity. Importantly, very high SRSF1 levels in the bone marrow of CML patients at presentation correlated with poorer clinical TKI responses. In summary, we find SRSF1 levels to be maintained in CD34+ CP CML progenitors by cytokines despite effective BCR-ABL1 inhibition, and that elevated levels promote impaired imatinib responses. Together, our data support an SRSF1/PRKCH/PLCH1 axis in contributing to cytokine-induced impaired imatinib sensitivity in CML.
Collapse
|
32
|
Lu T, Wei D, Yu K, Ma D, Xiong J, Fang Q, Wang J. Betulinic acid restores imatinib sensitivity in BCR-ABL1 kinase-independent, imatinib-resistant chronic myeloid leukemia by increasing HDAC3 ubiquitination and degradation. Ann N Y Acad Sci 2020; 1467:77-93. [PMID: 31930541 DOI: 10.1111/nyas.14298] [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: 09/23/2019] [Revised: 11/28/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
Although imatinib (IM) has been demonstrated to be an efficient treatment in chronic myeloid leukemia (CML), some patients still experience IM resistance and disease relapse. Through in vitro studies, we observed that HDAC3 levels were elevated in BCR-ABL1 kinase-independent, IM-resistant primary cells from CML patients and in IM-resistant K562 (K562R) cells and that downregulation of HDAC3 could enhance IM efficacy in K562R cells. Furthermore, betulinic acid (BA), a lupane-type pentacyclic triterpenoid saponin isolated from birch trees, restored IM sensitivity in the BCR-ABL1 kinase-independent, IM-resistant primary cells and in K562R cells, as well as in primary CD34+ bone marrow cells from CML patients. We found that BA restored IM sensitivity through inhibition of HDAC3 accumulation in cells, and that this was mediated by BA-dependent ubiquitination and degradation of HDAC3. BA at low dosage significantly increased IM antitumor effects on murine xenografts bearing K562R cells and inhibited HDAC3 expression in tumor tissue. Our findings demonstrated that HDAC3 is an essential factor in BCR-ABL1 kinase-independent IM resistance, and that BA in combination with IM may be a novel treatment strategy for overcoming IM resistance in CML.
Collapse
Affiliation(s)
- Tingting Lu
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Department of Clinical Laboratory Centre, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China
| | - Danna Wei
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Kunlin Yu
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China.,Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Dan Ma
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Jie Xiong
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Jishi Wang
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| |
Collapse
|
33
|
Mechanisms of Disease Progression and Resistance to Tyrosine Kinase Inhibitor Therapy in Chronic Myeloid Leukemia: An Update. Int J Mol Sci 2019; 20:ijms20246141. [PMID: 31817512 PMCID: PMC6940932 DOI: 10.3390/ijms20246141] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
Chronic myeloid leukemia (CML) is characterized by the presence of the BCR-ABL1 fusion gene, which encodes a constitutive active tyrosine kinase considered to be the pathogenic driver capable of initiating and maintaining the disease. Despite the remarkable efficacy of tyrosine kinase inhibitors (TKIs) targeting BCR-ABL1, some patients may not respond (primary resistance) or may relapse after an initial response (secondary resistance). In a small proportion of cases, development of resistance is accompanied or shortly followed by progression from chronic to blastic phase (BP), characterized by a dismal prognosis. Evolution from CP into BP is a multifactorial and probably multistep phenomenon. Increase in BCR-ABL1 transcript levels is thought to promote the onset of secondary chromosomal or genetic defects, induce differentiation arrest, perturb RNA transcription, editing and translation that together with epigenetic and metabolic changes may ultimately lead to the expansion of highly proliferating, differentiation-arrested malignant cells. A multitude of studies over the past two decades have investigated the mechanisms underlying the closely intertwined phenomena of drug resistance and disease progression. Here, we provide an update on what is currently known on the mechanisms underlying progression and present the latest acquisitions on BCR-ABL1-independent resistance and leukemia stem cell persistence.
