1
|
Mehrtabar E, Khalaji A, Pandeh M, Farhoudian A, Shafiee N, Shafiee A, Ojaghlou F, Mahdavi P, Soleymani-Goloujeh M. Impact of microRNA variants on PI3K/AKT signaling in triple-negative breast cancer: comprehensive review. Med Oncol 2024; 41:222. [PMID: 39120634 DOI: 10.1007/s12032-024-02469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Breast cancer (BC) is a significant cause of cancer-related mortality, and triple-negative breast cancer (TNBC) is a particularly aggressive subtype associated with high mortality rates, especially among younger females. TNBC poses a considerable clinical challenge due to its aggressive tumor behavior and limited therapeutic options. Aberrations within the PI3K/AKT pathway are prevalent in TNBC and correlate with increased therapeutic intervention resistance and poor outcomes. MicroRNAs (miRs) have emerged as crucial PI3K/AKT pathway regulators influencing various cellular processes involved in TNBC pathogenesis. The levels of miRs, including miR-193, miR-4649-5p, and miR-449a, undergo notable changes in TNBC tumor tissues, emphasizing their significance in cancer biology. This review explored the intricate interplay between miR variants and PI3K/AKT signaling in TNBC. The review focused on the molecular mechanisms underlying miR-mediated dysregulation of this pathway and highlighted specific miRs and their targets. In addition, we explore the clinical implications of miR dysregulation in TNBC, particularly its correlation with TNBC prognosis and therapeutic resistance. Elucidating the roles of miRs in modulating the PI3K/AKT signaling pathway will enhance our understanding of TNBC biology and unveil potential therapeutic targets. This comprehensive review aims to discuss current knowledge and open promising avenues for future research, ultimately facilitating the development of precise and effective treatments for patients with TNBC.
Collapse
Affiliation(s)
- Ehsan Mehrtabar
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Pandeh
- School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Aram Farhoudian
- School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nadia Shafiee
- Children's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefe Shafiee
- Board-Certified Cardiologist, Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ojaghlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parinaz Mahdavi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Soleymani-Goloujeh
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
2
|
Zhang S, Zhang X, Yang H, Liang T, Bai X. Hurdle or thruster: Glucose metabolism of T cells in anti-tumour immunity. Biochim Biophys Acta Rev Cancer 2024; 1879:189022. [PMID: 37993001 DOI: 10.1016/j.bbcan.2023.189022] [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: 07/14/2023] [Revised: 09/11/2023] [Accepted: 10/08/2023] [Indexed: 11/24/2023]
Abstract
Glucose metabolism is essential for the activation, differentiation and function of T cells and proper glucose metabolism is required to maintain effective T cell immunity. Dysregulation of glucose metabolism is a hallmark of cancer, and the tumour microenvironment (TME2) can create metabolic barriers in T cells that inhibit their anti-tumour immune function. Targeting glucose metabolism is a promising approach to improve the capacity of T cells in the TME. The efficacy of common immunotherapies, such as immune checkpoint inhibitors (ICIs3) and adoptive cell transfer (ACT4), can be limited by T-cell function, and the treatment itself can affect T-cell metabolism. Therefore, understanding the relationship between immunotherapy and T cell glucose metabolism helps to achieve more effective anti-tumour therapy. In this review, we provide an overview of T cell glucose metabolism and how T cell metabolic reprogramming in the TME regulates anti-tumour responses, briefly describe the metabolic patterns of T cells during ICI and ACT therapies, which suggest possible synergistic strategies.
Collapse
Affiliation(s)
- Sirui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Hanshen Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
3
|
Wiese W, Barczuk J, Racinska O, Siwecka N, Rozpedek-Kaminska W, Slupianek A, Sierpinski R, Majsterek I. PI3K/Akt/mTOR Signaling Pathway in Blood Malignancies-New Therapeutic Possibilities. Cancers (Basel) 2023; 15:5297. [PMID: 37958470 PMCID: PMC10648005 DOI: 10.3390/cancers15215297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/19/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Blood malignancies remain a therapeutic challenge despite the development of numerous treatment strategies. The phosphatidylinositol-3 kinase (PI3K)/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway plays a central role in regulating many cellular functions, including cell cycle, proliferation, quiescence, and longevity. Therefore, dysregulation of this pathway is a characteristic feature of carcinogenesis. Increased activation of PI3K/Akt/mTOR signaling enhances proliferation, growth, and resistance to chemo- and immunotherapy in cancer cells. Overactivation of the pathway has been found in various types of cancer, including acute and chronic leukemia. Inhibitors of the PI3K/Akt/mTOR pathway have been used in leukemia treatment since 2014, and some of them have improved treatment outcomes in clinical trials. Recently, new inhibitors of PI3K/Akt/mTOR signaling have been developed and tested both in preclinical and clinical models. In this review, we outline the role of the PI3K/Akt/mTOR signaling pathway in blood malignancies' cells and gather information on the inhibitors of this pathway that might provide a novel therapeutic opportunity against leukemia.
Collapse
Affiliation(s)
- Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (J.B.); (O.R.); (N.S.); (W.R.-K.)
| | - Julia Barczuk
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (J.B.); (O.R.); (N.S.); (W.R.-K.)
| | - Olga Racinska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (J.B.); (O.R.); (N.S.); (W.R.-K.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (J.B.); (O.R.); (N.S.); (W.R.-K.)
| | - Wioletta Rozpedek-Kaminska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (J.B.); (O.R.); (N.S.); (W.R.-K.)
| | - Artur Slupianek
- Department of Pathology, Fox Chase Cancer Center, Temple University, Philadelphia, PA 19111, USA;
| | - Radoslaw Sierpinski
- Faculty of Medicine, Cardinal Stefan Wyszyński University, 01-938 Warsaw, Poland;
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (J.B.); (O.R.); (N.S.); (W.R.-K.)
| |
Collapse
|
4
|
Bioactive peptide inhibits acute myeloid leukemia cell proliferation by downregulating ALKBH5-mediated m 6A demethylation of EIF4EBP1 and MLST8 mRNA. Cell Oncol (Dordr) 2022; 45:355-365. [PMID: 35579750 PMCID: PMC9187541 DOI: 10.1007/s13402-022-00666-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/05/2022] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose N6-methyladenosine (m6A), the most prevalent mRNA modification, plays an essential role in tumorigenesis. Notably, increasing interest has been directed to bioactive peptides (BPs) with antitumor activities. Here, we set out to investigate the potential of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis on prevention and treatment of acute myeloid leukemia (AML). Methods The biological effects of BP on AML cells were detected by MTT and ApoLive-Glo™ multiplex assays. The role of BP in tumor growth was determined by a subcutaneous xenograft model. The ALKBH5/MLST8/EIF4EBP1 axis was identified as a potential BP target in AML via methylated RNA immunoprecipitation sequencing (MeRIP-seq) combined with RNA sequencing (RNA-seq). Western blot, RT-qPCR, MeRIP-qPCR, dual-luciferase reporter and RNA stability assays were performed to validate the function and mode of action of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis. The clinical relevance of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis in AML was confirmed by TCGA data analysis. Results We found that BP can inhibit AML cell proliferation and promote apoptosis in vitro, and repress AML tumor growth in vivo. Mechanistically, we found that BP downregulated ALKBH5 expression, which in turn repressed m6A demethylation of MLST8 and EIF4EBP1 mRNAs. Reduction of the m6A levels of MLST8 and EIF4EBP1 facilitated MLST8 and EIF4EBP1 mRNA decay, resulting in inhibition of AML cell proliferation. Furthermore, we found that the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis closely correlates with AML patient prognosis. Conclusions Our data indicate that BP can inhibit acute myeloid leukemia cell proliferation by downregulating ALKBH5-mediated m6A demethylation of EIF4EBP1 and MLST8 mRNAs, which may have potential to prevent and treat this disease. Supplementary Information The online version contains supplementary material available at 10.1007/s13402-022-00666-9.
Collapse
|
5
|
BCR-ABL1 Tyrosine Kinase Complex Signaling Transduction: Challenges to Overcome Resistance in Chronic Myeloid Leukemia. Pharmaceutics 2022; 14:pharmaceutics14010215. [PMID: 35057108 PMCID: PMC8780254 DOI: 10.3390/pharmaceutics14010215] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/27/2023] Open
Abstract
The constitutively active BCR-ABL1 tyrosine kinase, found in t(9;22)(q34;q11) chromosomal translocation-derived leukemia, initiates an extremely complex signaling transduction cascade that induces a strong state of resistance to chemotherapy. Targeted therapies based on tyrosine kinase inhibitors (TKIs), such as imatinib, dasatinib, nilotinib, bosutinib, and ponatinib, have revolutionized the treatment of BCR-ABL1-driven leukemia, particularly chronic myeloid leukemia (CML). However, TKIs do not cure CML patients, as some develop TKI resistance and the majority relapse upon withdrawal from treatment. Importantly, although BCR-ABL1 tyrosine kinase is necessary to initiate and establish the malignant phenotype of Ph-related leukemia, in the later advanced phase of the disease, BCR-ABL1-independent mechanisms are also in place. Here, we present an overview of the signaling pathways initiated by BCR-ABL1 and discuss the major challenges regarding immunologic/pharmacologic combined therapies.
