1
|
Jia MX, Di DL, Liu ZZ, Wang HY, Chen L. Successful treatment of a chronic myeloid leukemia patient with extreme thrombocytosis by a combination of imatinib and interferon‑α: A case report. Exp Ther Med 2025; 29:50. [PMID: 39885917 PMCID: PMC11775754 DOI: 10.3892/etm.2025.12800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
Chronic myeloid leukemia with extreme thrombocytosis (CML-T), defined by a platelet count >1,000x109/l is a rare leukemia subtype. The present case report described a 66-year-old female CML-T patient presenting with a platelet count of 3,798x109/l, but a consistently normal spleen size. Following treatment with imatinib combined with interferon-α, the patient achieved hematological remission within 2 months, with a platelet count reduction to 311x109/l and complete cytogenetic remission after 10 months. The patient experienced myocardial infarction and liver injury during treatment, which was managed with supportive care. The present case suggested that imatinib combined with interferon-α may be a safe and effective treatment option for patients with CML-T and extreme thrombocytosis and suboptimal response to imatinib monotherapy.
Collapse
Affiliation(s)
- Meng-Xiao Jia
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| | - Da-Lin Di
- Department of Immunology, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhen-Zhen Liu
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| | - Hai-Ying Wang
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| | - Lei Chen
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
2
|
Yuan Y, Wei X, Xiong X, Wang X, Jiang W, Kuang Q, Zhu K, Chen C, Gan J, Li J, Yang J, Li L, Luo P. STAP2 promotes the progression of renal fibrosis via HSP27. J Transl Med 2024; 22:1018. [PMID: 39533293 PMCID: PMC11556045 DOI: 10.1186/s12967-024-05776-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Renal fibrosis is a key process in the progression from acute kidney injury (AKI) to chronic kidney disease (CKD), while the intricate mechanisms of renal fibrosis remain obscure. While the signal-transducing adaptor protein 2 (STAP2) was well-studied for its notable function in inflammation and immune-related disorders, its specific implication in renal fibrosis remains unclear. This study assessed the mechanism by which STAP2 could promote the progression of renal fibrosis. METHODS The expression level of STAP2 in fibrotic human samples, murine fibrosis models, and cellular fibrosis models was measured, respectively. Subsequently, immunoprecipitation (IP), mass spectrometry, and RNA sequencing (RNA-seq) were employed to identify HSP27 as an interacting protein and the PI3K-AKT signaling pathway. STAP2 was thereafter knocked down or overexpressed in both in vivo and in vitro models to assess the expression levels of pathway-related and fibrosis-related proteins. Finally, the important role of STAP2 in the fibrosis process in animal models induced by ischemia-reperfusion injury (IRI) and cisplatin was validated. RESULTS Functionally, in vivo assays demonstrated that the genetic knockout of STAP2 could remarkably mitigate epithelial-mesenchymal transition (EMT), diminish inflammatory cell infiltration, and reduce collagen deposition in mice with renal fibrosis. Conversely, in vitro assays employing STAP2-overexpressing cell models exacerbated the expression levels of fibrosis markers. The outcomes uncovered a potential mechanism by which STAP2 could modulate renal fibrosis through its impact on the expression level of phosphorylated HSP27, as well as modulating the PI3K/AKT signaling pathway. CONCLUSIONS This comprehensive investigation delineated the noticeable function of STAP2 in the advancement of renal fibrosis, and the outcomes might contribute to the development of targeted therapies concentrated on STAP2 to mitigate renal fibrosis.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao Wei
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Xi Xiong
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiong Wang
- Department of Pharmacy, Wuhan Third Hospital, Wuhan, 430060, China
| | - Wei Jiang
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University Huangshi, Huangshi, 435000, China
| | - Qihui Kuang
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Kai Zhu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen Chen
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430060, China
| | - Jingzheng Gan
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430060, China
| | - Junjie Li
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430060, China
| | - Jun Yang
- Department of Urology, Wuhan Third Hospital, Wuhan, 430060, China.
