1
|
Golla U, Patel S, Shah N, Talamo S, Bhalodia R, Claxton D, Dovat S, Sharma A. From Deworming to Cancer Therapy: Benzimidazoles in Hematological Malignancies. Cancers (Basel) 2024; 16:3454. [PMID: 39456548 PMCID: PMC11506385 DOI: 10.3390/cancers16203454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Drug repurposing is a strategy to discover new therapeutic uses for existing drugs, which have well-established toxicity profiles and are often more affordable. This approach has gained significant attention in recent years due to the high costs and low success rates associated with traditional drug development. Drug repositioning offers a more time- and cost-effective path for identifying new treatments. Several FDA-approved non-chemotherapy drugs have been investigated for their anticancer potential. Among these, anthelmintic benzimidazoles (such as albendazole, mebendazole, and flubendazole) have garnered interest due to their effects on microtubules and oncogenic signaling pathways. Blood cancers, which frequently develop resistance and have high mortality rates, present a critical need for effective therapies. This review highlights the recent advances in repurposing benzimidazoles for blood malignancies. These compounds induce cell cycle arrest, differentiation, tubulin depolymerization, loss of heterozygosity, proteasomal degradation, and inhibit oncogenic signaling to exert their anticancer effects. We also discuss current limitations and strategies to overcome them, emphasizing the potential of combining benzimidazoles with standard therapies for improved treatment of hematological cancers.
Collapse
Affiliation(s)
- Upendarrao Golla
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Satyam Patel
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Nyah Shah
- Department of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Stella Talamo
- Department of Medicine, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA;
| | - Riya Bhalodia
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.B.); (S.D.)
| | - David Claxton
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.B.); (S.D.)
| | - Arati Sharma
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| |
Collapse
|
2
|
Kaehler M, von Bubnoff N, Cascorbi I, Gorantla SP. Molecular biomarkers of leukemia: convergence-based drug resistance mechanisms in chronic myeloid leukemia and myeloproliferative neoplasms. Front Pharmacol 2024; 15:1422565. [PMID: 39104388 PMCID: PMC11298451 DOI: 10.3389/fphar.2024.1422565] [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: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Leukemia represents a diverse group of hematopoietic neoplasms that can be classified into different subtypes based on the molecular aberration in the affected cell population. Identification of these molecular classification is required to identify specific targeted therapeutic approaches for each leukemic subtype. In general, targeted therapy approaches achieve good responses in some leukemia subgroups, however, resistance against these targeted therapies is common. In this review, we summarize molecular drug resistance biomarkers in targeted therapies in BCR::ABL1-driven chronic myeloid leukemia (CML) and JAK2-driven myeloproliferative neoplasms (MPNs). While acquisition of secondary mutations in the BCR::ABL1 kinase domain is the a common mechanism associated with TKI resistance in CML, in JAK2-driven MPNs secondary mutations in JAK2 are rare. Due to high prevalence and lack of specific therapy approaches in MPNs compared to CML, identification of crucial pathways leading to inhibitor persistence in MPN model is utterly important. In this review, we focus on different alternative signaling pathways activated in both, BCR::ABL1-mediated CML and JAK2-mediated MPNs, by combining data from in vitro and in vivo-studies that could be used as potential biomarkers of drug resistance. In a nutshell, some common similarities, especially activation of PDGFR, Ras, PI3K/Akt signaling pathways, have been demonstrated in both leukemias. In addition, induction of the nucleoprotein YBX1 was shown to be involved in TKI-resistant JAK2-mediated MPN, as well as TKI-resistant CML highlighting deubiquitinating enzymes as potential biomarkers of TKI resistance. Taken together, whole exome sequencing of cell-based or patients-derived samples are highly beneficial to define specific resistance markers. Additionally, this might be helpful for the development of novel diagnostic tools, e.g., liquid biopsy, and novel therapeutic agents, which could be used to overcome TKI resistance in molecularly distinct leukemia subtypes.
Collapse
Affiliation(s)
- Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sivahari Prasad Gorantla
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
3
|
Biersack B, Höpfner M. Emerging role of MYB transcription factors in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:15. [PMID: 38835346 PMCID: PMC11149108 DOI: 10.20517/cdr.2023.158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/19/2024] [Accepted: 04/04/2024] [Indexed: 06/06/2024]
Abstract
Decades ago, the viral myeloblastosis oncogene v-myb was identified as a gene responsible for the development of avian leukemia. However, the relevance of MYB proteins for human cancer diseases, in particular for solid tumors, remained basically unrecognized for a very long time. The human family of MYB transcription factors comprises MYB (c-MYB), MYBL2 (b-MYB), and MYBL1 (a-MYB), which are overexpressed in several cancers and are associated with cancer progression and resistance to anticancer drugs. In addition to overexpression, the presence of activated MYB-fusion proteins as tumor drivers was described in certain cancers. The identification of anticancer drug resistance mediated by MYB proteins and their underlying mechanisms are of great importance in understanding failures of current therapies and establishing new and more efficient therapy regimens. In addition, new drug candidates targeting MYB transcription factor activity and signaling have emerged as a promising class of potential anticancer therapeutics that could tackle MYB-dependent drug-resistant cancers in a more selective way. This review describes the correlation of MYB transcription factors with the formation and persistence of cancer resistance to various approved and investigational anticancer drugs.
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Bayreuth 95440, Germany
| | - Michael Höpfner
- Institute for Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin 10117, Germany
| |
Collapse
|
4
|
Lee CH, Hsu KW, Hsieh YY, Li WT, Long Y, Lin CY, Chen SH. Unveiling IL6R and MYC as Targeting Biomarkers in Imatinib-Resistant Chronic Myeloid Leukemia through Advanced Non-Invasive Apoptosis Detection Sensor Version 2 Detection. Cells 2024; 13:616. [PMID: 38607055 PMCID: PMC11011921 DOI: 10.3390/cells13070616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/30/2024] [Accepted: 03/30/2024] [Indexed: 04/13/2024] Open
Abstract
The management of chronic myelogenous leukemia (CML) has seen significant progress with the introduction of tyrosine kinase inhibitors (TKIs), particularly Imatinib. However, a notable proportion of CML patients develop resistance to Imatinib, often due to the persistence of leukemia stem cells and resistance mechanisms independent of BCR::ABL1 This study investigates the roles of IL6R, IL7R, and MYC in Imatinib resistance by employing CRISPR/Cas9 for gene editing and the Non-Invasive Apoptosis Detection Sensor version 2 (NIADS v2) for apoptosis assessment. The results indicate that Imatinib-resistant K562 cells (K562-IR) predominantly express IL6R, IL7R, and MYC, with IL6R and MYC playing crucial roles in cell survival and sensitivity to Imatinib. Conversely, IL7R does not significantly impact cytotoxicity, either alone or in combination with Imatinib. Further genetic editing experiments confirm the protective functions of IL6R and MYC in K562-IR cells, suggesting their potential as therapeutic targets for overcoming Imatinib resistance in CML. This study contributes to understanding the mechanisms of Imatinib resistance in CML, proposing IL6R and MYC as pivotal targets for therapeutic strategies. Moreover, the utilization of NIADS v2 enhances our capability to analyze apoptosis and drug responses, contributing to a deeper understanding of CML pathogenesis and treatment options.