Collapse
|
6
|
Mars JC, Ghram M, Culjkovic-Kraljacic B, Borden KLB. The Cap-Binding Complex CBC and the Eukaryotic Translation Factor eIF4E: Co-Conspirators in Cap-Dependent RNA Maturation and Translation. Cancers (Basel) 2021; 13:6185. [PMID: 34944805 PMCID: PMC8699206 DOI: 10.3390/cancers13246185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022] Open
Abstract
The translation of RNA into protein is a dynamic process which is heavily regulated during normal cell physiology and can be dysregulated in human malignancies. Its dysregulation can impact selected groups of RNAs, modifying protein levels independently of transcription. Integral to their suitability for translation, RNAs undergo a series of maturation steps including the addition of the m7G cap on the 5' end of RNAs, splicing, as well as cleavage and polyadenylation (CPA). Importantly, each of these steps can be coopted to modify the transcript signal. Factors that bind the m7G cap escort these RNAs through different steps of maturation and thus govern the physical nature of the final transcript product presented to the translation machinery. Here, we describe these steps and how the major m7G cap-binding factors in mammalian cells, the cap binding complex (CBC) and the eukaryotic translation initiation factor eIF4E, are positioned to chaperone transcripts through RNA maturation, nuclear export, and translation in a transcript-specific manner. To conceptualize a framework for the flow and integration of this genetic information, we discuss RNA maturation models and how these integrate with translation. Finally, we discuss how these processes can be coopted by cancer cells and means to target these in malignancy.
Collapse
Affiliation(s)
- Jean-Clement Mars
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Mehdi Ghram
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Biljana Culjkovic-Kraljacic
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Katherine L B Borden
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Pavillion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
7
|
Chen X, Chen X, Huang Y, Lin J, Wu Y, Chen Y. TCP1 increases drug resistance in acute myeloid leukemia by suppressing autophagy via activating AKT/mTOR signaling. Cell Death Dis 2021; 12:1058. [PMID: 34750375 PMCID: PMC8575913 DOI: 10.1038/s41419-021-04336-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/29/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022]
Abstract
T-complex protein 1 (TCP1) is one of the subunits of chaperonin-containing T complex (CCT), which is involved in protein folding, cell proliferation, apoptosis, cell cycle regulation, and drug resistance. Investigations have demonstrated that TCP1 is a factor being responsible for drug resistance in breast and ovarian cancer. However, the TCP1 role in acute myeloid leukemia (AML) remains elusive. In the present study, we discovered that the TCP1 expression was elevated in AML patients and high TCP1 expression was associated with low complete response rate along with poor overall survival. TCP1 showed higher expression in the adriamycin-resistant leukemia cell line HL60/A and K562/A, comparing to their respective parent cells HL60 and K562 cells. TCP1 inhibition suppressed drug resistance in HL60/A and K562/A cells, whereas TCP1 overexpression in HL60 cells incremented drug resistance, both in vitro and in vivo. Mechanistic investigations revealed that TCP1 inhibited autophagy and adriamycin-induced cell apoptosis, and TCP1-mediated autophagy inhibition conferred resistance to adriamycin-induced cell apoptosis. Furthermore, TCP1 interacted with AKT and mTOR to activate AKT/mTOR signaling, which negatively regulates apoptosis and autophagy. Pharmacological inhibition of AKT/mTOR signal particularly activated autophagy and resensitized TCP1-overexpressing HL60 cells to adriamycin. These findings identify a novel role of TCP1 regarding drug resistance in AML, which advise a new strategy for overcoming drug resistance in AML through targeting TCP1/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaofang Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.,Department of Infectious Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xianling Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yiping Huang
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jia Lin
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yong Wu
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| | - Yuanzhong Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
8
|
Wu F, Chen Z, Liu J, Hou Y. The Akt-mTOR network at the interface of hematopoietic stem cell homeostasis. Exp Hematol 2021; 103:15-23. [PMID: 34464661 DOI: 10.1016/j.exphem.2021.08.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem cells (HSCs) are immature blood cells that exhibit multilineage differentiation capacity. Homeostasis is critical for HSC potential and lifelong hematopoiesis, and HSC homeostasis is tightly governed by both intrinsic molecular networks and microenvironmental signals. The evolutionarily conserved serine/threonine protein kinase B (PKB, also referred to as Akt)-mammalian target of rapamycin (mTOR) pathway is universal to nearly all multicellular organisms and plays an integral role in most cellular processes. Emerging evidence has revealed a central role of the Akt-mTOR network in HSC homeostasis, because it responds to multiple intracellular and extracellular signals and regulates various downstream targets, eventually affecting several cellular processes, including the cell cycle, mitochondrial metabolism, and protein synthesis. Dysregulated Akt-mTOR signaling greatly affects HSC self-renewal, maintenance, differentiation, survival, autophagy, and aging, as well as transformation of HSCs to leukemia stem cells. Here, we review recent works and provide an advanced understanding of how the Akt-mTOR network regulates HSC homeostasis, thus offering insights into future clinical applications.
Collapse
Affiliation(s)
- Feng Wu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhe Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China.
| | - Yu Hou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
9
|
Zhang H, Liu M, Wang X, Ren Y, Kim YM, Wang X, Lu X, Pang H, Liu G, Gu Y, Sun M, Shi Y, Zhang C, Zhang Y, Zhang J, Li S, Zhang L. Genomic Copy Number Variants in CML Patients With the Philadelphia Chromosome (Ph+): An Update. Front Genet 2021; 12:697009. [PMID: 34447409 PMCID: PMC8383316 DOI: 10.3389/fgene.2021.697009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background Submicroscopic segmental imbalances detected by array-comparative genomic hybridization (array-CGH) were discovered to be common in chronic myeloid leukemia (CML) patients with t(9;22) as the sole chromosomal anomaly. To confirm the findings of the previous study and expand the investigation, additional CML patients with t(9;22) as the sole chromosomal anomaly were recruited and copy number variants (CNVs) were searched for. Methods Karyotyping tests were performed on 106 CML patients during January 2010-September 2019 in our Genetics Laboratory. Eighty-four (79.2%) patients had the Philadelphia (Ph) chromosome as the sole chromosomal anomaly. Only 49(58.3%) of these 84 patients had sufficient marrow or leukemia blood materials to additionally be included in the array-CGH analysis. Fluorescence in situ hybridization (FISH) was carried out to confirm the genes covered by the deleted or duplicated regions of the CNVs. Results 11(22.4%) out of the 49 patients were found to have one to three somatic segmental somatic segmental (CNVs), including fourteen deletions and three duplications. The common region associated with deletions was on 9q33.3-34.12. Identified in five (45.5%) of the 11 positive patients with segmental CNVs, the deletions ranged from 106 kb to 4.1 Mb in size. Two (18.2%) cases had a deletion in the ABL1-BCR fusion gene on der (9), while three (27.3%) cases had a deletion in the ASS1 gene. The remaining CNVs were randomly distributed on different autosomes. Conclusion Subtle genomic CNVs are relatively common in CML patients without cytogenetically visible additional chromosomal aberrations (ACAs). Long-term studies investigating the potential impact on patient prognosis and treatment outcome is underway.
Collapse
Affiliation(s)
- Heyang Zhang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China
| | - Meng Liu
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xiaoxue Wang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yuan Ren
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Young Mi Kim
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xianfu Wang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xianglan Lu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hui Pang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Guangming Liu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Yue Gu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Respiratory and Intensive Care Medicine, The First Hospital of Jilin University, Changchun, China
| | - Mingran Sun
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Hematology and Oncology, Anshan Hospital of First Hospital of China Medical University, Shenyang, Anshan, China
| | - Yunpeng Shi
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chuan Zhang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Gansu Province Medical Genetics Center, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou, China
| | - Yaowen Zhang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Neurology, The Second Hospital of Jilin University, Changchun, China
| | - Jianqin Zhang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Pediatric Respiratory, Dalian Children's Hospital, Dalian, China
| | - Shibo Li
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Lijun Zhang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Mayerhofer C, Niemeyer CM, Flotho C. Current Treatment of Juvenile Myelomonocytic Leukemia. J Clin Med 2021; 10:3084. [PMID: 34300250 PMCID: PMC8305558 DOI: 10.3390/jcm10143084] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare pediatric leukemia characterized by mutations in five canonical RAS pathway genes. The diagnosis is made by typical clinical and hematological findings associated with a compatible mutation. Although this is sufficient for clinical decision-making in most JMML cases, more in-depth analysis can include DNA methylation class and panel sequencing analysis for secondary mutations. NRAS-initiated JMML is heterogeneous and adequate management ranges from watchful waiting to allogeneic hematopoietic stem cell transplantation (HSCT). Upfront azacitidine in KRAS patients can achieve long-term remissions without HSCT; if HSCT is required, a less toxic preparative regimen is recommended. Germline CBL patients often experience spontaneous resolution of the leukemia or exhibit stable mixed chimerism after HSCT. JMML driven by PTPN11 or NF1 is often rapidly progressive, requires swift HSCT and may benefit from pretransplant therapy with azacitidine. Because graft-versus-leukemia alloimmunity is central to cure high risk patients, the immunosuppressive regimen should be discontinued early after HSCT.