Collapse
|
34
|
Bonifacio M, Stagno F, Scaffidi L, Krampera M, Di Raimondo F. Management of Chronic Myeloid Leukemia in Advanced Phase. Front Oncol 2019; 9:1132. [PMID: 31709190 PMCID: PMC6823861 DOI: 10.3389/fonc.2019.01132] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Management of chronic myeloid leukemia (CML) in advanced phases remains a challenge also in the era of tyrosine kinase inhibitors (TKIs) treatment. Cytogenetic clonal evolution and development of resistant mutations represent crucial events that limit the benefit of subsequent therapies in these patients. CML is diagnosed in accelerated (AP) or blast phase (BP) in <5% of patients, and the availability of effective treatments for chronic phase (CP) has dramatically reduced progressions on therapy. Due to smaller number of patients, few randomized studies are available in this setting and evidences are limited. Nevertheless, three main scenarios may be drawn: (a) patients diagnosed in AP are at higher risk of failure as compared to CP patients, but if they achieve optimal responses with frontline TKI treatment their outcome may be similarly favorable; (b) patients diagnosed in BP may be treated with TKI alone or with TKI together with conventional chemotherapy regimens, and subsequent transplant decisions should rely on kinetics of response and individual transplant risk; (c) patients in CP progressing under TKI treatment represent the most challenging population and they should be treated with alternative TKI according to the mutational profile, optional chemotherapy in BP patients, and transplant should be considered in suitable cases after return to second CP. Due to lack of validated and reliable markers to predict blast crisis and the still unsatisfactory results of treatments in this setting, prevention of progression by careful selection of frontline treatment in CP and early treatment intensification in non-optimal responders remains the main goal. Personalized evaluation of response kinetics could help in identifying patients at risk for progression.
Collapse
Affiliation(s)
| | - Fabio Stagno
- Division of Hematology With BMT, AOU Policlinico “Vittorio Emanuele”, University of Catania, Catania, Italy
| | - Luigi Scaffidi
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Francesco Di Raimondo
- Division of Hematology With BMT, AOU Policlinico “Vittorio Emanuele”, University of Catania, Catania, Italy
| |
Collapse
|
35
|
Kawai H, Matsushita H, Suzuki R, Kitamura Y, Ogawa Y, Kawada H, Ando K. Overcoming Tyrosine Kinase Inhibitor Resistance in Transformed Cell Harboring SEPT9-ABL1 Chimeric Fusion Protein. Neoplasia 2019; 21:788-801. [PMID: 31276931 PMCID: PMC6611969 DOI: 10.1016/j.neo.2019.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Hematological malignancies harboring various ABL1 fusions are expected to be sensitive to tyrosine kinase inhibitors (TKIs), similar to those with BCR-ABL1. However, SEPT9-ABL1 exhibits TKI resistance both in vitro and in vivo. SEPT9-ABL1 has the same ABL1 region as seen in BCR-ABL1 but no point mutation in its kinase domain, which is one of the main mechanisms underlying TKI resistance in the leukemic cells harboring BCR-ABL1. The purpose of this study was to reveal the mechanism underlying TKI resistance induced by SEPT9-ABL1. We focused on the TP53 status because TKI-induced apoptosis in BCR-ABL1–positive cells is achieved through TP53. Mouse TP53 homologue TRP53 was downregulated and less phosphorylated in the cells expressing SEPT9-ABL1 than in those with BCR-ABL1, resulting in the prevention of apoptosis induced by TKIs. The CRM1 inhibitor KPT-330 accumulated nuclear TRP53 and NFKB1A (also known as IκBα), which is thought to capture TRP53 in the cytoplasm, and induced apoptosis in the hematopoietic cells expressing SEPT9-ABL1. In addition, the combination treatment of KPT-330 and imatinib, which induced the marked nuclear accumulation of PP2A and SET, reactivated PP2A through its dephosphorylation and inhibited SET expression, resulting in the effective induction of the apoptosis in the cells expressing SEPT9-ABL1. The combination treatment with KPT-330 and imatinib successfully reduced the subcutaneous masses expressing SEPT9-ABL1 and extended the survival of the mice intraperitoneally transplanted with SEPT9-ABL1–expressing cells. These results show that therapy with CRM1 inhibitors may be effective for overcoming TKI resistance induced by SEPT9-ABL1.
Collapse
Affiliation(s)
- Hidetsugu Kawai
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiromichi Matsushita
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan.
| | - Rikio Suzuki
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yuka Kitamura
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshiaki Ogawa
- Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiroshi Kawada
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Kiyoshi Ando
- Research Center for Cancer Stem Cell, Tokai University School of Medicine, Isehara, Kanagawa, Japan; Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
36
|
Meenakshi Sundaram DN, Jiang X, Brandwein JM, Valencia-Serna J, Remant KC, Uludağ H. Current outlook on drug resistance in chronic myeloid leukemia (CML) and potential therapeutic options. Drug Discov Today 2019; 24:1355-1369. [PMID: 31102734 DOI: 10.1016/j.drudis.2019.05.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/25/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Chronic myeloid leukemia cells are armed with several resistance mechanisms that can make current drugs ineffective. A better understanding of resistance mechanisms is yielding new approaches to management of the disease. Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm the hallmark of which, the breakpoint cluster region-Abelson (BCR-ABL) oncogene, has been the target of tyrosine kinase inhibitors (TKIs), which have significantly improved the survival of patients with CML. However, because of an increase in TKI resistance, it is becoming imperative to identify resistance mechanisms so that drug therapies can be better prescribed and new agents developed. In this review, we discuss the various BCR-ABL-dependent and -independent mechanisms of resistance observed in CML, and the range of therapeutic solutions available to overcome such resistance and to ultimately improve the survival of patients with CML.