Collapse
MESH Headings
- Humans
- Apoptosis
- Biomarkers
- Drug Resistance, Neoplasm
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Receptors, Interleukin-6
- Proto-Oncogene Proteins c-myc
Collapse
Affiliation(s)
- Chia-Hwa Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City 23561, Taiwan;
- Ph.D. Program in Medicine Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City 23561, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan;
| | - Kai-Wen Hsu
- Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan;
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung City 40402, Taiwan
- Program for Cancer Biology and Drug Discovery, Drug Development Center, China Medical University, Taichung City 40402, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Ting Li
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Yuqing Long
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK;
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Chun-Yu Lin
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan;
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Shu-Huey Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
5
|
Ye Y, Yang X, Li F, Liu W, Zhang W, Huang Z. c-myb is involved in CML progression and is a therapeutic target in the zebrafish CML model. Animal Model Exp Med 2024; 7:136-144. [PMID: 36300552 PMCID: PMC11079146 DOI: 10.1002/ame2.12282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/27/2022] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Despite the success of tyrosine kinase inhibitors in chronic myeloid leukemia (CML) therapy, CML still faces the challenges of drug resistance and progression to blast crisis. Twenty-five percent of patients have imatinib resistance and treatment difficulties due to heterogeneity after progression, but little is known about the mechanism. A key transcription factor in hematopoiesis, MYB, has been reported to increase abnormally in several types of aggressive blood disorders including CML. METHODS This study used a zebrafish model to explore the relationship between BCR/ABL1 and c-myb in CML progression. A CML zebrafish model was crossed with a c-myb hyperactivity transgenic line. RESULTS It was found that both exogenous BCR/ABL1 and c-myb could up-regulate the expression of neutrophil-related genes. More seriously, neutrophil accumulation was observed when BCR/ABL1 was combined with c-myb overexpression. Further studies showed that c-myb may be one of the downstream targets of BCR/ABL1 and the effect of BCR/ABL1 on neutrophils was c-myb dependent. Taking advantage of this inheritable in vivo model, it was shown that a combination of imatinib and flavopiridol, a cyclin-dependent kinase inhibitor targeting MYB, could more effectively alleviate the aggressive phenotype of the double transgene line. CONCLUSION In summary, this study suggests that c-myb acts downstream of BCR/ABL1 and is involved in CML progression and is therefore a risk factor and a valuable target for the treatment of CML progression. The model used in the study could be helpful in high-throughput drug screening in CML transformation.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Disease Models, Animal
- Disease Progression
- Flavonoids/pharmacology
- Flavonoids/therapeutic use
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Neutrophils/drug effects
- Neutrophils/metabolism
- Piperidines/pharmacology
- Piperidines/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Proto-Oncogene Proteins c-myb/genetics
- Proto-Oncogene Proteins c-myb/metabolism
- Zebrafish
Collapse
Affiliation(s)
- Yin Ye
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Xiaojun Yang
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Feifei Li
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Wei Liu
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Wenqing Zhang
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
6
|
Bansal M, Ansari S, Verma M. Role of miRNAs to control the progression of Chronic Myeloid Leukemia by their expression levels. Med Oncol 2024; 41:55. [PMID: 38216843 DOI: 10.1007/s12032-023-02278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 11/30/2023] [Indexed: 01/14/2024]
Abstract
Chronic Myeloid Leukemia (CML) is a myeloproliferative disorder distinguished by a specific genetic anomaly known as a reciprocal translocation between chromosomes 9 and 22. This translocation causes fusion between the BCR and ABL regions. Consequently, BCR::ABL oncoprotein is formed, which plays a significant role in driving CML progression. Imatinib, a tyrosine kinase inhibitor (TKI), became the first line of drugs against CML. However, with continuous treatment, patients developed resistance against it. Indeed, to address this challenge, microRNA-based therapy emerges as a promising approach. miRNAs are 20-25 nucleotides long and hold great significance in various cellular processes, including cell differentiation, proliferation, migration, and apoptosis. In several malignancies, it has been reported that miRNAs might help to promote or prevent tumourigenesis and abnormal expression because they could act as both oncogenes/tumor suppressors. Recently, because of their vital regulatory function in maintaining cell homeostasis, miRNAs might be used to control CML progression and in developing new therapies for TKI-resistant patients. They might also act as potential prognostic, diagnostic, and therapeutic biomarkers based on their expression profiles. Various annotation tools and microarray-based expression profiles can be used to predict dysregulated miRNAs and their target genes. The main purpose of this review is to provide brief insights into the role of dysregulated miRNAs in CML pathogenesis and to emphasize their clinical relevance, such as their significant potential as therapeutics against CML. Utilizing these miRNAs as a therapeutic approach by inhibition or amplification of their activity could unlock new doors for the therapy of CML.
Collapse
MESH Headings
- Humans
- MicroRNAs/genetics
- Fusion Proteins, bcr-abl
- Drug Resistance, Neoplasm/genetics
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Apoptosis
Collapse
Affiliation(s)
- Manvi Bansal
- School of Biotechnology, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Sana Ansari
- School of Biotechnology, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Malkhey Verma
- School of Biotechnology, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
7
|
Parsa-Kondelaji M, Musavi M, Barzegar F, Abbasian N, Rostami M, R Seyedtaghia M, S Hashemi S, Modi M, Nikfar B, A Momtazi-Borojeni A. Dysregulation of miRNA expression in patients with chronic myelogenous leukemia at diagnosis: a systematic review. Biomark Med 2023; 17:1021-1029. [PMID: 38230979 DOI: 10.2217/bmm-2023-0575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024] Open
Abstract
Aim: The present systematic review aimed to explore miRNAs as a potential biomarker for early diagnosis of chronic myeloid leukemia (CML). Materials & methods: A systematic search was conducted in three electronic databases, including Web of Science, Scopus and PubMed, to obtain relevant articles investigating the alteration of miRNA expression in patients with CML. Results: The authors found miRNAs whose expression changes are effective in the induction of CML disease. Among them, miR-21 and miR-155 were identified as the most common miRNAs with increased expression and miR-150 and miR-146 as the most common miRNAs with decreased expression. Conclusion: miRNAs can be used as an indicator for the early detection and treatment of CML phase.
Collapse
Affiliation(s)
- Mohammad Parsa-Kondelaji
- Department of Hematology & Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Musavi
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Faezeh Barzegar
- Department of Hematology & Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neda Abbasian
- Hematology & Blood Banking Center, Emam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Rostami
- Department of Hematology & Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad R Seyedtaghia
- Department of Medical Genetics & Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyyed S Hashemi
- Department of Medical Genetics & Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdiyeh Modi
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amir A Momtazi-Borojeni
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
8
|
Li J, Malouf C, Miles LA, Willis MB, Pietras EM, King KY. Chronic inflammation can transform the fate of normal and mutant hematopoietic stem cells. Exp Hematol 2023; 127:8-13. [PMID: 37647982 DOI: 10.1016/j.exphem.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
Chronic inflammation, although subtle, puts the body in a constant state of alertness and is associated with many diseases, including cancer and cardiovascular diseases. It leads hematopoietic cells to produce and release proinflammatory cytokines, which trigger specific signaling pathways in hematopoietic stem cells (HSCs) that cause changes in proliferation, differentiation, and migration. This response is essential when HSCs are needed to produce specific blood cells to eliminate an intruder, such as a pathogenic virus, but mutant HSCs can use these proinflammatory signals to their advantage and accelerate the development of hematologic disease or malignancy. Understanding this complex process is vital for monitoring and controlling disease progression in patients. In the 2023 International Society for Experimental Hematology winter webinar, Dr. Eric Pietras (University of Colorado Anschutz Medical Campus, United States) and Dr. Katherine Y. King (Baylor College of Medicine, United States) gave a presentation on this topic, which is summarized in this review article.