Collapse
Affiliation(s)
- Christina Mayerhofer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
| | - Charlotte M. Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
| |
Collapse
|
11
|
Liu Z, Zheng W, Liu Y, Zhou B, Zhang Y, Wang F. Targeting HSPA8 inhibits proliferation via downregulating BCR-ABL and enhances chemosensitivity in imatinib-resistant chronic myeloid leukemia cells. Exp Cell Res 2021; 405:112708. [PMID: 34157313 DOI: 10.1016/j.yexcr.2021.112708] [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: 04/18/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022]
Abstract
The resistance to tyrosine kinase inhibitors is currently a major problem for chronic myeloid leukemia (CML) treatment and HSPA8 is highly expressed and a hallmark of poor prognosis in several human cancers. However, its role in imatinib-resistant CML (IR-CML) cells remains undetermined. Here, we determined HSPA8 was overexpressed in IR-CML cells and associated with imatinib resistance. HSPA8 ablation could downregulate BCR-ABL/STAT5 and BCR-ABL/AKT signaling pathways, dramatically induce proliferation inhibition, autophagy, G0/G1 phase cell cycle arrest but not apoptosis in IR-CML cells. Significantly, HSPA8 ablation enhanced the antitumor activity of imatinib via promoting apoptosis in vitro and vivo. These findings unraveled that HSPA8 ablation inhibits proliferation via downregulating BCR-ABL and enhances chemosensitivity of imatinib in IR-CML cells, which investigate the role and molecular mechanism of HSPA8 in IR-CML cells and suggest that HSPA8 may be a potential target for IR-CML treatment.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - Wenlong Zheng
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| | - Yuan Liu
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Binghe Zhou
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| | - Yuqing Zhang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| | - Fan Wang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| |
Collapse
|
12
|
Cortes J, Lang F. Third-line therapy for chronic myeloid leukemia: current status and future directions. J Hematol Oncol 2021; 14:44. [PMID: 33736651 PMCID: PMC7976694 DOI: 10.1186/s13045-021-01055-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Chronic myeloid leukemia (CML) is driven by the BCR-ABL1 fusion protein, formed by a translocation between chromosomes 9 and 22 that creates the Philadelphia chromosome. The BCR-ABL1 fusion protein is an optimal target for tyrosine kinase inhibitors (TKIs) that aim for the adenosine triphosphate (ATP) binding site of ABL1. While these drugs have greatly improved the prognosis for CML, many patients ultimately fail treatment, some requiring multiple lines of TKI therapy. Mutations can occur in the ATP binding site of ABL1, causing resistance by preventing the binding of many of these drugs and leaving patients with limited treatment options. The approved TKIs are also associated with adverse effects that may lead to treatment discontinuation in some patients. Efficacy decreases with each progressive line of therapy; data suggest little clinical benefit of treatment with a third-line (3L), second-generation tyrosine kinase inhibitor (2GTKI) after failure of a first-generation TKI and a 2GTKI. Novel treatment options are needed for the patient population that requires treatment in the 3L setting and beyond. This review highlights the need for clear guidelines and new therapies for patients requiring 3L treatment and beyond.
Collapse
Affiliation(s)
- Jorge Cortes
- Georgia Cancer Center at Augusta University, 1410 Laney Walker Rd., CN2222, Augusta, GA, 30912, USA.
| | - Fabian Lang
- Department of Medicine, Hematology and Oncology, Goethe University Hospital, Building 33, 3rd floor, Room 246, Theodor-Stern-Kai 7, 60590, Frankfurt a. Main, Germany
| |
Collapse
|
13
|
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
|
14
|
Constantinou C, Charalambous C, Kanakis D, Kolokotroni O, Constantinou AI. Update on the Anti-Cancer Potency of Tocotrienols and α-Tocopheryl Polyethylene Glycol 1000 Succinate on Leukemic Cell Lines. Nutr Cancer 2020; 73:1302-1308. [PMID: 32698633 DOI: 10.1080/01635581.2020.1797128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The natural isoforms of vitamin E γ-tocotrienol (γ-ΤΤ) and δ-tocotrienol (δ-ΤΤ) and the synthetic derivative α-tocopheryl polyethylene glycol 1000 succinate (TPGS) have promising anticancer potency in a variety of cancer cell lines and animal models of cancer. Ongoing clinical trials are investigating the anti-tumor effectiveness of TTs in combination with chemotherapeutic agents in patients suffering from breast, colon, non-small cell lung and ovarian cancers. Despite extensive research on different types of cancer, the anticancer potency of TTs and TPGS has not been thoroughly investigated in leukemias. Given the fact that certain types of leukemias have very low survival rates and that patients suffer significantly from the toxic side effects of chemotherapeutic drugs, there is a need to develop novel treatments with increased specificity against cancer cells and reduced toxicity to the patients. The aim of this review is to report current evidence on the anticancer potency of TTs and TPGS on leukemic cells lines and to discuss future studies that could be carried out to investigate the role of these agents in the management of leukemias.
Collapse
Affiliation(s)
- Constantina Constantinou
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Christiana Charalambous
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Dimitrios Kanakis
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Ourania Kolokotroni
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia, Cyprus
| | - Andreas I Constantinou
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| |
Collapse
|
15
|
mTOR and STAT3 Pathway Hyper-Activation is Associated with Elevated Interleukin-6 Levels in Patients with Shwachman-Diamond Syndrome: Further Evidence of Lymphoid Lineage Impairment. Cancers (Basel) 2020; 12:cancers12030597. [PMID: 32150944 PMCID: PMC7139896 DOI: 10.3390/cancers12030597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/29/2022] Open
Abstract
Shwachman–Diamond syndrome (SDS) is a rare inherited bone marrow failure syndrome, resulting in neutropenia and a risk of myeloid neoplasia. A mutation in a ribosome maturation factor accounts for almost all of the cases. Lymphoid involvement in SDS has not been well characterized. We recently reported that lymphocyte subpopulations are reduced in SDS patients. We have also shown that the mTOR-STAT3 pathway is hyper-activated in SDS myeloid cell populations. Here we show that mTOR-STAT3 signaling is markedly upregulated in the lymphoid compartment of SDS patients. Furthermore, our data reveal elevated IL-6 levels in cellular supernatants obtained from lymphoblasts, bone marrow mononuclear and mesenchymal stromal cells, and plasma samples obtained from a cohort of 10 patients. Of note, everolimus-mediated inhibition of mTOR signaling is associated with basal state of phosphorylated STAT3. Finally, inhibition of mTOR-STAT3 pathway activation leads to normalization of IL-6 expression in SDS cells. Altogether, our data strengthen the hypothesis that SDS affects both lymphoid and myeloid blood compartment and suggest everolimus as a potential therapeutic agent to reduce excessive mTOR-STAT3 activation in SDS.
Collapse
|
16
|
Özgür Yurttaş N, Eşkazan AE. Novel therapeutic approaches in chronic myeloid leukemia. Leuk Res 2020; 91:106337. [PMID: 32200189 DOI: 10.1016/j.leukres.2020.106337] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
The tyrosine kinase inhibitors (TKIs) have revolutionized the management of chronic myeloid leukemia (CML) and BCR-ABL1 inhibitors form the mainstay of CML treatment. Although patients with CML generally do well under TKI therapy, there is a subgroup of patients who are resistant and/or intolerant to TKIs. In these group of patients, there is the need of additional treatment strategies. In this review, we provide an update on the current knowledge of these novel treatment approaches that can be used alone and/or in combination with TKIs.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Clinical Trials as Topic
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Everolimus/therapeutic use
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/immunology
- Gene Expression
- Histone Deacetylase Inhibitors/therapeutic use
- Homoharringtonine/therapeutic use
- Humans
- Immunotherapy/methods
- Interferon-alpha/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy/methods
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Piperidines/therapeutic use
- Polyethylene Glycols/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/therapeutic use
- Pyridines/therapeutic use
- Quinolones/therapeutic use
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Nurgül Özgür Yurttaş
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ahmet Emre Eşkazan
- Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
17
|
Mechanisms of Cardiovascular Toxicity of BCR-ABL1 Tyrosine Kinase Inhibitors in Chronic Myelogenous Leukemia. Curr Hematol Malig Rep 2020; 15:20-30. [DOI: 10.1007/s11899-020-00560-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
Phospho-Profiling Linking Biology and Clinics in Pediatric Acute Myeloid Leukemia. Hemasphere 2019; 4:e312. [PMID: 32072137 PMCID: PMC7000467 DOI: 10.1097/hs9.0000000000000312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023] Open
Abstract
Supplemental Digital Content is available in the text Aberrant activation of key signaling-molecules is a hallmark of acute myeloid leukemia (AML) and may have prognostic and therapeutic implications. AML summarizes several disease entities with a variety of genetic subtypes. A comprehensive model spanning from signal activation patterns in major genetic subtypes of pediatric AML (pedAML) to outcome prediction and pre-clinical response to signaling inhibitors has not yet been provided. We established a high-throughput flow-cytometry based method to assess activation of hallmark phospho-proteins (phospho-flow) in 166 bone-marrow derived pedAML samples under basal and cytokine stimulated conditions. We correlated levels of activated phospho-proteins at diagnosis with relapse incidence in intermediate (IR) and high risk (HR) subtypes. In parallel, we screened a set of signaling inhibitors for their efficacy against primary AML blasts in a flow-cytometry based ex vivo cytotoxicity assay and validated the results in a murine xenograft model. Certain phospho-signal patterns differ between genetic subtypes of pedAML. Some are consistently seen through all AML subtypes such as pSTAT5. In IR/HR subtypes high levels of GM-CSF stimulated pSTAT5 and low levels of unstimulated pJNK correlated with increased relapse risk overall. Combination of GM-CSF/pSTAT5high and basal/pJNKlow separated three risk groups among IR/HR subtypes. Out of 10 tested signaling inhibitors, midostaurin most effectively affected AML blasts and simultaneously blocked phosphorylation of multiple proteins, including STAT5. In a mouse xenograft model of KMT2A-rearranged pedAML, midostaurin significantly prolonged disease latency. Our study demonstrates the applicability of phospho-flow for relapse-risk assessment in pedAML, whereas functional phenotype-driven ex vivo testing of signaling inhibitors may allow individualized therapy.
Collapse
|
19
|
Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacol Res 2019; 150:104511. [DOI: 10.1016/j.phrs.2019.104511] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/19/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022]
|
20
|
Gatz SA, Aladowicz E, Casanova M, Chisholm JC, Kearns PR, Fulda S, Geoerger B, Schäfer BW, Shipley JM. A Perspective on Polo-Like Kinase-1 Inhibition for the Treatment of Rhabdomyosarcomas. Front Oncol 2019; 9:1271. [PMID: 31824851 PMCID: PMC6882953 DOI: 10.3389/fonc.2019.01271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Rhabdomyosarcomas are the most common pediatric soft tissue sarcoma and are a major cause of death from cancer in young patients requiring new treatment options to improve outcomes. High-risk patients include those with metastatic or relapsed disease and tumors with PAX3-FOXO1 fusion genes that encode a potent transcription factor that drives tumourigenesis through transcriptional reprogramming. Polo-Like Kinase-1 (PLK1) is a serine/threonine kinase that phosphorylates a wide range of target substrates and alters their activity. PLK1 functions as a pleiotropic master regulator of mitosis and regulates DNA replication after stress. Taken together with high levels of expression that correlate with poor outcomes in many cancers, including rhabdomyosarcomas, it is an attractive therapeutic target. This is supported in rhabdomyosarcoma models by characterization of molecular and phenotypic effects of reducing and inhibiting PLK1, including changes to the PAX3-FOXO1 fusion protein. However, as tumor re-growth has been observed, combination strategies are required. Here we review preclinical evidence and consider biological rationale for PLK1 inhibition in combination with drugs that promote apoptosis, interfere with activity of PAX3-FOXO1 and are synergistic with microtubule-destabilizing drugs such as vincristine. The preclinical effects of low doses of the PLK1 inhibitor volasertib in combination with vincristine, which is widely used in rhabdomyosarcoma treatment, show particular promise in light of recent clinical data in the pediatric setting that support achievable volasertib doses predicted to be effective. Further development of novel therapeutic strategies including PLK1 inhibition may ultimately benefit young patients with rhabdomyosarcoma and other cancers.