Collapse
Affiliation(s)
| | - Xiaoyan Jiang
- Terry Fox Laboratory, British Columbia Cancer Agency and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | | - Juliana Valencia-Serna
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | - K C Remant
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
37
|
|
38
|
Stella S, Zammit V, Vitale SR, Pennisi MS, Massimino M, Tirrò E, Forte S, Spitaleri A, Antolino A, Siracusa S, Accurso V, Mannina D, Impera S, Musolino C, Russo S, Malato A, Mineo G, Musso M, Porretto F, Martino B, Di Raimondo F, Manzella L, Vigneri P, Stagno F. Clinical Implications of Discordant Early Molecular Responses in CML Patients Treated with Imatinib. Int J Mol Sci 2019; 20:ijms20092226. [PMID: 31064152 PMCID: PMC6539817 DOI: 10.3390/ijms20092226] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 11/16/2022] Open
Abstract
A reduction in BCR-ABL1/ABL1IS transcript levels to <10% after 3 months or <1% after 6 months of tyrosine kinase inhibitor therapy are associated with superior clinical outcomes in chronic myeloid leukemia (CML) patients. In this study, we investigated the reliability of multiple BCR-ABL1 thresholds in predicting treatment outcomes for 184 subjects diagnosed with CML and treated with standard-dose imatinib mesylate (IM). With a median follow-up of 61 months, patients with concordant BCR-ABL1/ABL1IS transcripts below the defined thresholds (10% at 3 months and 1% at 6 months) displayed significantly superior rates of event-free survival (86.1% vs. 26.6%) and deep molecular response (≥ MR4; 71.5% vs. 16.1%) compared to individuals with BCR-ABL1/ABL1IS levels above these defined thresholds. We then analyzed the outcomes of subjects displaying discordant molecular transcripts at 3- and 6-month time points. Among these patients, those with BCR-ABL1/ABL1IS values >10% at 3 months but <1% at 6 months fared significantly better than individuals with BCR-ABL1/ABL1IS <10% at 3 months but >1% at 6 months (event-free survival 68.2% vs. 32.7%; p < 0.001). Likewise, subjects with BCR-ABL1/ABL1IS at 3 months >10% but <1% at 6 months showed a higher cumulative incidence of MR4 compared to patients with BCR-ABL1/ABL1IS <10% at 3 months but >1% at 6 months (75% vs. 18.2%; p < 0.001). Finally, lower BCR-ABL1/GUSIS transcripts at diagnosis were associated with BCR-ABL1/ABL1IS values <1% at 6 months (p < 0.001). Our data suggest that when assessing early molecular responses to therapy, the 6-month BCR-ABL1/ABL1IS level displays a superior prognostic value compared to the 3-month measurement in patients with discordant oncogenic transcripts at these two pivotal time points.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Female
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Male
- Middle Aged
- Protein Kinase Inhibitors/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Valentina Zammit
- Division of Hematology and Bone Marrow Transplant, AOU Policlinico - V. Emanuele, 95123 Catania, Italy.
| | - Silvia Rita Vitale
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy.
| | - Antonio Spitaleri
- Division of Hematology and Bone Marrow Transplant, AOU Policlinico - V. Emanuele, 95123 Catania, Italy.
| | - Agostino Antolino
- Department of Transfusional Medicine, Maria Paternò-Arezzo Hospital, 97100 Ragusa, Italy.
| | - Sergio Siracusa
- Division of Hematology, A.O.U. Policlinico "P. Giaccone", University of Palermo, 90127 Palermo, Italy.
| | - Vincenzo Accurso
- Division of Hematology, A.O.U. Policlinico "P. Giaccone", University of Palermo, 90127 Palermo, Italy.
| | - Donato Mannina
- Division of Hematology, Papardo Hospital, 98158 Messina, Italy.
| | - Stefana Impera
- Division of Oncology and Hematology, ARNAS Garibaldi-Nesima, 95122 Catania, Italy.
| | - Caterina Musolino
- Division of Hematology, University of Messina, 98125 Messina, Italy.
| | - Sabina Russo
- Division of Hematology, University of Messina, 98125 Messina, Italy.
| | - Alessandra Malato
- Division of Hematology and Bone Marrow Transplant, Villa Sofia-Cervello Hospital, 90146 Palermo, Italy.
| | - Giuseppe Mineo
- Division of Hematology, San Vincenzo Hospital, 98039 Taormina, Italy.
| | - Maurizio Musso
- Division of Hematology, La Maddalena Hospital, 90146 Palermo, Italy.
| | | | - Bruno Martino
- Hematology Department, Grande Ospedale Metropolitano, Reggio Calabria, 89124 Reggio Calabria, Italy.
| | - Francesco Di Raimondo
- Division of Hematology and Bone Marrow Transplant, AOU Policlinico - V. Emanuele, 95123 Catania, Italy.