Collapse
Affiliation(s)
- Jingjing Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia.
| | | | - Linde A Miles
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Mara B Willis
- Center for Cell and Gene Therapy and Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX
| | - Eric M Pietras
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Katherine Y King
- Center for Cell and Gene Therapy and Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX
| |
Collapse
|
9
|
Chai C, Sui K, Tang J, Yu H, Yang C, Zhang H, Li SC, Zhong JF, Wang Z, Zhang X. BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution. Theranostics 2023; 13:3943-3963. [PMID: 37554265 PMCID: PMC10405834 DOI: 10.7150/thno.83178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/12/2023] [Indexed: 08/10/2023] Open
Abstract
Rationale: In the bone marrow microenvironment (BMME), mesenchymal stem/stromal cells (MSCs) control the self-renewal of both healthy and cancerous hematopoietic stem/progenitor cells (HSPCs). We previously showed that in vivo leukemia-derived MSCs change neighbor MSCs into leukemia-permissive states and boost leukemia cell proliferation, survival, and chemotherapy resistance. But the mechanisms behind how the state changes are still not fully understood. Methods: Here, we took a reverse engineering approach to determine BCR-ABL1+ leukemia cells activated transcriptional factor C/EBPβ, resulting in miR130a/b-3p production. Then, we back-tracked from clinical specimen transcriptome sequencing to cell co-culture, molecular and cellular assays, flow cytometry, single-cell transcriptome, and transcriptional regulation to determine the molecular mechanisms of BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications. Results: BCR-ABL1-driven exosome-miR130a/b-3p mediated gap-junction Cx43 (a.k.a., GJA1) BMSC intercellular communications for subclonal evolution in leukemic microenvironment by targeting BMSCs-expressed HLAs, thereby potentially maintaining BMSCs with self-renewal properties and reduced BMSC immunogenicity. The Cx43low and miR-130a/bhigh subclonal MSCs subsets of differentiation state could be reversed to Cx43high and miR-130a/blow subclones of the higher stemness state in Cx43-overexpressed subclonal MSCs. Both miR-130a and miR-130b might only inhibit Cx43 translation or degrade Cx43 proteins and did not affect Cx43 mRNA stability. The subclonal evolution was further confirmed by single-cell transcriptome profiling of MSCs, which suggested that Cx43 regulated their stemness and played normal roles in immunomodulation antigen processing. Thus, upregulated miR-130a/b promoted osteogenesis and adipogenesis from BMSCs, thereby decreasing cancer progression. Our clinical data validated that the expression of many genes in human major histocompatibility was negatively associated with the stemness of MSCs, and several immune checkpoint proteins contributing to immune escape in tumors were overexpressed after either miR-130a or miR-130b overexpression, such as CD274, LAG3, PDCD1, and TNFRSF4. Not only did immune response-related cytokine-cytokine receptor interactions and PI3K-AKT pathways, including EGR3, TNFRSF1B, but also NDRG2 leukemic-associated inflammatory factors, such as IFNB1, CXCL1, CXCL10, and CCL7 manifest upon miR-130a/b overexpression. Either BCR siRNAs or ABL1 siRNAs assay showed significantly decreased miR-130a and miR-130b expression, and chromatin immunoprecipitation sequencing confirmed that the regulation of miR-130a and miR-130b expression is BCR-ABL1-dependent. BCR-ABL1 induces miR-130a/b expression through the upregulation of transcriptional factor C/EBPβ. C/EBPβ could bind directly to the promoter region of miR-130b-3p, not miR-130a-3p. BCR-ABL1-driven exosome-miR130a-3p could interact with Cx43, and further impact GJIC in TME. Conclusion: Our findings shed light on how leukemia BCR-ABL1-driven exosome-miR130b-3p could interact with gap-junction Cx43, and further impact GJIC in TME, implications for leukemic therapies of subclonal evolution.
Collapse
Affiliation(s)
- Chengyan Chai
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Ke Sui
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Jun Tang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
| | - Hao Yu
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Chao Yang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Hongyang Zhang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC), 1201 La Veta Ave., Orange, CA 92868-3874, United States of America
- Department of Neurology, University of California-Irvine School of Medicine, 200 S. Manchester Ave. Ste. 206, Orange, CA 92868, United States of America
| | - Jiang F. Zhong
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354, United States of America
| | - Zheng Wang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Xi Zhang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
10
|
Xu Y, Wang Z, Zhang L, Gao C, Li F, Li X, Ke Y, Liu HM, Hu Z, Wei L, Chen ZS. Differentiation of imatinib -resistant chronic myeloid leukemia cells with BCR-ABL-T315I mutation induced by Jiyuan Oridonin A. J Cancer 2023; 14:1182-1194. [PMID: 37215441 PMCID: PMC10197941 DOI: 10.7150/jca.83219] [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: 02/04/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Chronic myeloid leukemia (CML) results from BCR-ABL oncogene, which blocks CML cells differentiation and protects these cells from apoptosis. T315I mutated BCR-ABL is the main cause of the resistance mediated by imatinib and second generation BCR-ABL inhibitor. CML with the T315I mutation has been considered to have poor prognosis. Here, we determined the effect of Jiyuan oridonin A (JOA), an ent-kaurene diterpenoid compound, on the differentiation blockade in imatinib-sensitive, particularly, imatinib-resistant CML cells with BCR-ABL-T315I mutation by cell proliferation assay, apoptosis analysis, cell differentiation analysis, cell cycle analysis and colony formation assay. We also investigated the possible molecular mechanism by mRNA sequencing, qRT-PCR and Western blotting. We found that JOA at lower concentration significantly inhibited the proliferation of CML cells expressing mutant BCR-ABL (T315I mutation included) and wild-type BCR-ABL, which was due to that JOA induced the cell differentiation and the cell cycle arrest at G0/G1 phase. Interestingly, JOA possessed stronger anti-leukemia activity than its analogues such as OGP46 and Oridonin, which has been investigated extensively. Mechanistically, the cell differentiation mediated by JOA may be originated from the inhibition of BCR-ABL/c-MYC signaling in CML cells expressing wild-type BCR-ABL and BCR-ABL-T315I. JOA displayed the activity of inhibiting the BCR-ABL and promoted differentiation of not only imatinib -sensitive but also imatinib -resistant cells with BCR-ABL mutation, which could become a potent lead compound to overcome the imatinib -resistant induced by inhibitors of BCR-ABL tyrosine kinase in CML therapy.