Collapse
Affiliation(s)
- Susanne A. Gatz
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Ewa Aladowicz
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | | | - Julia C. Chisholm
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Pamela R. Kearns
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
| | - Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Paediatric and Adolescent Oncology, Université Paris-Saclay, Villejuif, France
| | - Beat W. Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Janet M. Shipley
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
21
|
Vaske OM, Bjork I, Salama SR, Beale H, Tayi Shah A, Sanders L, Pfeil J, Lam DL, Learned K, Durbin A, Kephart ET, Currie R, Newton Y, Swatloski T, McColl D, Vivian J, Zhu J, Lee AG, Leung SG, Spillinger A, Liu HY, Liang WS, Byron SA, Berens ME, Resnick AC, Lacayo N, Spunt SL, Rangaswami A, Huynh V, Torno L, Plant A, Kirov I, Zabokrtsky KB, Rassekh SR, Deyell RJ, Laskin J, Marra MA, Sender LS, Mueller S, Sweet-Cordero EA, Goldstein TC, Haussler D. Comparative Tumor RNA Sequencing Analysis for Difficult-to-Treat Pediatric and Young Adult Patients With Cancer. JAMA Netw Open 2019; 2:e1913968. [PMID: 31651965 PMCID: PMC6822083 DOI: 10.1001/jamanetworkopen.2019.13968] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPORTANCE Pediatric cancers are epigenetic diseases; therefore, considering tumor gene expression information is necessary for a complete understanding of the tumorigenic processes. OBJECTIVE To evaluate the feasibility and utility of incorporating comparative gene expression information into the precision medicine framework for difficult-to-treat pediatric and young adult patients with cancer. DESIGN, SETTING, AND PARTICIPANTS This cohort study was conducted as a consortium between the University of California, Santa Cruz (UCSC) Treehouse Childhood Cancer Initiative and clinical genomic trials. RNA sequencing (RNA-Seq) data were obtained from the following 4 clinical sites and analyzed at UCSC: British Columbia Children's Hospital (n = 31), Lucile Packard Children's Hospital at Stanford University (n = 80), CHOC Children's Hospital and Hyundai Cancer Institute (n = 46), and the Pacific Pediatric Neuro-Oncology Consortium (n = 24). The study dates were January 1, 2016, to March 22, 2017. EXPOSURES Participants underwent tumor RNA-Seq profiling as part of 4 separate clinical trials at partner hospitals. The UCSC either downloaded RNA-Seq data from a partner institution for analysis in the cloud or provided a Docker pipeline that performed the same analysis at a partner institution. The UCSC then compared each participant's tumor RNA-Seq profile with more than 11 000 uniformly analyzed tumor profiles from pediatric and young adult patients with cancer, downloaded from public data repositories. These comparisons were used to identify genes and pathways that are significantly overexpressed in each patient's tumor. Results of the UCSC analysis were presented to clinical partners. MAIN OUTCOMES AND MEASURES Feasibility of a third-party institution (UCSC Treehouse Childhood Cancer Initiative) to obtain tumor RNA-Seq data from patients, conduct comparative analysis, and present analysis results to clinicians; and proportion of patients for whom comparative tumor gene expression analysis provided useful clinical and biological information. RESULTS Among 144 samples from children and young adults (median age at diagnosis, 9 years; range, 0-26 years; 72 of 118 [61.0%] male [26 patients sex unknown]) with a relapsed, refractory, or rare cancer treated on precision medicine protocols, RNA-Seq-derived gene expression was potentially useful for 99 of 144 samples (68.8%) compared with DNA mutation information that was potentially useful for only 34 of 74 samples (45.9%). CONCLUSIONS AND RELEVANCE This study's findings suggest that tumor RNA-Seq comparisons may be feasible and highlight the potential clinical utility of incorporating such comparisons into the clinical genomic interpretation framework for difficult-to-treat pediatric and young adult patients with cancer. The study also highlights for the first time to date the potential clinical utility of harmonized publicly available genomic data sets.
Collapse
Affiliation(s)
- Olena M. Vaske
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Isabel Bjork
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Sofie R. Salama
- University of California, Santa Cruz Genomics Institute, Santa Cruz
- Howard Hughes Medical Institute, University of California, Santa Cruz
| | - Holly Beale
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Avanthi Tayi Shah
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Lauren Sanders
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Jacob Pfeil
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Du L. Lam
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Katrina Learned
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Ann Durbin
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Ellen T. Kephart
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Rob Currie
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Yulia Newton
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Teresa Swatloski
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Duncan McColl
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - John Vivian
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Jingchun Zhu
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Alex G. Lee
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Stanley G. Leung
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Aviv Spillinger
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Heng-Yi Liu
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Winnie S. Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | - Sara A. Byron
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | | | - Adam C. Resnick
- Center for Data Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Norman Lacayo
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Sheri L. Spunt
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Arun Rangaswami
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Van Huynh
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Lilibeth Torno
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Ashley Plant
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Ivan Kirov
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | | | - S. Rod Rassekh
- British Columbia Children’s Hospital Research Institute, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Rebecca J. Deyell
- British Columbia Children’s Hospital Research Institute, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | | | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard S. Sender
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Sabine Mueller
- Department of Neurology, University of California, San Francisco
- Department of Neurosurgery, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
| | | | - Theodore C. Goldstein
- University of California, Santa Cruz Genomics Institute, Santa Cruz
- Now with Anthem, Inc, Palo Alto, California
| | - David Haussler
- University of California, Santa Cruz Genomics Institute, Santa Cruz
- Howard Hughes Medical Institute, University of California, Santa Cruz
| |
Collapse
|
22
|
Castro I, Sampaio-Marques B, Ludovico P. Targeting Metabolic Reprogramming in Acute Myeloid Leukemia. Cells 2019; 8:cells8090967. [PMID: 31450562 PMCID: PMC6770240 DOI: 10.3390/cells8090967] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
The cancer metabolic reprogramming allows the maintenance of tumor proliferation, expansion and survival by altering key bioenergetics, biosynthetic and redox functions to meet the higher demands of tumor cells. In addition, several metabolites are also needed to perform signaling functions that further promote tumor growth and progression. These metabolic alterations have been exploited in different cancers, including acute myeloid leukemia, as novel therapeutic strategies both in preclinical models and clinical trials. Here, we review the complexity of acute myeloid leukemia (AML) metabolism and discuss how therapies targeting different aspects of cellular metabolism have demonstrated efficacy and how they provide a therapeutic window that should be explored to target the metabolic requirements of AML cells.
Collapse
Affiliation(s)
- Isabel Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal.
| |
Collapse
|
23
|
Visnjic D, Dembitz V, Lalic H. The Role of AMPK/mTOR Modulators in the Therapy of Acute Myeloid Leukemia. Curr Med Chem 2019; 26:2208-2229. [PMID: 29345570 DOI: 10.2174/0929867325666180117105522] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 01/01/2018] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Differentiation therapy of acute promyelocytic leukemia with all-trans retinoic acid represents the most successful pharmacological therapy of acute myeloid leukemia (AML). Numerous studies demonstrate that drugs that inhibit mechanistic target of rapamycin (mTOR) and activate AMP-kinase (AMPK) have beneficial effects in promoting differentiation and blocking proliferation of AML. Most of these drugs are already in use for other purposes; rapalogs as immunosuppressants, biguanides as oral antidiabetics, and 5-amino-4-imidazolecarboxamide ribonucleoside (AICAr, acadesine) as an exercise mimetic. Although most of these pharmacological modulators have been widely used for decades, their mechanism of action is only partially understood. In this review, we summarize the role of AMPK and mTOR in hematological malignancies and discuss the possible role of pharmacological modulators in proliferation and differentiation of leukemia cells.
Collapse
Affiliation(s)
- Dora Visnjic
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 12, 10 000 Zagreb, Croatia
| | - Vilma Dembitz
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 12, 10 000 Zagreb, Croatia
| | - Hrvoje Lalic
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 12, 10 000 Zagreb, Croatia
| |
Collapse
|
24
|
Ibrahim SM, Bakhashab S, Ilyas AM, Pushparaj PN, Karim S, Khan JA, Abuzenadah AM, Chaudhary AG, Al-Qahtani MH, Ahmed F. WYE-354 restores Adriamycin sensitivity in multidrug-resistant acute myeloid leukemia cell lines. Oncol Rep 2019; 41:3179-3188. [PMID: 30942458 PMCID: PMC6489006 DOI: 10.3892/or.2019.7093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/05/2019] [Indexed: 12/15/2022] Open
Abstract
Multidrug resistance (MDR) is a major reason for the failure of acute myeloid leukemia (AML) therapy. Agents that reverse MDR and sensitize AML cells to chemotherapy are of great clinical significance. The present study developed Adriamycin (Adr)-resistant cell lines, namely K562/Adr200 and K562/Adr500, which exhibited MDR. The upregulation of ATP-binding cassette subfamily B member 1 (ABCB1) was confirmed as the mechanism of resistance by reverse transcription-quantitative polymerase chain reaction and western blot analyses. Subsequently, the role of the mammalian target of rapamycin (mTOR) kinase inhibitor, WYE-354, in sensitizing the K562/Adr200 and K562/Adr500 cell lines to Adr was evaluated. At sub-cytotoxic concentrations, WYE-354 increased Adr cytotoxicity in the K562/Adr200 and K562/Adr500 cells. WYE-354 restored Adr sensitivity in the resistant cells by inhibiting ABCB1-mediated substrate efflux, thereby leading to an accumulation of Adr, an increase in Adr-mediated G2/M cell cycle arrest and the induction of apoptosis. Furthermore, WYE-354 stimulated the ATPase activity of ABCB1, which was consistent with in silico predictions using a human ABCB1 mouse homology model, indicating that WYE-354 is a potent substrate of ABCB1. WYE-354 did not regulate the expression of ABCB1 at the concentrations used in the present study. These findings indicate that WYE-354 may be a competitive inhibitor of ABCB1-mediated efflux and a potential candidate in combination with standard chemotherapy for overcoming MDR. Further clinical investigations are warranted to validate this combination in vivo.