- Department of Surgery, Medical and Surgical Specialities, University of Catania, 95123 Catania, Italy.
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95123 Catania, Italy.
| | - Fabio Stagno
- Division of Hematology and Bone Marrow Transplant, AOU Policlinico - V. Emanuele, 95123 Catania, Italy.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Chronic myeloid leukemia (CML) is hallmarked by the presence of fusion protein kinase derived from a reciprocal translocation between chromosome 9 and 22, breakpoint cluster region (BCR)-Abelson leukemia virus (ABL) 1, causing aberrant regulation of the downstream pathways leading to unchecked CML leukemia stem cells (LSCs) proliferation. Since the discovery of tyrosine kinase inhibitors (TKI), CML, once a fatal disease, has become a chronic illness if managed appropriately. Changing treatment landscape has unsurfaced the challenge of TKI resistance that is clinically difficult to overcome. RECENT FINDINGS In this review, we discuss the concept of TKI resistance and pathways leading to the resistance which allows for a survival advantage to CML LSCs. Aside from BCR-ABL-dependent mechanisms of resistance which involves aberrant expression in the regulatory pumps involving efflux and influx of the TKI affecting drug bioavailability, activation of alternate survival pathways may be accountable for primary or secondary resistance. Activation of these pathways, intrinsically and extrinsically to LSCs, may be mediated through various upstream and downstream signaling as well as conditions affecting the microenvironment. Several therapeutic approaches that combine TKI with an additional agent that inhibits the activation of an alternate pathway have been studied as part of clinical trials which we will discuss here. SUMMARY We categorize the resistance into BCR-ABL-dependent and BCR-ABL-independent subgroups to further describe the complex molecular pathways which can potentially serve as a therapeutic target. We further discuss novel combination strategies currently in early or advanced phase clinical trials aimed to overcome the TKI resistance. We further highlight the need for further research despite the tremendous strides already made in the management of CML.
Collapse
|
40
|
Bach DH, Long NP, Luu TTT, Anh NH, Kwon SW, Lee SK. The Dominant Role of Forkhead Box Proteins in Cancer. Int J Mol Sci 2018; 19:E3279. [PMID: 30360388 PMCID: PMC6213973 DOI: 10.3390/ijms19103279] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/16/2022] Open
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that are significantly implicated in cancer, with various critical roles in biological processes. Herein, we provide an overview of several key members of the FOXA, FOXC, FOXM1, FOXO and FOXP subfamilies. Important pathophysiological processes of FOX transcription factors at multiple levels in a context-dependent manner are discussed. We also specifically summarize some major aspects of FOX transcription factors in association with cancer research such as drug resistance, tumor growth, genomic alterations or drivers of initiation. Finally, we suggest that targeting FOX proteins may be a potential therapeutic strategy to combat cancer.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | | | | | - Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
41
|
miR-217 sensitizes chronic myelogenous leukemia cells to tyrosine kinase inhibitors by targeting pro-oncogenic anterior gradient 2. Exp Hematol 2018; 68:80-88.e2. [PMID: 30195077 DOI: 10.1016/j.exphem.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/30/2018] [Accepted: 09/01/2018] [Indexed: 12/21/2022]
Abstract
BCR-ABL1-independent mechanisms had been thought to mediate drug resistance to tyrosine kinase inhibitors (TKIs) in patients with chronic myelogenous leukemia (CML). The pro-oncogenic anterior gradient 2 (AGR2) mediates drug resistance of cancer cells. In this study, we observed an increased level of AGR2 in TKI-resistant CML cells. Silence of AGR2 in dasatinib-resistant K562 (K562DR) cells led to restored sensitivity to dasatinib both in vitro and in vivo. Exposure to dasatinib induced upregulation of AGR2 in K562 cells, which indicated a probable treatment-related drug resistance. We further investigated the potential interaction between microRNA (miRNA) and AGR2 in K562DR cells and found that downregulation of miR-217 was associated with overexpression of AGR2 in K562DR cells. Luciferase reporter assay identified that miR-217 negatively regulated expression of AGR2 through binding the 3'-untranslated region of AGR2. Hypermethylation of the CpG island on the promoter region of the MIR217 gene is a probable reason for the downregulation of miR-217 in dasatinib-treated K562 cells. Forced expression of miR-217 led to decreased expression of AGR2 as well as compromised TKI-resistant potential of K562DR cells. Similarly, overexpression of miR-217 resensitized K562DR cells to dasatinib treatment in a murine xenograft transplantation model. TKI treatment-induced drug resistance is correlated with a decrease of miR-217 and upregulation of AGR2. The miR-217/AGR2 interaction might be a potential therapeutic target in treating CML patients with TKI resistance.