Collapse
Affiliation(s)
- Yun Xu
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Ziting Wang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Lei Zhang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Congying Gao
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Fahui Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Xueming Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Yu Ke
- School of Pharmacy, Zhengzhou University, Zhengzhou, 450052, China
| | - Hong-Min Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenbo Hu
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang 261042, China
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| |
Collapse
|
11
|
Rassaei N, Abbaszade Dibavar M, Soleimani M, Atashi A, Mohammadi MH, Allahbakhshian Farsani M, Shahsavan S. The effect of microvesicles derived from K562 cells on proliferation and apoptosis of human bone marrow mesenchymal stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:295-300. [PMID: 36865039 PMCID: PMC9922373 DOI: 10.22038/ijbms.2023.66903.14675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/30/2022] [Indexed: 03/04/2023]
Abstract
Objectives Microvesicles (MVs) are small membrane-bound particles that act as a vehicle to transfer their contents, such as proteins, RNAs, and miRNAs, to the target cells, making them undergo several changes. Depending on the origin and the target cell, MVs may cause cell survival or apoptosis. This study investigated the effects of MVs released from the leukemic K562 cell line on the human bone marrow mesenchymal stem cells (hBM-MSCs) to evaluate changes in the survival or apoptosis of the cells in an in vitro system. Materials and Methods In this experimental study, we added the isolated MVs from the K562 cell line to hBM-MSCs, and after three and then seven days, subsequently cell count, cell viability, transmission electron microscopy, tracing MVs by carboxyfluorescein diacetate, succinimidyl ester (CFSE) solution, flow cytometry analysis for Annexin-V/PI staining and qPCR for the evaluation of BCL-2, KI67, and BAX expression were carried out. On the 10th day of the culture, hBM-MSCs were examined by Oil red O and Alizarin Red staining to evaluate their differentiation into adipocytes and osteoblasts. Results There was a significant decrease in cell viability and KI67 and BCL-2 expression; however, BAX was significantly upregulated in the hBM-MSCs compared to control groups. Annexin-V/PI staining results also showed the apoptotic effects of K562-MVs on hBM-MSCs. Moreover, the differentiation of hBM-MSCs into adipocytes and osteoblasts was not observed. Conclusion MVs from the leukemic cell line could affect the viability of normal hBM-MSCs and induce cell apoptosis.
Collapse
Affiliation(s)
- Neda Rassaei
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University,Tehran, Iran
| | - Mahnoosh Abbaszade Dibavar
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University,Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Hossein Mohammadi
- HSCT Research Center, Laboratory Hematology and Blood Banking Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding author: Mohammad Hossein Mohammadi. HSCT Research Center, Laboratory Hematology and Blood Banking Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Tel: +98-21-22718531;
| | - Mehdi Allahbakhshian Farsani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Shahsavan
- HSCT Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Ghazimoradi MH, Karimpour-Fard N, Babashah S. The Promising Role of Non-Coding RNAs as Biomarkers and Therapeutic Targets for Leukemia. Genes (Basel) 2023; 14:131. [PMID: 36672872 PMCID: PMC9859176 DOI: 10.3390/genes14010131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Early-stage leukemia identification is crucial for effective disease management and leads to an improvement in the survival of leukemia patients. Approaches based on cutting-edge biomarkers with excellent accuracy in body liquids provide patients with the possibility of early diagnosis with high sensitivity and specificity. Non-coding RNAs have recently received a great deal of interest as possible biomarkers in leukemia due to their participation in crucial oncogenic processes such as proliferation, differentiation, invasion, apoptosis, and their availability in body fluids. Recent studies have revealed a strong correlation between leukemia and the deregulated non-coding RNAs. On this basis, these RNAs are also great therapeutic targets. Based on these advantages, we tried to review the role of non-coding RNAs in leukemia. Here, the significance of several non-coding RNA types in leukemia is highlighted, and their potential roles as diagnostic, prognostic, and therapeutic targets are covered.
Collapse
Affiliation(s)
- Mohammad H. Ghazimoradi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| | - Naeim Karimpour-Fard
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| |
Collapse
|
13
|
Identification of key microRNAs as predictive biomarkers of Nilotinib response in chronic myeloid leukemia: a sub-analysis of the ENESTxtnd clinical trial. Leukemia 2022; 36:2443-2452. [PMID: 35999259 DOI: 10.1038/s41375-022-01680-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 11/08/2022]
Abstract
Despite the effectiveness of tyrosine kinase inhibitors (TKIs) against chronic myeloid leukemia (CML), they are not usually curative as some patients develop drug-resistance or are at risk of disease relapse when treatment is discontinued. Studies have demonstrated that primitive CML cells display unique miRNA profiles in response to TKI treatment. However, the utility of miRNAs in predicting treatment response is not yet conclusive. Here, we analyzed differentially expressed miRNAs in CD34+ CML cells pre- and post-nilotinib (NL) therapy from 58 patients enrolled in the Canadian sub-analysis of the ENESTxtnd phase IIIb clinical trial which correlated with sensitivity of CD34+ cells to NL treatment in in vitro colony-forming cell (CFC) assays. We performed Cox Proportional Hazard (CoxPH) analysis and applied machine learning algorithms to generate multivariate miRNA panels which can predict NL response at treatment-naïve or post-treatment time points. We demonstrated that a combination of miR-145 and miR-708 are effective predictors of NL response in treatment-naïve patients whereas miR-150 and miR-185 were significant classifiers at 1-month and 3-month post-NL therapy. Interestingly, incorporation of NL-CFC output in these panels enhanced predictive performance. Thus, this novel predictive model may be developed into a prognostic tool for use in the clinic.
Collapse
|
14
|
Chen CW, Huang NK, Lee YL, Fan CK, Chen YC, Liu CW, Huang HM. Activin A downregulates the CD69-MT2A axis via p38MAPK to induce erythroid differentiation that sensitizes BCR-ABL-positive cells to imatinib. Exp Cell Res 2022; 417:113219. [DOI: 10.1016/j.yexcr.2022.113219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/25/2022] [Accepted: 05/19/2022] [Indexed: 11/04/2022]
|
15
|
Elias MH, Syed Mohamad SF, Abdul Hamid N. A Systematic Review of Candidate miRNAs, Its Targeted Genes and Pathways in Chronic Myeloid Leukemia-An Integrated Bioinformatical Analysis. Front Oncol 2022; 12:848199. [PMID: 35330714 PMCID: PMC8940286 DOI: 10.3389/fonc.2022.848199] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic myeloid leukaemia is blood cancer due to a reciprocal translocation, resulting in a BCR-ABL1 oncogene. Although tyrosine kinase inhibitors have been successfully used to treat CML, there are still cases of resistance. The resistance occurred mainly due to the mutation in the tyrosine kinase domain of the BCR-ABL1 gene. However, there are still many cases with unknown causes of resistance as the etiopathology of CML are not fully understood. Thus, it is crucial to figure out the complete pathogenesis of CML, and miRNA can be one of the essential pathogeneses. The objective of this study was to systematically review the literature on miRNAs that were differentially expressed in CML cases. Their target genes and downstream genes were also explored. An electronic search was performed via PubMed, Scopus, EBSCOhost MEDLINE, and Science Direct. The following MeSH (Medical Subject Heading) terms were used: chronic myeloid leukaemia, genes and microRNAs in the title or abstract. From 806 studies retrieved from the search, only clinical studies with in-vitro experimental evidence on the target genes of the studied miRNAs in CML cells were included. Two independent reviewers independently scrutinised the titles and abstracts before examining the eligibility of studies that met the inclusion criteria. Study design, sample size, sampling type, and the molecular method used were identified for each study. The pooled miRNAs were analysed using DIANA tools, and target genes were analysed with DAVID, STRING and Cytoscape MCODE. Fourteen original research articles on miRNAs in CML were included, 26 validated downstream genes and 187 predicted target genes were analysed and clustered into 7 clusters. Through GO analysis, miRNAs’ target genes were localised throughout the cells, including the extracellular region, cytosol, and nucleus. Those genes are involved in various pathways that regulate genomic instability, proliferation, apoptosis, cell cycle, differentiation, and migration of CML cells.