Collapse
Affiliation(s)
- Sara M Ibrahim
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sherin Bakhashab
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asad M Ilyas
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Peter N Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jalaluddin A Khan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Adel M Abuzenadah
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Adeel G Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammed H Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Farid Ahmed
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
25
|
|
26
|
Alves R, Gonçalves AC, Jorge J, Alves J, Alves da Silva A, Freitas-Tavares P, Nascimento Costa JM, Almeida AM, Sarmento-Ribeiro AB. Everolimus in combination with Imatinib overcomes resistance in Chronic myeloid leukaemia. Med Oncol 2019; 36:30. [DOI: 10.1007/s12032-019-1253-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/31/2019] [Indexed: 11/28/2022]
|
27
|
Targeting mTOR in Acute Lymphoblastic Leukemia. Cells 2019; 8:cells8020190. [PMID: 30795552 PMCID: PMC6406494 DOI: 10.3390/cells8020190] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/12/2019] [Accepted: 02/16/2019] [Indexed: 12/12/2022] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) is an aggressive hematologic disorder and constitutes approximately 25% of cancer diagnoses among children and teenagers. Pediatric patients have a favourable prognosis, with 5-years overall survival rates near 90%, while adult ALL still correlates with poorer survival. However, during the past few decades, the therapeutic outcome of adult ALL was significantly ameliorated, mainly due to intensive pediatric-based protocols of chemotherapy. Mammalian (or mechanistic) target of rapamycin (mTOR) is a conserved serine/threonine kinase belonging to the phosphatidylinositol 3-kinase (PI3K)-related kinase family (PIKK) and resides in two distinct signalling complexes named mTORC1, involved in mRNA translation and protein synthesis and mTORC2 that controls cell survival and migration. Moreover, both complexes are remarkably involved in metabolism regulation. Growing evidence reports that mTOR dysregulation is related to metastatic potential, cell proliferation and angiogenesis and given that PI3K/Akt/mTOR network activation is often associated with poor prognosis and chemoresistance in ALL, there is a constant need to discover novel inhibitors for ALL treatment. Here, the current knowledge of mTOR signalling and the development of anti-mTOR compounds are documented, reporting the most relevant results from both preclinical and clinical studies in ALL that have contributed significantly into their efficacy or failure.
Collapse
|
28
|
Tian T, Li X, Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int J Mol Sci 2019; 20:ijms20030755. [PMID: 30754640 PMCID: PMC6387042 DOI: 10.3390/ijms20030755] [Citation(s) in RCA: 383] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) pathway plays a crucial role in regulation of cell survival, metabolism, growth and protein synthesis in response to upstream signals in both normal physiological and pathological conditions, especially in cancer. Aberrant mTOR signaling resulting from genetic alterations from different levels of the signal cascade is commonly observed in various types of cancers. Upon hyperactivation, mTOR signaling promotes cell proliferation and metabolism that contribute to tumor initiation and progression. In addition, mTOR also negatively regulates autophagy via different ways. We discuss mTOR signaling and its key upstream and downstream factors, the specific genetic changes in the mTOR pathway and the inhibitors of mTOR applied as therapeutic strategies in eight solid tumors. Although monotherapy and combination therapy with mTOR inhibitors have been extensively applied in preclinical and clinical trials in various cancer types, innovative therapies with better efficacy and less drug resistance are still in great need, and new biomarkers and deep sequencing technologies will facilitate these mTOR targeting drugs benefit the cancer patients in personalized therapy.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Xiaoyi Li
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| |
Collapse
|
29
|
Pergolizzi B, Panuzzo C, Ali MS, Lo Iacono M, Levron CL, Ponzone L, Prelli M, Cilloni D, Calautti E, Bozzaro S, Bracco E. Mammals and Dictyostelium rictor mutations swapping reveals two essential Gly residues for mTORC2 activity and integrity. J Cell Sci 2019; 132:jcs.236505. [DOI: 10.1242/jcs.236505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022] Open
Abstract
mTORC2 regulates a variety of vital cellular processes, and its aberrant functioning is often associated with various diseases. Rictor is a peculiar and distinguishing mTORC2 component playing a pivotal role in controlling its assembly and activity. Among living organisms Rictor is conserved from unicellular eukaryotes to metazoan. We replaced two distinct, but conserved, glycines in both the Dictyostelium piaA gene and its human ortholog, rictor. The two conserved residues are spaced by approximately 50 aminoacids and both are embedded within a conserved region falling in between the Ras-GEFN2 and Rictor_V domains. The effects of point mutations on the mTORC2 activity and integrity were assessed by biochemical and functional assays.In both cases, the reciprocal exchange between mammals and Dictyostelium rictor and piaA gene point mutations impaired mTORC2 activity and integrity.Our data indicate that the two Gly residues are essential for the maintenance of mTORC2 activity and integrity in organisms that appear to be distantly related, suggesting a primeval role in the assembly and proper TOR complex 2 functioning.
Collapse
Affiliation(s)
- Barbara Pergolizzi
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - M. Shahzad Ali
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Marco Lo Iacono
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Chiara Levra Levron
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Luca Ponzone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Marta Prelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Enzo Calautti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Salvatore Bozzaro
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Enrico Bracco
- Department of Oncology, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| |
Collapse
|
30
|
Goroshchuk O, Kolosenko I, Vidarsdottir L, Azimi A, Palm-Apergi C. Polo-like kinases and acute leukemia. Oncogene 2019; 38:1-16. [PMID: 30104712 DOI: 10.1038/s41388-018-0443-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/09/2018] [Accepted: 07/20/2018] [Indexed: 12/17/2022]
Abstract
Acute leukemia is a common malignancy among children and adults worldwide and many patients suffer from chronic health issues using current therapeutic approaches. Therefore, there is a great need for the development of novel and more specific therapies with fewer side effects. The family of Polo-like kinases (Plks) is a group of five serine/threonine kinases that play an important role in cell cycle regulation and are critical targets for therapeutic invention. Plk1 and Plk4 are novel targets for cancer therapy as leukemic cells often express higher levels than normal cells. In contrast, Plk2 and Plk3 are considered to be tumor suppressors. Several small molecule inhibitors have been developed for targeting Plk1 inhibition. Despite reaching phase III clinical trials, one of the ATP-competitive Plk1 inhibitor, volasertib, did not induce an objective clinical response and even caused lethal side effects in some patients. In order to improve the specificity of the Plk1 inhibitors and reduce off-target side effects, novel RNA interference (RNAi)-based therapies have been developed. In this review, we summarize the mechanisms of action of the Plk family members in acute leukemia, describe preclinical studies and clinical trials involving Plk-targeting drugs and discuss novel approaches in Plk targeting.
Collapse
Affiliation(s)
- Oksana Goroshchuk
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Iryna Kolosenko
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Linda Vidarsdottir
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Alireza Azimi
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Palm-Apergi
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
31
|
Chen L, Zhao H, Wang C, Hu N. TUG1 knockdown enhances adriamycin cytotoxicity by inhibiting glycolysis in adriamycin-resistant acute myeloid leukemia HL60/ADR cells. RSC Adv 2019; 9:10897-10904. [PMID: 35515331 PMCID: PMC9062713 DOI: 10.1039/c9ra00306a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/29/2019] [Indexed: 01/16/2023] Open
Abstract
Taurine-upregulated gene 1 (TUG1) has been reported as an oncogenic long non-coding RNA (lncRNA) in acute myeloid leukemia (AML). Nevertheless, the roles and molecular mechanism of TUG1 in drug resistance of AML cells are still unclear. Glycolysis level was evaluated by detecting glucose consumption and lactate production. qRT-PCR and Western blot were performed to detect TUG1, hexokinase2 (HK2) and pyruvate kinase isoenzyme M2 (PKM2) expressions. Adriamycin (ADR) cytotoxicity and apoptosis were assessed by MTT assay and flow cytometry, respectively. The changes of the protein kinase B (Akt) pathway were determined by Western blot analysis of phosphorylated-Akt (p-Akt) (ser473) and Akt. Our results showed that glycolysis was increased in HL60/ADR cells, as evidenced by the elevated glucose consumption and lactate production, as well as the increased HK2 and PKM2 expressions at mRNA and protein levels. TUG1 was up-regulated in HL60/ADR cells and TUG knockdown inhibited glycolysis. TUG1 knockdown enhanced ADR-induced cytotoxicity and apoptosis in HL60/ADR cells. TUG1 knockdown inhibited the Akt pathway and activation of the Akt pathway by 740Y-P attenuated the effects of TUG1 knockdown on ADR-induced cytotoxicity and apoptosis, as well as glycolysis in HL60/ADR cells. Taken together, TUG1 knockdown enhances adriamycin cytotoxicity in HL60/ADR cells via inhibiting the glycolysis by inactivating the Akt pathway. Taurine-upregulated gene 1 (TUG1) has been reported as an oncogenic long non-coding RNA (lncRNA) in acute myeloid leukemia (AML).![]()
Collapse
Affiliation(s)
- Li Chen
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| | - Hongmian Zhao
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| | - Chao Wang
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| | - Ning Hu
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| |
Collapse
|
32
|
Identification and targeting of novel CDK9 complexes in acute myeloid leukemia. Blood 2018; 133:1171-1185. [PMID: 30587525 DOI: 10.1182/blood-2018-08-870089] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/07/2018] [Indexed: 01/09/2023] Open
Abstract
Aberrant activation of mTOR signaling in acute myeloid leukemia (AML) results in a survival advantage that promotes the malignant phenotype. To improve our understanding of factors that contribute to mammalian target of rapamycin (mTOR) signaling activation and identify novel therapeutic targets, we searched for unique interactors of mTOR complexes through proteomics analyses. We identify cyclin dependent kinase 9 (CDK9) as a novel binding partner of the mTOR complex scaffold protein, mLST8. Our studies demonstrate that CDK9 is present in distinct mTOR-like (CTOR) complexes in the cytoplasm and nucleus. In the nucleus, CDK9 binds to RAPTOR and mLST8, forming CTORC1, to promote transcription of genes important for leukemogenesis. In the cytoplasm, CDK9 binds to RICTOR, SIN1, and mLST8, forming CTORC2, and controls messenger RNA (mRNA) translation through phosphorylation of LARP1 and rpS6. Pharmacological targeting of CTORC complexes results in suppression of growth of primitive human AML progenitors in vitro and elicits strong antileukemic responses in AML xenografts in vivo.