Collapse
|
42
|
Borges CDS, Ferreira AF, Almeida VH, Gomes FG, Berzoti-Coelho MG, Cacemiro MDC, Nunes NS, Figueiredo-Pontes LL, Simões BP, Castro FA, Monteiro RQ. Crosstalk between BCR-ABL and protease-activated receptor 1 (PAR1) suggests a novel target in chronic myeloid leukemia. Exp Hematol 2018; 66:50-62. [PMID: 30076949 DOI: 10.1016/j.exphem.2018.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 06/19/2018] [Accepted: 07/26/2018] [Indexed: 12/31/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm characterized by the presence of the Philadelphia chromosome, which generates the oncogene BCR-ABL1. Protease-activated receptor 1 (PAR1) is involved in tumor progression and angiogenesis. We have previously reported that PAR1 expression is elevated in human leukemias that display a more aggressive clinical behavior, including the blast crisis of CML. In this study, we analyzed the crosstalk between the oncoprotein BCR-ABL and PAR1 in CML. Leukemic cell lines transfected with the BCR-ABL1 oncogene showed significantly higher expression levels of PAR1 compared with that of wild-type counterparts. This phenomenon was reversed by treatment with tyrosine kinase inhibitors (TKIs). Conversely, treatment with the PAR1 antagonist SCH79797 inhibited BCR-ABL expression. The PAR1 antagonist induced apoptosis in a dose- and time-dependent manner. Higher vascular endothelial growth factor (VEGF) levels were observed in cells transfected with BCR-ABL1 than in their wild-type counterparts. VEGF expression was strongly inhibited after treatment with either TKIs or the PAR1 antagonist. Finally, we evaluated PAR1 expression in CML patients who were either in the blast or chronic phases and had either received TKI treatment or no treatment. A significant decrease in PAR1 expression was observed in treatment-responsive patients, as opposed to a significant increase in PAR1 expression levels in treatment-resistant patients. Patients classified as high risk according to the Sokal index showed higher PAR1 expression levels. Our results demonstrate the crosstalk between BCR-ABL and PAR1. These data may offer important insight into the development of new therapeutic strategies for CML.
Collapse
Affiliation(s)
- Camilla de S Borges
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline F Ferreira
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Vitor H Almeida
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fausto G Gomes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Gabriela Berzoti-Coelho
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Maira da Costa Cacemiro
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Natalia S Nunes
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Lorena L Figueiredo-Pontes
- Hematology Division, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Belinda P Simões
- Hematology Division, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Fabíola A Castro
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Robson Q Monteiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
43
|
Srutova K, Curik N, Burda P, Savvulidi F, Silvestri G, Trotta R, Klamova H, Pecherkova P, Sovova Z, Koblihova J, Stopka T, Perrotti D, Polakova KM. BCR-ABL1 mediated miR-150 downregulation through MYC contributed to myeloid differentiation block and drug resistance in chronic myeloid leukemia. Haematologica 2018; 103:2016-2025. [PMID: 30049824 PMCID: PMC6269310 DOI: 10.3324/haematol.2018.193086] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022] Open
Abstract
The fusion oncoprotein BCR-ABL1 exhibits aberrant tyrosine kinase activity and it has been proposed that it deregulates signaling networks involving both transcription factors and non-coding microRNAs that result in chronic myeloid leukemia (CML). Previously, microRNA expression profiling showed deregulated expression of miR-150 and miR-155 in CML. In this study, we placed these findings into the broader context of the MYC/miR-150/MYB/miR-155/PU.1 oncogenic network. We propose that up-regulated MYC and miR-155 in CD34+ leukemic stem and progenitor cells, in concert with BCR-ABL1, impair the molecular mechanisms of myeloid differentiation associated with low miR-150 and PU.1 levels. We revealed that MYC directly occupied the -11.7 kb and -0.35 kb regulatory regions in the MIR150 gene. MYC occupancy was markedly increased through BCR-ABL1 activity, causing inhibition of MIR150 gene expression in CML CD34+ and CD34- cells. Furthermore, we found an association between reduced miR-150 levels in CML blast cells and their resistance to tyrosine kinase inhibitors (TKIs). Although TKIs successfully disrupted BCR-ABL1 kinase activity in proliferating CML cells, this treatment did not efficiently target quiescent leukemic stem cells. The study presents new evidence regarding the MYC/miR-150/MYB/miR-155/PU.1 leukemic network established by aberrant BCR-ABL1 activity. The key connecting nodes of this network may serve as potential druggable targets to overcome resistance of CML stem and progenitor cells.