| | - Lili Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Pengcheng Luo
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
3
|
Li Y. DNA Adducts in Cancer Chemotherapy. J Med Chem 2024; 67:5113-5143. [PMID: 38552031 DOI: 10.1021/acs.jmedchem.3c02476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
DNA adducting drugs, including alkylating agents and platinum-containing drugs, are prominent in cancer chemotherapy. Their mechanisms of action involve direct interaction with DNA, resulting in the formation of DNA addition products known as DNA adducts. While these adducts are well-accepted to induce cancer cell death, understanding of their specific chemotypes and their role in drug therapy response remain limited. This perspective aims to address this gap by investigating the metabolic activation and chemical characterization of DNA adducts formed by the U.S. FDA-approved drugs. Moreover, clinical studies on DNA adducts as potential biomarkers for predicting patient responses to drug efficacy are examined. The overarching goal is to engage the interest of medicinal chemists and stimulate further research into the use of DNA adducts as biomarkers for guiding personalized cancer treatment.
Collapse
|
4
|
Jiang L, Hong H, Xiang S, Li H, Ji J, Lan M, Luo B. Integrated gene co-expression network analysis and experimental validation revealed potential targets of human urine extract CDA-II in treating chronic myeloid leukemia. Genomics 2024; 116:110806. [PMID: 38325533 DOI: 10.1016/j.ygeno.2024.110806] [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: 09/11/2023] [Revised: 01/24/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Cell differentiation agent II (CDA-II) exhibits potent anti-proliferative and apoptosis-inducing properties against a variety of cancer cells. However, its mechanism of action in chronic myeloid leukemia (CML) remains unclear. METHODS Cell counting Kit 8 (CCK-8) and flow cytometry were used to investigate the effects of CDA-II on the biological characteristics of K562 cells. Gene (mRNA and lncRNA) expression profiles were analyzed by bioinformatics to screen differentially expressed genes and to perform enrichment analysis. The Pearson correlation coefficients of lncRNAs and mRNAs were calculated using gene expression values, and a lncRNA/mRNA co-expression network was constructed. The MCODE and cytoHubba plugins were used to analyze the co-expression network. RESULTS The Results, derived from CCK-8 and flow cytometry, indicated that CDA-II exerts dual effects on K562 cells: it inhibits their proliferation and induces apoptosis. From bioinformatics analysis, we identified 316 mRNAs and 32 lncRNAs. These mRNAs were predominantly related to the meiotic cell cycle, DNA methylation, transporter complex and peptidase regulator activity, complement and coagulation cascades, protein digestion and absorption, and cell adhesion molecule signaling pathways. The co-expression network comprised of 163 lncRNA/mRNA interaction pairs. Notably, our analysis results implicated clustered histone gene families and five lncRNAs in the biological effects of CDA-II on K562 cells. CONCLUSION This study highlights the hub gene and lncRNA/mRNA co-expression network as crucial elements in the context of CDA-II treatment of CML. This insight not only enriches our understanding of CDA-II's mechanism of action but also might provide valuable clues for subsequent experimental studies of CDA-II, and potentially contribute to the discovery of new therapeutic targets for CML.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Critical Care Medicine, Intensive Care Unit, The People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Qingxiu District, Nanning 530021, China
| | - Haoyuan Hong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | - Shulin Xiang
- Department of Critical Care Medicine, Intensive Care Unit, The People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Qingxiu District, Nanning 530021, China
| | - Han Li
- Department of Hematology, The People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Qingxiu District, Nanning 530021, China
| | - Jianyu Ji
- Department of Critical Care Medicine, Intensive Care Unit, The People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Qingxiu District, Nanning 530021, China
| | - Mei Lan
- Department of Hematology, The People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Qingxiu District, Nanning 530021, China
| | - Bin Luo
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR 999078, China; Department of Hematology, The People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Qingxiu District, Nanning 530021, China.