Collapse
|
33
|
Huang Q, Xie L, Chen X, Yu H, Lv Y, Huang X, Ying J, Zheng C, Cheng Y, Huang J. Synthesis and anticancer activity of novel rapamycin C-28 containing triazole moiety compounds. Arch Pharm (Weinheim) 2018; 351:e1800123. [PMID: 30357890 DOI: 10.1002/ardp.201800123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 09/16/2018] [Accepted: 09/26/2018] [Indexed: 11/10/2022]
Abstract
Rapamycin is an mTOR allosteric inhibitor with multiple functions such as immunosuppressive, anticancer, and lifespan prolonging activities. Its C-43 semi-synthetic derivatives temsirolimus and everolimus have been used as mTOR targeting anticancer drugs in the clinic. Following our previous research on antitumor rapalogs modified on the C-43 position, 13 novel rapamycin triazole hybrids (6a-g, 7a-f) were designed and synthesized on the C-28 position of rapamycin via Huisgen's reaction. Anticancer assays indicated that the targeted derivatives containing phenyl and 4-methylphenyl groups showed an obvious raise in anticancer activity. On the contrary, the compounds with methoxyl, amine, and halogen groups on the benzene ring displayed lower anticancer activity. Compound 6c, as the most active compound, showed a stronger inhibition effect as compared with rapamycin for almost all of the tested cell lines (p < 0.01), except PC-3. Meanwhile, the effect of 6c on inducing apoptosis and cell cycle arrest in A549 cells was more powerful than that of rapamycin. In addition, 6c inhibited the phosphorylation of mTOR and its downstream key kinases 4EBP1 and p70S6K1 in A549 cells, indicating that 6c also effectively inhibits the mTORC1 signaling pathway as rapamycin. On the basis of these findings, 6c may have the potential to be developed as a new mTOR inhibitor against specific cancers.
Collapse
Affiliation(s)
- Qingwen Huang
- Fujian University of Traditional Chinese Medicine, Fuzhou, P. R. China.,Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Lijun Xie
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Xiaoming Chen
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Hui Yu
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Yubing Lv
- Hangzhou Huadong Medicine Group Pharmaceutical Research Institute Co. Ltd., Hangzhou, P. R. China
| | - XueHui Huang
- Hangzhou Huadong Medicine Group Pharmaceutical Research Institute Co. Ltd., Hangzhou, P. R. China
| | - Jiayin Ying
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Congshen Zheng
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Yuanrong Cheng
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| | - Jie Huang
- Fujian University of Traditional Chinese Medicine, Fuzhou, P. R. China.,Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, P. R. China
| |
Collapse
|
34
|
Chemical modulation of autophagy as an adjunct to chemotherapy in childhood and adolescent brain tumors. Oncotarget 2018; 9:35266-35277. [PMID: 30443293 PMCID: PMC6219655 DOI: 10.18632/oncotarget.26186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023] Open
Abstract
Brain tumors are the leading cause of cancer-related death in children and are the most challenging childhood cancer in relation to diagnosis, treatment, and outcome. One potential novel strategy to improve outcomes in cancer involves the manipulation of autophagy, a fundamental process in all cells. In cancer, autophagy can be thought of as having a "Janus"-like duality. On one face, especially in the early phases of cancer formation, autophagy can act as a cellular housekeeper to eliminate damaged organelles and recycle macromolecules, thus acting as tumor suppressor. On the other face, at later stages of tumor progression, autophagy can function as a pro-survival pathway in response to metabolic stresses such as nutrient depravation, hypoxia and indeed to chemotherapy itself, and can support cell growth by supplying much needed energy. In the context of chemotherapy, autophagy may, in some cases, mediate resistance to treatment. We present an overview of the relevance of autophagy in central nervous system tumors including how its chemical modulation can serve as a useful adjunct to chemotherapy, and use this knowledge to consider how targeting of autophagy may be relevant in pediatric brain tumors.
Collapse
|
35
|
Zadi Heydarabad M, Vatanmakanian M, Abdolalizadeh J, Mohammadi H, Azimi A, Mousavi Ardehaie R, Movasaghpour A, Farshdousti Hagh M. Apoptotic effect of resveratrol on human T-ALL cell line CCRF-CEM is unlikely exerted through alteration of BAX and BCL2 promoter methylation. J Cell Biochem 2018; 119:10033-10040. [PMID: 30132966 DOI: 10.1002/jcb.27333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/28/2018] [Indexed: 11/09/2022]
Abstract
One of the fundamental barriers leading to failure of leukemia therapy is the resistance against conventional chemotherapies, common modality used to cure leukemia. Having the potential to trigger apoptosis in various human leukemia cell lines, resveratrol is regarded as a robust agent in chemotherapy regimens. The current study was aimed to assess whether the apoptotic effect of resveratrol on T-cell acute lymphoblastic leukemia cell line, CCRF-CEM, is exerted through DNA methylation of BAX and BCL2 gene promoters. For this purpose, the CCRF-CEM cells were treated by resveratrol under standard cell culture. To analyze the promoter DNA methylation changes, we used methylation-specific polymerase chain reaction technique following the resveratrol treatment at different dosages and time intervals. Based on our previous study, the resveratrol treatment can trigger apoptosis in CCRF-CEM cell line via upregulation of apoptotic BAX gene and downregulation of antiapoptotic BCL2 gene. Despite these alterations in gene expression, the current study reveals no changes in DNA methylation patterns of subjected genes following the resveratrol treatment. Unchanged status of DNA methylation of BAX and BCL2 genes may suggest that resveratrol causes the gene expression changes through a distinct mechanism which requires further studies to be understood.
Collapse
Affiliation(s)
| | - Mousa Vatanmakanian
- Department of Hematology, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ako Azimi
- Department of Basic Science, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Reza Mousavi Ardehaie
- Department of Biochemistry and Clinical Laboratory, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliakbar Movasaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
36
|
Massimino M, Stella S, Tirrò E, Romano C, Pennisi MS, Puma A, Manzella L, Zanghì A, Stagno F, Di Raimondo F, Vigneri P. Non ABL-directed inhibitors as alternative treatment strategies for chronic myeloid leukemia. Mol Cancer 2018; 17:56. [PMID: 29455672 PMCID: PMC5817805 DOI: 10.1186/s12943-018-0805-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/01/2018] [Indexed: 02/07/2023] Open
Abstract
The introduction of ABL Tyrosine Kinase Inhibitors (TKIs) has significantly improved the outcome of Chronic Myeloid Leukemia (CML) patients that, in large part, achieve satisfactory hematological, cytogenetic and molecular remissions. However, approximately 15-20% fail to obtain optimal responses according to the current European Leukemia Network recommendation because of drug intolerance or resistance.Moreover, a plethora of evidence suggests that Leukemic Stem Cells (LSCs) show BCR-ABL1-independent survival. Hence, they are unresponsive to TKIs, leading to disease relapse if pharmacological treatment is discontinued.All together, these biological events generate a subpopulation of CML patients in need of alternative therapeutic strategies to overcome TKI resistance or to eradicate LSCs in order to allow cure of the disease.In this review we update the role of "non ABL-directed inhibitors" targeting signaling pathways downstream of the BCR-ABL1 oncoprotein and describe immunological approaches activating specific T cell responses against CML cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Combined Modality Therapy
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy
- Signal Transduction/drug effects
- Treatment Outcome
Collapse
Affiliation(s)
- Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Chiara Romano
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Adriana Puma
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Antonino Zanghì
- Department of Surgical Medical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Fabio Stagno
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Francesco Di Raimondo
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Department of Surgery, Medical and Surgical Specialties, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy.
| |
Collapse
|
37
|
Alameen AAM, Simioni C, Martelli AM, Zauli G, Ultimo S, McCubrey JA, Gonelli A, Marisi G, Ulivi P, Capitani S, Neri LM. Healthy CD4+ T lymphocytes are not affected by targeted therapies against the PI3K/Akt/mTOR pathway in T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:55690-55703. [PMID: 27494886 PMCID: PMC5342446 DOI: 10.18632/oncotarget.10984] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 12/19/2022] Open
Abstract
An attractive molecular target for novel anti-cancer therapies is the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway which is commonly deregulated in many types of cancer. Nevertheless, the effects of PI3K/Akt/mTOR inhibitors on T lymphocytes, a key component of immune responses, have been seldom explored. In this study we investigated the effects on human CD4+ T-cells of a panel of PI3K/Akt/mTOR inhibitors: BGT226, Torin-2, MK-2206, and ZSTK474. We also assessed their efficacy against two acute leukemia T cell lines. T lymphocytes were stimulated with phytohemagglutinin. Inhibitor effects on cell cycle and apoptosis were analyzed by flow cytometry, while cytotoxicity was assessed by MTT assays. In addition, the activation status of the pathway as well as induction of autophagy were analyzed by Western blotting. Quiescent healthy T lymphocytes were unaffected by the drugs whereas mitogen-stimulated lymphocytes as well as leukemic cell lines displayed a cell cycle block, caspase-dependent apoptosis, and dephosphorylation of key components of the signaling pathway. Autophagy was also induced in proliferating lymphocytes and in JURKAT and MOLT-4 cell lines. When autophagy was inhibited by 3-methyladenine or Bafilomycin A1, drug cytotoxicity was increased, indicating that autophagy is a protective mechanism. Therefore, our findings suggest that PI3K/Akt/mTOR inhibitors preserve lymphocyte viability. This is a valuable result to be taken into account when selecting drugs for targeted cancer therapy in order to minimize detrimental effects on immune function.