Collapse
Affiliation(s)
- Marjanu Hikmah Elias
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Malaysia
| | - Syarifah Faezah Syed Mohamad
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Malaysia.,Faculty of Applied Sciences, Universiti Teknologi MARA Cawangan Pahang, Jengka, Malaysia
| | - Nazefah Abdul Hamid
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Malaysia
| |
Collapse
|
16
|
The role of microRNAs in the development, progression and drug resistance of chronic myeloid leukemia and their potential clinical significance. Life Sci 2022; 296:120437. [DOI: 10.1016/j.lfs.2022.120437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 12/26/2022]
|
17
|
Small Non-Coding RNAs in Leukemia. Cancers (Basel) 2022; 14:cancers14030509. [PMID: 35158777 PMCID: PMC8833386 DOI: 10.3390/cancers14030509] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
In 2020, more than 60,500 people were diagnosed with leukemia in the USA, and more than 23,000 died. The incidence of leukemia is still rising, and drug resistance development is a serious concern for patients' wellbeing and survival. In the past two decades, small non-coding RNAs have been studied to evaluate their functions and possible role in cancer pathogenesis. Small non-coding RNAs are short RNA molecules involved in several cellular processes by regulating the expression of genes. An increasing body of evidence collected by many independent studies shows that the expression of these molecules is tissue specific, and that their dysregulation alters the expression of genes involved in tumor development, progression and drug response. Indeed, small non-coding RNAs play a pivotal role in the onset, staging, relapse and drug response of hematological malignancies and cancers in general. These findings strongly suggest that small non-coding RNAs could function as biomarkers and possible targets for therapy. Thus, in this review, we summarize the regulatory mechanisms of small non-coding RNA expression in different types of leukemia and assess their potential clinical implications.
Collapse
|
18
|
Hu D, Shao W, Liu L, Wang Y, Yuan S, Liu Z, Liu J, Zhang J. Intricate crosstalk between MYB and noncoding RNAs in cancer. Cancer Cell Int 2021; 21:653. [PMID: 34876130 PMCID: PMC8650324 DOI: 10.1186/s12935-021-02362-4] [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: 09/16/2021] [Accepted: 11/24/2021] [Indexed: 11/10/2022] Open
Abstract
MYB is often overexpressed in malignant tumors and plays a carcinogenic role in the initiation and development of cancer. Deletion of the MYB regulatory C-terminal domain may be a driving mutation leading to tumorigenesis, therefore, different tumor mechanisms produce similar MYB proteins. As MYB is a transcription factor, priority has been given to identifying the genes that it regulates. All previous attention has been focused on protein-coding genes. However, an increasing number of studies have suggested that MYB can affect the complexity of cancer progression by regulating tumor-associated noncoding RNAs (ncRNAs), such as microRNAs, long-non-coding RNAs and circular RNAs. ncRNAs can regulate the expression of numerous downstream genes at the transcription, RNA processing and translation levels, thereby having various biological functions. Additionally, ncRNAs play important roles in regulating MYB expression. This review focuses on the intricate crosstalk between oncogenic MYB and ncRNAs, which play a pivotal role in tumorigenesis, including proliferation, apoptosis, angiogenesis, metastasis, senescence and drug resistance. In addition, we discuss therapeutic strategies for crosstalk between MYB and ncRNAs to prevent the occurrence and development of cancer.
Collapse
Affiliation(s)
- Dingyu Hu
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenjun Shao
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Li Liu
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yanyan Wang
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shunling Yuan
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaoping Liu
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Ji Zhang
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China. .,Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, Guangdong, China.
| |
Collapse
|
19
|
Curik N, Polivkova V, Burda P, Koblihova J, Laznicka A, Kalina T, Kanderova V, Brezinova J, Ransdorfova S, Karasova D, Rejlova K, Bakardjieva M, Kuzilkova D, Kundrat D, Linhartova J, Klamova H, Salek C, Klener P, Hrusak O, Machova Polakova K. Somatic Mutations in Oncogenes Are in Chronic Myeloid Leukemia Acquired De Novo via Deregulated Base-Excision Repair and Alternative Non-Homologous End Joining. Front Oncol 2021; 11:744373. [PMID: 34616685 PMCID: PMC8488388 DOI: 10.3389/fonc.2021.744373] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022] Open
Abstract
Somatic mutations are a common molecular mechanism through which chronic myeloid leukemia (CML) cells acquire resistance to tyrosine kinase inhibitors (TKIs) therapy. While most of the mutations in the kinase domain of BCR-ABL1 can be successfully managed, the recurrent somatic mutations in other genes may be therapeutically challenging. Despite the major clinical relevance of mutation-associated resistance in CML, the mechanisms underlying mutation acquisition in TKI-treated leukemic cells are not well understood. This work demonstrated de novo acquisition of mutations on isolated single-cell sorted CML clones growing in the presence of imatinib. The acquisition of mutations was associated with the significantly increased expression of the LIG1 and PARP1 genes involved in the error-prone alternative nonhomologous end-joining pathway, leading to genomic instability, and increased expression of the UNG, FEN and POLD3 genes involved in the base-excision repair (long patch) pathway, allowing point mutagenesis. This work showed in vitro and in vivo that de novo acquisition of resistance-associated mutations in oncogenes is the prevalent method of somatic mutation development in CML under TKIs treatment.
Collapse
Affiliation(s)
- Nikola Curik
- Institute of Hematology and Blood Transfusion, Prague, Czechia.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | | | - Pavel Burda
- Institute of Hematology and Blood Transfusion, Prague, Czechia.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Jitka Koblihova
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Adam Laznicka
- Institute of Hematology and Blood Transfusion, Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Tomas Kalina
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Veronika Kanderova
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Jana Brezinova
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | | | | | - Katerina Rejlova
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Marina Bakardjieva
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Daniela Kuzilkova
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - David Kundrat
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Jana Linhartova
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Hana Klamova
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Cyril Salek
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czechia.,First Department of Internal Medicine-Department of Hematology, Charles University General Hospital in Prague, Prague, Czechia
| | - Ondrej Hrusak
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Katerina Machova Polakova
- Institute of Hematology and Blood Transfusion, Prague, Czechia.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
20
|
Kalushkova A, Nylund P, Párraga AA, Lennartsson A, Jernberg-Wiklund H. One Omics Approach Does Not Rule Them All: The Metabolome and the Epigenome Join Forces in Haematological Malignancies. EPIGENOMES 2021; 5:epigenomes5040022. [PMID: 34968247 PMCID: PMC8715477 DOI: 10.3390/epigenomes5040022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 02/01/2023] Open
Abstract
Aberrant DNA methylation, dysregulation of chromatin-modifying enzymes, and microRNAs (miRNAs) play a crucial role in haematological malignancies. These epimutations, with an impact on chromatin accessibility and transcriptional output, are often associated with genomic instability and the emergence of drug resistance, disease progression, and poor survival. In order to exert their functions, epigenetic enzymes utilize cellular metabolites as co-factors and are highly dependent on their availability. By affecting the expression of metabolic enzymes, epigenetic modifiers may aid the generation of metabolite signatures that could be utilized as targets and biomarkers in cancer. This interdependency remains often neglected and poorly represented in studies, despite well-established methods to study the cellular metabolome. This review critically summarizes the current knowledge in the field to provide an integral picture of the interplay between epigenomic alterations and the cellular metabolome in haematological malignancies. Our recent findings defining a distinct metabolic signature upon response to enhancer of zeste homolog 2 (EZH2) inhibition in multiple myeloma (MM) highlight how a shift of preferred metabolic pathways may potentiate novel treatments. The suggested link between the epigenome and the metabolome in haematopoietic tumours holds promise for the use of metabolic signatures as possible biomarkers of response to treatment.