Collapse
Affiliation(s)
- Klara Srutova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Nikola Curik
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Institute of Pathological Physiology, First Medical Faculty, Charles University, Prague, Czech Republic
| | - Pavel Burda
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Institute of Pathological Physiology, First Medical Faculty, Charles University, Prague, Czech Republic
| | - Filipp Savvulidi
- Institute of Pathological Physiology, First Medical Faculty, Charles University, Prague, Czech Republic
| | - Giovannino Silvestri
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore, MD, USA
| | - Rossana Trotta
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore, MD, USA
| | - Hana Klamova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Institute of Clinical and Experimental Hematology, First Medical Faculty, Charles University, Prague, Czech Republic
| | - Pavla Pecherkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Zofie Sovova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jitka Koblihova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Tomas Stopka
- BIOCEV, First Medical Faculty, Charles University, Vestec, Czech Republic
| | - Danilo Perrotti
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore, MD, USA
| | - Katerina Machova Polakova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic .,Institute of Clinical and Experimental Hematology, First Medical Faculty, Charles University, Prague, Czech Republic
| |
Collapse
|
44
|
Targeting PFKFB3 sensitizes chronic myelogenous leukemia cells to tyrosine kinase inhibitor. Oncogene 2018; 37:2837-2849. [DOI: 10.1038/s41388-018-0157-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 01/10/2018] [Accepted: 01/14/2018] [Indexed: 01/20/2023]
|
45
|
Wagle MC, Kirouac D, Klijn C, Liu B, Mahajan S, Junttila M, Moffat J, Merchant M, Huw L, Wongchenko M, Okrah K, Srinivasan S, Mounir Z, Sumiyoshi T, Haverty PM, Yauch RL, Yan Y, Kabbarah O, Hampton G, Amler L, Ramanujan S, Lackner MR, Huang SMA. A transcriptional MAPK Pathway Activity Score (MPAS) is a clinically relevant biomarker in multiple cancer types. NPJ Precis Oncol 2018; 2:7. [PMID: 29872725 PMCID: PMC5871852 DOI: 10.1038/s41698-018-0051-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/29/2018] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
KRAS- and BRAF-mutant tumors are often dependent on MAPK signaling for proliferation and survival and thus sensitive to MAPK pathway inhibitors. However, clinical studies have shown that MEK inhibitors are not uniformly effective in these cancers indicating that mutational status of these oncogenes does not accurately capture MAPK pathway activity. A number of transcripts are regulated by this pathway and are recurrently identified in genome-based MAPK transcriptional signatures. To test whether the transcriptional output of only 10 of these targets could quantify MAPK pathway activity with potential predictive or prognostic clinical utility, we created a MAPK Pathway Activity Score (MPAS) derived from aggregated gene expression. In vitro, MPAS predicted sensitivity to MAPK inhibitors in multiple cell lines, comparable to or better than larger genome-based statistical models. Bridging in vitro studies and clinical samples, median MPAS from a given tumor type correlated with cobimetinib (MEK inhibitor) sensitivity of cancer cell lines originating from the same tissue type. Retrospective analyses of clinical datasets showed that MPAS was associated with the sensitivity of melanomas to vemurafenib (HR: 0.596) and negatively prognostic of overall or progression-free survival in both adjuvant and metastatic CRC (HR: 1.5 and 1.4), adrenal cancer (HR: 1.7), and HER2+ breast cancer (HR: 1.6). MPAS thus demonstrates potential clinical utility that warrants further exploration.