Collapse
Affiliation(s)
- Ayman A M Alameen
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Department of Chemical Pathology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Simona Ultimo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,LTTA Center, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
38
|
Zhang XH, Wang XY, Zhou ZW, Bai H, Shi L, Yang YX, Zhou SF, Zhang XC. The combination of digoxin and GSK2606414 exerts synergistic anticancer activity against leukemia in vitro and in vivo. Biofactors 2017; 43:812-820. [PMID: 28817203 DOI: 10.1002/biof.1380] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/14/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Digoxin is a member of cardiac glycosides and recent studies show that digoxin plays anticancer role in several types of cancer. However, the anticancer effects and mechanism of digoxin in leukemia is largely unknown. Her, our data show that digoxin treatment significantly inhibits leukemia cell viability. In addition, digoxin treatment significantly induced apoptosis and G2/M cell cycle arrest in leukemia cells. Furthermore, we demonstrated that digoxin treatment inactivate that oncogenic pathway Akt/mTOR signaling in leukemia cells. In addition, our data show that digoxin treatment induces activation of unfolded protein response (UPR) signaling in leukemia cells. Interestingly, our in vitro and in vivo experiments show that combination treatment of digoxin and UPR inhibitor can synergistically suppress leukemia growth and induces apoptosis and cell cycle arrest compared to single drug treatment. In summary, our findings indicate that digoxin has potential anticancer effects on leukemia. The combination of digoxin and UPR signaling inhibitor can exerts synergistic anticancer activity against leukemia. © 2017 BioFactors, 43(6):812-820, 2017.
Collapse
Affiliation(s)
- Xue-Hong Zhang
- Department of Pediatrics, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xin-Yu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
- Department of Pharmacy and Institute of Clinical Pharmacology, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Zhi-Wei Zhou
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hua Bai
- Department of Pediatrics, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Lin Shi
- Department of Pediatrics, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Yin-Xue Yang
- Department of Colorectal Surgery, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xiao-Chun Zhang
- Department of Pediatrics, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| |
Collapse
|
39
|
Zhang X, Rastogi P, Shah B, Zhang L. B lymphoblastic leukemia/lymphoma: new insights into genetics, molecular aberrations, subclassification and targeted therapy. Oncotarget 2017; 8:66728-66741. [PMID: 29029550 PMCID: PMC5630450 DOI: 10.18632/oncotarget.19271] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/07/2017] [Indexed: 12/18/2022] Open
Abstract
B lymphoblastic leukemia/lymphoma (B-ALL) is a clonal hematopoietic stem cell neoplasm derived from B-cell progenitors, which mostly occurs in children and adolescents and is regarded as one of top leading causes of death related to malignancies in this population. Despite the majority of patients with B-ALL have fairly good response to conventional chemotherapeutic interventions followed by hematopoietic stem cell transplant for the last decades, a subpopulation of patients show chemo-resistance and a high relapse rate. Adult B-ALL exhibits similar clinical course but worse prognosis in comparison to younger individuals. Ample evidences have shown that the clinical behavior, response rate and clinical outcome of B-ALL rely largely on its genetic and molecular profiles, such as the presence of BCR-ABL1 fusion gene which is an independent negative prognostic predictor. New B-ALL subtypes have been recognized with recurrent genetic abnormalities, including B-ALL with intrachromosomal amplification of chromosome 21 (iAMP21), B-ALL with translocations involving tyrosine kinases or cytokine receptors (“BCR-ABL1-like ALL”). Genome-wide genetic profiling studies on B-ALL have extended our understanding of genomic landscape of B-ALL, and genetic mutations involved in various key pathways have been illustrated. These include CRLF2 and PAX5 alterations, TP53, CREBBP and ERG mutations, characteristic genetic aberrations in BCR-ABL1-like B-ALL and others. The review further provides new insights into clinical implication of the genetic aberrations in regard to targeted therapy development.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Prerna Rastogi
- Department of Pathology, University of Iowa College of Medicine, Iowa City, Iowa, USA
| | - Bijal Shah
- Department of Hematological Malignancies, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Ling Zhang
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
40
|
Control of B lymphocyte development and functions by the mTOR signaling pathways. Cytokine Growth Factor Rev 2017; 35:47-62. [PMID: 28583723 DOI: 10.1016/j.cytogfr.2017.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022]
Abstract
Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase originally discovered as the molecular target of the immunosuppressant rapamycin. mTOR forms two compositionally and functionally distinct complexes, mTORC1 and mTORC2, which are crucial for coordinating nutrient, energy, oxygen, and growth factor availability with cellular growth, proliferation, and survival. Recent studies have identified critical, non-redundant roles for mTORC1 and mTORC2 in controlling B cell development, differentiation, and functions, and have highlighted emerging roles of the Folliculin-Fnip protein complex in regulating mTOR and B cell development. In this review, we summarize the basic mechanisms of mTOR signaling; describe what is known about the roles of mTORC1, mTORC2, and the Folliculin/Fnip1 pathway in B cell development and functions; and briefly outline current clinical approaches for targeting mTOR in B cell neoplasms. We conclude by highlighting a few salient questions and future perspectives regarding mTOR in B lineage cells.
Collapse
|
41
|
Espinoza JL, Elbadry MI, Taniwaki M, Harada K, Trung LQ, Nakagawa N, Takami A, Ishiyama K, Yamauchi T, Takenaka K, Nakao S. The simultaneous inhibition of the mTOR and MAPK pathways with Gnetin-C induces apoptosis in acute myeloid leukemia. Cancer Lett 2017; 400:127-136. [PMID: 28456658 DOI: 10.1016/j.canlet.2017.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/08/2017] [Accepted: 04/14/2017] [Indexed: 11/27/2022]
Abstract
Acute myelogenous leukemia (AML) is a clinically heterogeneous disease that is frequently associated with relapse and a poor prognosis. Among the various subtypes, AML with the monosomal karyotype (AML-MK) has an extremely unfavorable prognosis. We performed screening to identify antitumor compounds that are capable of inducing apoptosis in primary leukemia cells harboring the AML-MK karyotype and identified a naturally occurring stilbene, Gnetin-C, with potent anti-tumor activities against AML cells from patients with various cytogenetic abnormalities, including patients with the AML-MK karyotype. Gnetin-C simultaneously inhibits the ERK1/2 and the AKT/mTOR pathways, two signals that are essential for the survival of leukemia cells. A combination of Gnetin-C with low doses of chemotherapeutic drugs led to synergistic anti-tumor effects against AML cells. In an immunodeficient mouse model of human leukemia, Gnetin-C attenuated the formation of leukemia, depleted leukemia cells and improved survival. These findings suggest that Gnetin-C has antitumor activities in AML and supports the therapeutic potential of blocking two different pathways in AML.
Collapse
Affiliation(s)
- J Luis Espinoza
- Department of Hematology and Oncology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.
| | - Mahmoud I Elbadry
- Department of Hematology and Oncology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Masafumi Taniwaki
- Center for Molecular Diagnostic and Therapeutic, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kenichi Harada
- Department of Human Pathology, Graduate School of Medical Science, Kanazawa University, Takara Machi 13-1, Kanazawa, Ishikawa, 920-8640, Japan
| | - Ly Quoc Trung
- Department of Hematology and Oncology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Noriharu Nakagawa
- Department of Hematology and Oncology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Akiyoshi Takami
- Department of Internal Medicine, Division of Hematology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Ken Ishiyama
- Department of Hematology and Oncology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Katsuto Takenaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shinji Nakao
- Department of Hematology and Oncology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
42
|
Deng L, Jiang L, Lin XH, Tseng KF, Liu Y, Zhang X, Dong RH, Lu ZG, Wang XJ. The PI3K/mTOR dual inhibitor BEZ235 suppresses proliferation and migration and reverses multidrug resistance in acute myeloid leukemia. Acta Pharmacol Sin 2017; 38:382-391. [PMID: 28042875 DOI: 10.1038/aps.2016.121] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/21/2016] [Indexed: 02/06/2023] Open
Abstract
Aberrant activation of the PI3K/Akt/mTOR pathway contributes to the proliferation of malignant cells, and may confer resistance to chemotherapy in various malignancies, including acute myeloid leukemia (AML). Chemoresistance is the major reason for relapse in AML. RAD001 (everolimus) has been used at d1 and d7 of an induction chemotherapy regimen for AML, which has acceptable toxicity and may improve conventional chemotherapeutic treatment. Dual inhibitors of PI3K and mTOR overcome some of the intrinsic disadvantages of rapamycin and its derivatives. In this study, we evaluated the effects of BEZ235, a PI3K/mTOR dual inhibitor, on the multidrug-resistant AML cell lines HL-60/VCR and K562/ADR in vitro. BEZ235 dose-dependently inhibited the viability of HL-60/VCR and K562/ADR cells with the IC50 values of 66.69 and 71.44 nmol/L, respectively. BEZ235 (25-100 nmol/L) dose-dependently inhibited the migration of the two AML cell lines, and it also significantly sensitized the two AML cell lines to VCR and ADR. After treatment with BEZ235, the miR-1-3p levels were markedly increased in HL-60/VCR cells. Using TargetScan analysis and luciferase assays, we showed that miR-1-3p targeted BAG4, EDN1 and ABCB1, the key regulators of cell apoptosis, migration and multidrug resistance, and significantly decreased their levels in the two AML cell lines. Transfection of HL-60/VCR and K562/ADR cells with miR-1-3p-AMO to inhibit miR-1-3p could reverse the anti-proliferation effects of BEZ235. In conclusion, the PI3K/mTOR dual inhibitor BEZ235 effectively chemosensitizes AML cells via increasing miR-1-3p and subsequently down-regulating BAG4, EDN1 and ABCB1.