Collapse
Affiliation(s)
- Antonia Kalushkova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
- Correspondence:
| | - Patrick Nylund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Alba Atienza Párraga
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, NEO, Karolinska Institutet, 14157 Huddinge, Sweden;
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| |
Collapse
|
21
|
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
|
22
|
Li Q, Huang Z, Peng Y, Wang X, Jiang G, Wang T, Mou K, Feng W. RanBP3 Regulates Proliferation, Apoptosis and Chemosensitivity of Chronic Myeloid Leukemia Cells via Mediating SMAD2/3 and ERK1/2 Nuclear Transport. Front Oncol 2021; 11:698410. [PMID: 34504783 PMCID: PMC8421687 DOI: 10.3389/fonc.2021.698410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Abnormal subcellular localization of proteins is an important cause of tumorigenesis and drug resistance. Chromosome region maintenance 1 (CRM1), the nuclear export regulator of most proteins, has been confirmed to be over-expressed in various malignancies and is regarded as an efficient target. But the potential role of the CRM1 cofactor RanBP3 (Ran Binding Protein 3) is left unrevealed in chronic myeloid leukemia (CML). Here, we first detected the level of RanBP3 in CML and found an elevated RanBP3 expression in CML compared with control. Then we used shRNA lentivirus to down-regulated RanBP3 in imatinib sensitive K562 cells and resistant K562/G01 cells and found RanBP3 silencing inhibited cell proliferation by up-regulating p21, induced caspase3-related cell apoptosis, and enhanced the drug sensitivity of IM in vitro. Notably, we observed that RanBP3 silencing restored imatinib sensitivity of K562 cells in NOD/SCID mice. Mechanistically, the nuclear aggregation of SMAD2/3 revealed that tumor suppressor axis (TGF-β)-SMAD2/3-p21 was the anti-proliferation program related to RanBP3 knockdown, and the decrease of cytoplasmic ERK1/2 caused by RanBP3 interference leaded to the down-regulation of anti-apoptosis protein p(Ser112)-BAD, which was the mechanism of increased cell apoptosis and enhanced chemosensitivity to imatinib in CML. In summary, this study revealed the expression and potential role of RanBP3 in CML, suggesting that targeting RanBP3 alone or combined with TKIs could improve the clinical response of CML.
Collapse
Affiliation(s)
- Qian Li
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Department of Clinical Hematology, Chongqing Medical University, Chongqing, China
| | - Zhenglan Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Department of Clinical Hematology, Chongqing Medical University, Chongqing, China
| | - Yuhang Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Department of Clinical Hematology, Chongqing Medical University, Chongqing, China
| | - Xin Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guoyun Jiang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Department of Clinical Hematology, Chongqing Medical University, Chongqing, China
| | - Teng Wang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Mou
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Department of Clinical Hematology, Chongqing Medical University, Chongqing, China
| | - Wenli Feng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Department of Clinical Hematology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
23
|
Chen N, Meng Z, Song J, Kong L, Zhang Y, Guo S, Zhang X, Lu X, Jiang L, Chen R, Jiao Z, Zhao L. miR-506 in patients with chronic myeloid leukemia and its effect on apoptosis of K562 cells. Am J Transl Res 2021; 13:9413-9420. [PMID: 34540060 PMCID: PMC8430173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/07/2020] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To explore the expression of miR-506 in chronic myeloid leukemia (CML) and its influence on the biological function of CML cells. METHODS Altogether 84 CML patients from February 2012 to September 2014 were obtained as the observation group (OG), and 71 healthy people were taken as the control group (CG). miR-506 was tested using RT-qPCR, and the 5-year survival of patients with high and low expression of miR-506 was compared with the median value of miR-506 as the limit. ROC curve was applied to detect the value of miR-506 in diagnosing CML and predicting the 5-year survival of patients, and K562 cell line was transfected with miR-506 inhibitor and miR-506 mimic for observing its effects on the cell proliferation and apoptosis. RESULTS miR-506 in CML patients was evidently lower than that in healthy people, the AUC of diagnosis of miR-506 was 0.883, the total survival of patients with low miR-506 was evidently lower than those with high miR-506, and the AUC of predicted survival of patients was 0.778. The proliferation of cells transfected with miR-506 inhibitor was promoted, the apoptosis and the survival rate reduced. CONCLUSION miR-506 is evidently reduced in CML, and may be applied as a diagnostic and predictive treatment for CML and 5-year related survival; it can also can hinder the viability of K562 cells and promote apoptosis.
Collapse
Affiliation(s)
- Nafei Chen
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Zhen Meng
- Department of Hematology, Hudson International Peace Hospital, Heng Shui City People’s HospitalHengshui 053000, Hebei Province, China
| | - Jiaojie Song
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Lingfang Kong
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Yehua Zhang
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Suli Guo
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Xiaokun Zhang
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Xin Lu
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Licai Jiang
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Ran Chen
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Zongjiu Jiao
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| | - Liyun Zhao
- Department of Hematology, Xingtai People’s HospitalXingtai 054000, Hebei Province, China
| |
Collapse
|
24
|
Habib EM, Nosiar NA, Eid MA, Taha AM, Sherief DE, Hassan AE, Abdel Ghafar MT. MiR-150 Expression in Chronic Myeloid Leukemia: Relation to Imatinib Response. Lab Med 2021; 53:58-64. [PMID: 34350970 DOI: 10.1093/labmed/lmab040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To assess the circulating micro-RNA-150 (miR-150) expression in patients with chronic myeloid leukemia (CML) in relation to imatinib response. METHODS Sixty patients with CML and 20 age- and sex-matched control subjects were enrolled. Circulating miR-150 levels were assessed by quantitative real-time polymerase chain reaction on days 0, 14, and 90 of imatinib therapy for patients and once for control subjects. RESULTS The baseline miR-150 expression was significantly lower in patients with CML than in control subjects with subsequent elevation at 14 and 90 days after the start of imatinib treatment. Early treatment response (ETR) at 90 days was the main study outcome. The miR-150 expression had a significantly higher level in patients with CML with ETR. On multivariate analysis, miR-150 on day 14 was significantly related to ETR in patients with CML with predictive efficacy (area under the curve = 0.838, 72.9% sensitivity, and 84.2% specificity). CONCLUSION We found that miR-150 expression on day 14 of imatinib treatment is a useful early predictive candidate for imatinib response in patients with CML.