Collapse
Affiliation(s)
- Marie-Claire Wagle
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Daniel Kirouac
- 2Department of Pre-Clinical and Translational PKPD, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Christiaan Klijn
- 3Department of Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Bonnie Liu
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shilpi Mahajan
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Melissa Junttila
- 4Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - John Moffat
- 5Department of Biochemical and Cellular pharmacology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Mark Merchant
- 4Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Ling Huw
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Matthew Wongchenko
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Kwame Okrah
- 6Department of Biostatistics, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shrividhya Srinivasan
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Zineb Mounir
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Teiko Sumiyoshi
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Peter M Haverty
- 3Department of Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Robert L Yauch
- 4Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Yibing Yan
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Omar Kabbarah
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Garret Hampton
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Lukas Amler
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Saroja Ramanujan
- 2Department of Pre-Clinical and Translational PKPD, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Mark R Lackner
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shih-Min A Huang
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA.,7Present Address: Bristol-Myers Squibb, 3551 Lawrenceville Princeton, Lawrence Township, NJ 08648 USA
| |
Collapse
|
46
|
Massimino M, Stella S, Tirrò E, Romano C, Pennisi MS, Puma A, Manzella L, Zanghì A, Stagno F, Di Raimondo F, Vigneri P. Non ABL-directed inhibitors as alternative treatment strategies for chronic myeloid leukemia. Mol Cancer 2018; 17:56. [PMID: 29455672 PMCID: PMC5817805 DOI: 10.1186/s12943-018-0805-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/01/2018] [Indexed: 02/07/2023] Open
Abstract
The introduction of ABL Tyrosine Kinase Inhibitors (TKIs) has significantly improved the outcome of Chronic Myeloid Leukemia (CML) patients that, in large part, achieve satisfactory hematological, cytogenetic and molecular remissions. However, approximately 15-20% fail to obtain optimal responses according to the current European Leukemia Network recommendation because of drug intolerance or resistance.Moreover, a plethora of evidence suggests that Leukemic Stem Cells (LSCs) show BCR-ABL1-independent survival. Hence, they are unresponsive to TKIs, leading to disease relapse if pharmacological treatment is discontinued.All together, these biological events generate a subpopulation of CML patients in need of alternative therapeutic strategies to overcome TKI resistance or to eradicate LSCs in order to allow cure of the disease.In this review we update the role of "non ABL-directed inhibitors" targeting signaling pathways downstream of the BCR-ABL1 oncoprotein and describe immunological approaches activating specific T cell responses against CML cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Combined Modality Therapy
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy
- Signal Transduction/drug effects
- Treatment Outcome
Collapse
Affiliation(s)
- Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Chiara Romano
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Adriana Puma
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Antonino Zanghì
- Department of Surgical Medical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Fabio Stagno
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Francesco Di Raimondo
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Department of Surgery, Medical and Surgical Specialties, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy.
| |
Collapse
|
47
|
Soverini S, Mancini M, Bavaro L, Cavo M, Martinelli G. Chronic myeloid leukemia: the paradigm of targeting oncogenic tyrosine kinase signaling and counteracting resistance for successful cancer therapy. Mol Cancer 2018; 17:49. [PMID: 29455643 PMCID: PMC5817796 DOI: 10.1186/s12943-018-0780-6] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Deregulated activity of BCR-ABL1, a nonreceptor tyrosine kinase encoded by the fusion gene resulting from the t(9;22)(q34;q11) chromosomal translocation, is thought to be the driver event responsible for initiation and maintenance of chronic myeloid leukemia (CML). BCR-ABL1 was one of the first tyrosine kinases to be implicated in a human malignancy and the first to be successfully targeted. Imatinib mesylate, the first tyrosine kinase inhibitor (TKI) to be approved for therapeutic use, was hailed as a magic bullet against cancer and remains one of the safest and most effective anticancer agents ever developed. Second- and third-generation TKIs were later introduced to prevent or counteract the problem of drug resistance, that may arise in a small proportion of patients. They are more potent molecules, but have been associated to more serious side effects and complications. Patients achieving stable optimal responses to TKI therapy are predicted to have the same life expectancy of the general population. However, TKIs do not ‘cure’ CML. Only a small proportion of cases may attempt therapy discontinuation without experiencing subsequent relapse. The great majority of patients will have to assume TKIs indefinitely – which raises serious pharmacoeconomic concerns and is now shifting the focus from efficacy to compliance and quality of life issues. Here we retrace the steps that have led from the biological acquisitions regarding BCR-ABL1 structure and function to the development of inhibitory strategies and we discuss drug resistance mechanism and how they can be addressed.