Collapse
|
43
|
Ragon BK, Kantarjian H, Jabbour E, Ravandi F, Cortes J, Borthakur G, DeBose L, Zeng Z, Schneider H, Pemmaraju N, Garcia-Manero G, Kornblau S, Wierda W, Burger J, DiNardo CD, Andreeff M, Konopleva M, Daver N. Buparlisib, a PI3K inhibitor, demonstrates acceptable tolerability and preliminary activity in a phase I trial of patients with advanced leukemias. Am J Hematol 2017; 92:7-11. [PMID: 27673440 DOI: 10.1002/ajh.24568] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/01/2016] [Accepted: 09/26/2016] [Indexed: 01/03/2023]
Abstract
Phosphatidylinositol-3-kinase (PI3K) signaling plays a crucial role in oncogene-mediated tumor growth and proliferation. Buparlisib (BKM120) is an oral pan-class I PI3K inhibitor. This phase I study was conducted to determine the dose limiting toxicity (DLT) and maximum tolerated dose (MTD) of BKM120 in patients (pts) with relapsed/refractory acute leukemias. Fourteen pts (12 acute myeloid leukemia, 1 acute lymphoblastic leukemia, and 1 mixed phenotype leukemia) were enrolled. Twelve pts received BKM-120 80 mg/day and two 100 mg/day. The MTD was 80 mg/day. Of the 14 patients treated, the best response was stable disease in one patient that lasted 82 days. The median survival for all patients was 75 days (range 10-568). Three patients with a 3q26 chromosome abnormality had a significantly improved median survival of 360 days (range 278-568) as compared to a median survival of 57 days (range, 10-125) among the 11 other patients. The most frequent drug-related toxicities included confusion, mucositis, dysphagia, and fatigue. Western blot profiling revealed a decrease in p-pS6K/total pS6K in 5/7 (71%) available patient samples with a mean quantitative inhibition of 65% (range, 32-100%) and a decrease in p-FOXO3/total FOXO3 in 4/6 (67%) samples with a mean quantitative inhibition of 93% (range, 89-100%). BKM120 administered at 80 mg/day showed modest efficacy and was tolerable in advanced acute leukemias. Am. J. Hematol. 92:7-11, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Brittany Knick Ragon
- Hematology/Oncology Fellowship, Division of Cancer Medicine; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hagop Kantarjian
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Elias Jabbour
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Farhad Ravandi
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Jorge Cortes
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Gautam Borthakur
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - LaKiesha DeBose
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Zhihong Zeng
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Heather Schneider
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naveen Pemmaraju
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Steven Kornblau
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - William Wierda
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Jan Burger
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Courtney D DiNardo
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Michael Andreeff
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Marina Konopleva
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naval Daver
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
44
|
Petrov I, Suntsova M, Mutorova O, Sorokin M, Garazha A, Ilnitskaya E, Spirin P, Larin S, Zhavoronkov A, Kovalchuk O, Prassolov V, Roumiantsev A, Buzdin A. Molecular pathway activation features of pediatric acute myeloid leukemia (AML) and acute lymphoblast leukemia (ALL) cells. Aging (Albany NY) 2016; 8:2936-2947. [PMID: 27870639 PMCID: PMC5182073 DOI: 10.18632/aging.101102] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 11/04/2016] [Indexed: 12/11/2022]
Abstract
Acute lymphoblast leukemia (ALL) is characterized by overproduction of immature white blood cells in the bone marrow. ALL is most common in the childhood and has high (>80%) cure rate. In contrast, acute myeloid leukemia (AML) has far greater mortality rate than the ALL and is most commonly affecting older adults. However, AML is a leading cause of childhood cancer mortality. In this study, we compare gene expression and molecular pathway activation patterns in three normal blood, seven pediatric ALL and seven pediatric AML bone marrow samples. We identified 172/94 and 148/31 characteristic gene expression/pathway activation signatures, clearly distinguishing pediatric ALL and AML cells, respectively, from the normal blood. The pediatric AML and ALL cells differed by 139/34 gene expression/pathway activation biomarkers. For the adult 30 AML and 17 normal blood samples, we found 132/33 gene expression/pathway AML-specific features, of which only 7/2 were common for the adult and pediatric AML and, therefore, age-independent. At the pathway level, we found more differences than similarities between the adult and pediatric forms. These findings suggest that the adult and pediatric AMLs may require different treatment strategies.
Collapse
MESH Headings
- Adolescent
- Adult
- Age Factors
- Biomarkers, Tumor
- Bone Marrow
- Case-Control Studies
- Child
- Child, Preschool
- Female
- Gene Expression
- Gene Expression Profiling
- Humans
- Infant
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Male
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
Collapse
Affiliation(s)
- Ivan Petrov
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
- First Oncology Research and Advisory Center, Moscow, 117997, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow region, 141700, Russia
| | - Maria Suntsova
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Olga Mutorova
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
- Morozov Pediatric Clinical Hospital, Moscow, 101000, Russia
| | - Maxim Sorokin
- National Research Centre “Kurchatov Institute”, Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, 123182, Russia
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
| | - Andrew Garazha
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow region, 141700, Russia
| | - Elena Ilnitskaya
- First Oncology Research and Advisory Center, Moscow, 117997, Russia
| | - Pavel Spirin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Mosow, Russia,119991
| | - Sergey Larin
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
| | - Alex Zhavoronkov
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
- First Oncology Research and Advisory Center, Moscow, 117997, Russia
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Vladimir Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Mosow, Russia,119991
| | - Alexander Roumiantsev
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
| | - Anton Buzdin
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
- National Research Centre “Kurchatov Institute”, Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, 123182, Russia
| |
Collapse
|
45
|
Chen X, Xiong W, Li H. Comparison of microRNA expression profiles in K562-cells-derived microvesicles and parental cells, and analysis of their roles in leukemia. Oncol Lett 2016; 12:4937-4948. [PMID: 28105201 PMCID: PMC5228523 DOI: 10.3892/ol.2016.5308] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/20/2016] [Indexed: 02/07/2023] Open
Abstract
Microvesicles (MVs) are 30-1,000-nm extracellular vesicles that are released from a multitude of cell types and perform diverse cellular functions, including intercellular communication, antigen presentation, and transfer of proteins, messenger RNA and microRNA (also known as miR). MicroRNAs have been demonstrated to be aberrantly expressed in leukemia, and the overall microRNA expression profile may differentiate normal blood cells vs. leukemia cells. MVs containing microRNAs may enable intercellular cross-talk in vivo. This prompted us to investigate specific variations of microRNA expression patterns in MVs derived from leukemia cells. The present study examined the microRNA expression profile of MVs from chronic myeloid leukemia K562 cells and that of MVs from normal human volunteers' peripheral blood cells. The potential targets of the differentially expressed microRNAs were predicted using computational searches. Bioinformatic analyses of the predicted target genes were performed for further evaluation. The present study analyzed microRNAs of MVs derived from leukemia and normal cells, and characterized specific microRNAs expression. The results revealed that MVs derived from K562 cells expressed 181 microRNAs of the 888 microRNAs assessed. Further analysis revealed that 16 microRNAs were downregulated, while 7 were upregulated in these MVs. In addition, significant differences in microRNA expression profiles between MVs derived from K562 cells and K562 cells were identified. The present results revealed that 77 and 122 microRNAs were only expressed in MVs derived from K562 cells and in K562 cells, respectively. There were 104 microRNAs co-expressed in MVs derived from K562 cells and in K562 cells. Target gene-related pathway analyses demonstrated that the majority of the dysregulated microRNAs were involved in pathways associated with leukemia, particularly the mitogen-activated protein kinase (MAPK) and the p53 signaling pathways. By further conducting microRNA gene network analysis, the present study revealed that the miR-15a/b, miR-16, miR-17 and miR-30 families were likely to play a role in the regulation of the MAPK signaling pathway. Since K562 cells presented the t(9;22) translocation, the current study further examined the predicted function of 12 microRNAs located in chromosomes 9 [Homo sapiens (hsa)-let-7a, hsa-let-7f, miR-126, miR-126*, miR-23b, miR-24, miR-27b and miR-7] and 22 (hsa-let-7b, miR-1249, miR-130b and miR-185), which were expressed both in MVs derived from K562 cells and in K562 cells. The present study identified microRNAs of MVs from leukemia and normal cells, and characterized the expression of specific microRNAs. The current study is also the first to identify and characterize distinct microRNA expression between MVs derived from K562 cells and K562 cells. These findings highlight that a number of microRNAs from leukemia-derived MVs may contribute to the development of hematopoietic malignancies. Further investigation may reveal the function of these differentially expressed microRNAs and may provide potential targets for novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaomei Chen
- Center for Biotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Wei Xiong
- Center of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang University School of Medicine at Binjiang, Hangzhou, Zhejiang 310009, P.R. China
| | - Huiyu Li
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|