Collapse
Affiliation(s)
- Eman M Habib
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - Nahla A Nosiar
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - Manal A Eid
- Department of Clinical Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Atef M Taha
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia E Sherief
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - Asmaa E Hassan
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | | |
Collapse
|
25
|
Wang L, Li L, Chen R, Huang X, Ye X. Understanding and Monitoring Chronic Myeloid Leukemia Blast Crisis: How to Better Manage Patients. Cancer Manag Res 2021; 13:4987-5000. [PMID: 34188552 PMCID: PMC8236273 DOI: 10.2147/cmar.s314343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/13/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic myeloid leukemia (CML) is triggered primarily by the t(9;22) (q34.13; q11.23) translocation. This reciprocal chromosomal translocation leads to the formation of the BCR-ABL fusion gene. Patients in the chronic phase (CP) experience a good curative effect with tyrosine kinase inhibitors. However, cases are treatment refractory, with a dismal prognosis, when the disease has progressed to the accelerated phase (AP) or blast phase (BP). Until now, few reports have provided a comprehensive description of the mechanisms involved at different molecular levels. Indeed, the underlying pathogenesis of CML evolution comprises genetic aberrations, chromosomal translocations (except for the Philadelphia chromosome), telomere biology, and epigenetic anomalies. Herein, we provide knowledge of the biology responsible for blast transformation of CML at several levels, such as genetics, telomere biology, and epigenetic anomalies. Because of the limited treatment options available and poor outcomes, only the therapeutic response is monitored regularly, which involves BCR-ABL transcript level assessment and immunologic surveillance, with the optimal treatment strategy for patients in CP adapted to evaluate disease recurrence or progression. Overall, selecting optimal treatment endpoints to predict survival and successful TFR improves the quality of life of patients. Thus, identifying risk factors and developing risk-adapted therapeutic options may contribute to a better outcome for advanced-phase patients.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Program in Clinical Medicine, School of Medicine of Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Li Li
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Rongrong Chen
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Program in Clinical Medicine, School of Medicine of Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xianbo Huang
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiujin Ye
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
26
|
Peixoto da Silva S, Caires HR, Bergantim R, Guimarães JE, Vasconcelos MH. miRNAs mediated drug resistance in hematological malignancies. Semin Cancer Biol 2021; 83:283-302. [PMID: 33757848 DOI: 10.1016/j.semcancer.2021.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/11/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
Despite improvements in the therapeutic approaches for hematological malignancies in the last decades, refractory disease still occurs, and cancer drug resistance still remains a major hurdle in the clinical management of these cancer patients. The investigation of this problem has been extensive and different mechanism and molecules have been associated with drug resistance. MicroRNAs (miRNAs) have been described as having an important action in the emergence of cancer, including hematological tumors, and as being major players in their progression, aggressiveness and response to treatments. Moreover, miRNAs have been strongly associated with cancer drug resistance and with the modulation of the sensitivity of cancer cells to a wide array of anticancer drugs. Furthermore, this role has also been reported for miRNAs packaged into extracellular vesicles (EVs-miRNAs), which in turn have been described as essential for the horizontal transfer of drug resistance to sensitive cells. Several studies have been suggesting the use of miRNAs as biomarkers for drug response and clinical outcome prediction, as well as promising therapeutic tools in hematological diseases. Indeed, the combination of miRNA-based therapeutic tools with conventional drugs contributes to overcome drug resistance. This review addresses the role of miRNAs in the pathogenesis of hematological malignances, namely multiple myeloma, leukemias and lymphomas, highlighting their important action (either in their cell-free circulating form or within circulating EVs) in drug resistance and their potential clinical applications.
Collapse
Affiliation(s)
- Sara Peixoto da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal
| | - Hugo R Caires
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal
| | - Rui Bergantim
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal; Clinical Hematology, Hospital São João, 4200-319, Porto, Portugal; Clinical Hematology, FMUP - Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - José E Guimarães
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal; Clinical Hematology, FMUP - Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal; Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário, IUCSCESPU, 4585-116, Gandra, Paredes, Portugal
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
27
|
Regulation of MYB by distal enhancer elements in human myeloid leukemia. Cell Death Dis 2021; 12:223. [PMID: 33637692 PMCID: PMC7910426 DOI: 10.1038/s41419-021-03515-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/31/2022]
Abstract
MYB plays vital roles in regulating proliferation and differentiation of hematopoietic progenitor cells, dysregulation of MYB has been implicated in the pathogenesis of leukemia. Although the transcription of MYB has been well studied, its detailed underlying regulatory mechanisms still remain elusive. Here, we detected the long-range interaction between the upstream regions, −34k and −88k, and the MYB promoter in K562, U937, and HL-60 cells using circularized chromosome conformation capture (4C) assay, which declined when MYB was downregulated during chemical-induced differentiation. The enrichment of enhancer markers, H3K4me1 and H3K27ac, and enhancer activity at the −34k and −88k regions were confirmed by ChIP-qPCR and luciferase assay respectively. ChIP-qPCR showed the dynamic binding of GATA1, TAL1, and CCAAT/enhancer-binding protein (C/EBPβ) at −34k and −88k during differentiation of K562 cells. Epigenome editing by a CRISPR-Cas9-based method showed that H3K27ac at −34k enhanced TF binding and MYB expression, while DNA methylation inhibited MYB expression. Taken together, our data revealed that enhancer elements at −34k are required for MYB expression, TF binding, and epigenetic modification are closely involved in this process in human myeloid leukemia cells.
Collapse
|
28
|
LncRNA highly upregulated in liver cancer regulates imatinib resistance in chronic myeloid leukemia via the miR-150-5p/MCL1 axis. Anticancer Drugs 2021; 32:427-436. [PMID: 33587348 DOI: 10.1097/cad.0000000000001019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML) is a type of myeloproliferative neoplasm. Aberrant expression of long noncoding RNA highly upregulated in liver cancer (HULC) has been implicated in tumor progression, including CML. This study aimed to investigate the role of HULC in CML. The levels of HULC, miR-150-5p and myeloid cell leukemia 1 (MCL1) were examined by quantitative real-time PCR or western blot assay. Cell counting kit-8 assay was used to detect cell viability and half inhibition concentration. Cell apoptosis was monitored by flow cytometry and western blot. The interaction among HULC, miR-150-5p and MCL1 was validated by dual-luciferase reporter assay. The expression of phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT) and phosphorylation-AKT was evaluated using western blot assay. HULC and MCL1 were upregulated, whereas miR-150-5p was downregulated in bone marrow mononuclear cells of CML patients and CML cells. HULC overexpression increased imatinib resistance in K562 cells, and HULC depletion enhanced imatinib sensitivity in imatinib-resistant cells (K562-R). Mechanically, HULC was a sponge of miR-150-5p. HULC contributed to imatinib resistance through regulation of miR-150-5p. MCL1 bound to miR-150-5p and reversed the effect of HULC on imatinib resistance. HULC regulated the PI3K/AKT pathway via the miR-150-5p/MCL1 axis. These findings indicated that HULC enhanced imatinib resistance in CML by modulating the miR-150-5p/MCL1 axis, providing a promising biomarker for CML.
Collapse
|
29
|
Yung Y, Lee E, Chu HT, Yip PK, Gill H. Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22020659. [PMID: 33440869 PMCID: PMC7827471 DOI: 10.3390/ijms22020659] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 02/02/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in haematopoiesis. They are characterized by stem cell-derived clonal myeloproliferation. The key MPNs comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). CML is defined by the presence of the Philadelphia (Ph) chromosome and BCR-ABL1 fusion gene. Despite effective cytoreductive agents and targeted therapy, complete CML/MPN stem cell eradication is rarely achieved. In this review article, we discuss the novel agents and combination therapy that can potentially abnormal hematopoietic stem cells in CML and MPNs and the CML/MPN stem cell-sustaining bone marrow microenvironment.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Autophagy
- Biomarkers, Tumor
- Cell Survival/drug effects
- Cell Transformation, Neoplastic/genetics
- Combined Modality Therapy
- Disease Susceptibility
- Genetic Predisposition to Disease
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Molecular Targeted Therapy
- Myeloproliferative Disorders/etiology
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/therapy
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Philadelphia Chromosome
- Signal Transduction/drug effects
- Stem Cell Niche
- Tumor Microenvironment
Collapse
Affiliation(s)
| | | | | | | | - Harinder Gill
- Correspondence: ; Tel.: +852-2255-4542; Fax: +852-2816-2863
| |
Collapse
|
30
|
Minciacchi VR, Kumar R, Krause DS. Chronic Myeloid Leukemia: A Model Disease of the Past, Present and Future. Cells 2021; 10:cells10010117. [PMID: 33435150 PMCID: PMC7827482 DOI: 10.3390/cells10010117] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic myeloid leukemia (CML) has been a "model disease" with a long history. Beginning with the first discovery of leukemia and the description of the Philadelphia Chromosome and ending with the current goal of achieving treatment-free remission after targeted therapies, we describe here the journey of CML, focusing on molecular pathways relating to signaling, metabolism and the bone marrow microenvironment. We highlight current strategies for combination therapies aimed at eradicating the CML stem cell; hopefully the final destination of this long voyage.