Collapse
Affiliation(s)
- Simona Soverini
- Hematology/Oncology "L. e A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Manuela Mancini
- Hematology/Oncology "L. e A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Luana Bavaro
- Hematology/Oncology "L. e A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Michele Cavo
- Hematology/Oncology "L. e A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Giovanni Martinelli
- Hematology/Oncology "L. e A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
48
|
Khosravi A, Alizadeh S, Jalili A, Shirzad R, Saki N. The impact of Mir-9 regulation in normal and malignant hematopoiesis. Oncol Rev 2018; 12:348. [PMID: 29774136 PMCID: PMC5939831 DOI: 10.4081/oncol.2018.348] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNA-9 (MiR-9) dysregulation has been observed in various cancers. Recently, MiR-9 is considered to have a part in hematopoiesis and hematologic malignancies. However, its importance in blood neoplasms is not yet well defined. Thus, this study was conducted in order to assess the significance of MiR-9 role in the development of hematologic neoplasia, prognosis, and treatment approaches. We have shown that a large number of MiR-9 targets (such as FOXOs, SIRT1, CCND1, ID2, CCNG1, Ets, and NFkB) play essential roles in leukemogenesis and that it is overexpressed in different leukemias. Our findings indicated MiR-9 downregulation in a majority of leukemias. However, its overexpression was reported in patients with dysregulated MiR-9 controlling factors (such as MLLr). Additionally, prognostic value of MiR-9 has been reported in some types of leukemia. This study generally emphasizes on the critical role of MiR-9 in hematologic malignancies as a prognostic factor and a therapeutic target.
Collapse
Affiliation(s)
- Abbas Khosravi
- Transfusion Research Center, High Institute for Research and Education in Transfusion Medi-cine, Tehran
| | - Shaban Alizadeh
- Hematology Department, Allied Medical School, Tehran University of Medical Sciences, Tehran
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology at Cell Science Re-search Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran
| | - Reza Shirzad
- WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jun-dishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
49
|
The regulation of FOXO1 and its role in disease progression. Life Sci 2017; 193:124-131. [PMID: 29158051 DOI: 10.1016/j.lfs.2017.11.030] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/14/2017] [Accepted: 11/16/2017] [Indexed: 12/27/2022]
Abstract
Cell proliferation, apoptosis, autophagy, oxidative stress and metabolic dysregulation are the basis of many diseases. Forkhead box transcription factor O1 (FOXO1) changes in response to cellular stimulation and maintains tissue homeostasis during the above-mentioned physiological and pathological processes. Substantial evidences indicate that FOXO1's function depends on the modulation of downstream targets such as apoptosis- and autophagy-associated genes, anti-oxidative stress enzymes, cell cycle arrest genes, and metabolic and immune regulators. In addition, oxidative stress, high glucose and other stimulations induce the regulation of FOXO1 activity via PI3k-Akt, JNK, CBP, Sirtuins, ubiquitin E3 ligases, etc., which mediate multiple signalling pathways. Subsequent post-transcriptional modifications, including phosphorylation, ubiquitination, acetylation, deacetylation, arginine methylation and O-GlcNAcylation, activate or inhibit FOXO1. The regulation of FOXO1 and its role might provide a significant avenue for the prevention and treatment of diseases. However, the subtle mechanisms of the post-transcriptional modifications and the effect of FOXO1 remain elusive and even conflicting in the development of many diseases. The determination of these questions potentially has implications for further research regarding FOXO1 signalling and the identification of targeted drugs.
Collapse
|
50
|
Patel AB, O'Hare T, Deininger MW. Mechanisms of Resistance to ABL Kinase Inhibition in Chronic Myeloid Leukemia and the Development of Next Generation ABL Kinase Inhibitors. Hematol Oncol Clin North Am 2017; 31:589-612. [PMID: 28673390 PMCID: PMC5505321 DOI: 10.1016/j.hoc.2017.04.007] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Chronic myeloid leukemia is increasingly viewed as a chronic illness; most patients have a life expectancy close to that of the general population. Despite progress made using BCR-ABL1 tyrosine kinase inhibitors (TKIs), drug resistance via BCR-ABL1-dependent and BCR-ABL1-independent mechanisms continues to be an issue. BCR-ABL1-dependent resistance is primarily mediated through oncoprotein kinase domain mutations and usually results in overt resistance to TKIs. However, BCR-ABL1-independent resistance in the setting of effective BCR-ABL1 inhibition is recognized as a major contributor to minimal residual disease. Efforts to eradicate persistent leukemic stem cells have focused on combination therapy.
Collapse
MESH Headings
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biological Availability
- Biomarkers
- Cell Survival/drug effects
- Cell Survival/genetics
- Dose-Response Relationship, Drug
- Drug Discovery
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/chemistry
- Fusion Proteins, bcr-abl/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- Immunotherapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Models, Molecular
- Molecular Targeted Therapy
- Mutation
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Signal Transduction/drug effects
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Ami B Patel
- Department of Hematology and Oncology, Huntsman Cancer Institute, 2000 Circle of Hope Drive, The University of Utah, Salt Lake City, UT 84112, USA
| | - Thomas O'Hare
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| | - Michael W Deininger
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA.
| |
Collapse
|