Collapse
MESH Headings
- Epigenesis, Genetic
- History, 20th Century
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/history
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Models, Biological
- Molecular Targeted Therapy
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Valentina R. Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
| | - Daniela S. Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
- German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt, Germany
- Faculty of Medicine, Medical Clinic II, Johann Wolfgang Goethe University, 60596 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-63395-500; Fax: +49-69-63395-519
| |
Collapse
|
31
|
Embryonic Program Activated during Blast Crisis of Chronic Myelogenous Leukemia (CML) Implicates a TCF7L2 and MYC Cooperative Chromatin Binding. Int J Mol Sci 2020; 21:ijms21114057. [PMID: 32517078 PMCID: PMC7312032 DOI: 10.3390/ijms21114057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/25/2022] Open
Abstract
Chronic myeloid leukemia (CML) is characterized by an inherent genetic instability, which contributes to the progression of the disease towards an accelerated phase (AP) and blast crisis (BC). Several cytogenetic and genomic alterations have been reported in the progression towards BC, but the precise molecular mechanisms of this event are undetermined. Transcription Factor 7 like 2 (TFC7L2) is a member of the TCF family of proteins that are known to activate WNT target genes such as Cyclin D1. TCF7L2 has been shown to be overexpressed in acute myeloid leukemia (AML) and represents a druggable target. We report here that TCF7L2 transcription factor expression was found to be correlated to blast cell numbers during the progression of the disease. In these cells, TCF7L2 CHIP-sequencing highlighted distal cis active enhancer, such as elements in SMAD3, ATF5, and PRMT1 genomic regions and a proximal active transcriptional program of 144 genes. The analysis of CHIP-sequencing of MYC revealed a significant overlapping of TCF7L2 epigenetic program with MYC. The β-catenin activator lithium chloride and the MYC-MAX dimerization inhibitor 10058-F4 significantly modified the expression of three epigenetic targets in the BC cell line K562. These results suggest for the first time the cooperative role of TCF7L2 and MYC during CML-BC and they strengthen previous data showing a possible involvement of embryonic genes in this process.
Collapse
|
32
|
Yu Y, Kou D, Liu B, Huang Y, Li S, Qi Y, Guo Y, Huang T, Qi X, Jia L. LncRNA MEG3 contributes to drug resistance in acute myeloid leukemia by positively regulating ALG9 through sponging miR-155. Int J Lab Hematol 2020; 42:464-472. [PMID: 32359033 DOI: 10.1111/ijlh.13225] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/07/2020] [Accepted: 04/11/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The development of drug resistance is the main obstacle for successful treatment in acute myeloid leukemia (AML). Noncoding RNAs have been implicated in biological function in AML drug resistance. Aberrant protein glycosylation is associated with AML progression. The aim of the study was to explore the potential regulatory mechanism of lncRNA MEG3/miR-155/ALG9 axis in drug resistance of AML. METHODS QRT-PCR and Western blot were used for comparison analyses of ALG9, MEG3, and miR-155 levels. CCK-8 and colony formation assays were determined for drug sensitivity and proliferative capability of AML cells. Luciferase reporter assay was used to confirm the targets of miR-155. RESULTS The mannosyltransferase ALG9 and MEG3 was downregulated in peripheral blood mononuclear cells (PBMCs) of M5/multidrug resistance (MDR) AML patients and adriamycin (ADR)-resistant AML cell lines, which determined a positive correlation in AML patients. Low expression of ALG9 and MEG3 predicted poor prognosis of AML patients. The altered level of ALG9 was found corresponding to the drug-resistant phenotype and sphere formation of AML cells. MiR-155 was overexpressed in M5/MDR patients and ADR-resistant AML cells, as well as inversely correlated to ALG9 expression. MEG3 was a direct target of miR-155 and could sponge miR-155 in AML cells. MEG3 interacted with miR-155 to regulate ALG9 expression, which reversed the effects of ALG9 regulation on proliferation and drug resistance in AML cells. CONCLUSION MEG3 sponged miR-155 by competing endogenous RNA (ceRNA) mechanism, which further modulated ALG9 expression and AML procession, providing a novel therapeutic target for AML chemoresistance.
Collapse
Affiliation(s)
- Yanan Yu
- College of Laboratory Medicine, Dalian Medical University, Dalian, China.,Department of Emergency, Affiliated Dalian Friend-ship Hospital of Dalian Medical University, Dalian, China
| | - Daqing Kou
- Department of Clinical Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bing Liu
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Yiran Huang
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Shuangda Li
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Yu Qi
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Yanru Guo
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Tong Huang
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Xia Qi
- College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, 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
|
Hu T, Chong Y, Cai B, Liu Y, Lu S, Cowell JK. DNA methyltransferase 1-mediated CpG methylation of the miR-150-5p promoter contributes to fibroblast growth factor receptor 1-driven leukemogenesis. J Biol Chem 2019; 294:18122-18130. [PMID: 31628193 DOI: 10.1074/jbc.ra119.010144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/09/2019] [Indexed: 01/10/2023] Open
Abstract
MicroRNA-150-5p (miR-150-5p) plays a complex role in normal early hematopoietic development and is also implicated in the development of various different leukemias. We have reported previously that, in myeloid and lymphoid malignancies associated with dysregulated fibroblast growth factor receptor 1 (FGFR1) activities, miR-150-5p is down-regulated compared with healthy cells. Here, using murine cells, we found that this down-regulation is accompanied by CpG methylation of the miR-150-5p promoter region. Of note, analysis of human acute lymphoblastic leukemia (ALL) cohorts also revealed an inverse relationship between miR-150-5p expression and disease progression. We also found that the DNA methyltransferase 1 (DNMT1) enzyme is highly up-regulated in FGFR1-driven leukemias and lymphomas and that FGFR1 inhibition reduces DNMT1 expression. DNMT1 knockdown in stem cell leukemia/lymphoma (SCLL) cells increased miR-150-5p levels and reduced levels of the MYB proto-oncogene transcription factor, a key regulator of leukemogenesis. FGFR1 directly activates the MYC proto-oncogene basic helix-loop-helix transcription factor, which, as we show here, binds and activates the DNMT1 promoter. MYC knockdown decreased DNMT1 expression, which, in turn, increased miR-150-5p expression. One of the known targets of miR-150-5p is MYB, and treatment of leukemic cells with the MYB inhibitor mebendazole dose-dependently increased apoptosis and reduced cell viability. Moreover, mebendazole treatment of murine xenografts models of FGFR1-driven leukemias enhanced survival. These findings provide evidence that MYC activates MYB by up-regulating DNMT1, which silences miR-150-5p and promotes SCLL progression. We propose that inclusion of mebendazole in a combination therapy with FGFR1 inhibitors may be a valuable option to manage SCLL.
Collapse
Affiliation(s)
- Tianxiang Hu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912
| | - Yating Chong
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912
| | - Baohuan Cai
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912; Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun Liu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912; Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sumin Lu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912
| | - John K Cowell
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912.
| |
Collapse
|