| |
Collapse
|
5
|
Tsubaki M, Takeda T, Koumoto Y, Usami T, Matsuda T, Seki S, Sakai K, Nishio K, Nishida S. Activation of ERK1/2 by MOS and TPL2 leads to dasatinib resistance in chronic myeloid leukaemia cells. Cell Prolif 2023:e13420. [PMID: 36847709 DOI: 10.1111/cpr.13420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 03/01/2023] Open
Abstract
The development of BCR::ABL1 tyrosine kinase inhibitors (TKIs), such as dasatinib, has dramatically improved survival in cases of chronic myeloid leukaemia (CML). However, the development of resistance to BCR::ABL1 TKIs is a clinical problem. BCR::ABL1 TKI resistance is known to have BCR::ABL1-dependent or BCR::ABL1-independent mechanisms, but the mechanism of BCR::ABL1 independence is not well understood. In the present study, we investigated the mechanism of BCR::ABL1-independent dasatinib resistance. The expression and activation level of genes or proteins were evaluated using array CGH, real time PCR, or western blot analysis. Gene expression was modulated using siRNA-mediated knockdown. Cell survival was assessed by using trypan blue dye method. We found that dasatinib-resistant K562/DR and KU812/DR cells did not harbour a BCR::ABL1 mutation but had elevated expression and/or activation of MOS, TPL2 and ERK1/2. In addition, MOS siRNA, TPL2 siRNA and trametinib resensitized dasatinib-resistant cells to dasatinib. Moreover, expression levels of MOS in dasatinib non-responder patients with CML were higher than those in dasatinib responders, and the expression of TPL2 tended to increase in dasatinib non-responder patients compared with that in responder patients. Our results indicate that activation of ERK1/2 by elevated MOS and TPL2 expression is involved in dasatinib resistance, and inhibition of these proteins overcomes dasatinib resistance. Therefore, MOS, TPL2 and ERK1/2 inhibitors may be therapeutically useful for treating BCR::ABL1-independent dasatinib-resistant CML.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Yuuichi Koumoto
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Takehiro Usami
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| |
Collapse
|
6
|
Chai Y, Chen F, Li Z, Yang P, Zhou Q, Liu W, Xi Y. Mechanism of salidroside in the treatment of chronic myeloid leukemia based on the network pharmacology and molecular docking. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:384-395. [PMID: 36369630 DOI: 10.1007/s12094-022-02990-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Salidroside is a phenolic natural product, which is a kind of Rhodiola rosea. It has been confirmed that it has inhibitory effects on chronic myeloid leukemia, but the specific performance of its molecular effects is still unclear. OBJECTIVE To systematically study the pharmacological mechanism of salidroside on chronic myeloid leukemia by means of network pharmacology. METHODS First, the possible target genes of salidroside were predicted through the Traditional Chinese Medicine Pharmacology Database and Analysis Platform, the target gene names were converted into standardized gene names using the Uniprot website. At the same time, the related target genes of chronic myeloid leukemia were collected from GeneCards and DisGenet; Collect summary data and screen for commonly targeted genes. Then, the above-mentioned intersected genes were imported into the String website to construct the protein-protein interaction (PPI) network, and the Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were further analyzed. To investigate the overall pharmacological effects of salidroside on chronic myeloid leukemia, we constructed a drug component-target gene-disease (CTD) network. Finally, molecular docking was performed to verify the possible binding conformation between salidroside and the candidate target. RESULTS A total of 126 salidroside target genes were retrieved, and 106 of them had interactions with chronic myeloid leukemia. The pharmacological effects of salidroside on chronic myeloid leukemia are related to some important oncogenes and signaling pathways. Molecular docking studies confirmed that the main role of salidroside binding to the target genes is hydrogen bonding. CONCLUSIONS We revealed the potential mechanism of action of salidroside against chronic myeloid leukemia, verified by network pharmacology combined with molecular docking. However, salidroside is a promising drug for the prevention and treatment of chronic myeloid leukemia, and further research is needed to prove it.
Collapse
Affiliation(s)
- Yihong Chai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Feng Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zijian Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Department of Hematology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Panpan Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Department of Hematology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Qi Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Wenling Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yaming Xi
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China. .,Department of Hematology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
7
|
Studies on 1,4-Quinone Derivatives Exhibiting Anti-Leukemic Activity along with Anti-Colorectal and Anti-Breast Cancer Effects. Molecules 2022; 28:molecules28010077. [PMID: 36615273 PMCID: PMC9822417 DOI: 10.3390/molecules28010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC), breast cancer, and chronic myeloid leukemia (CML) are life-threatening malignancies worldwide. Although potent therapeutic and screening strategies have been developed so far, these cancer types are still major public health problems. Therefore, the exploration of more potent and selective new agents is urgently required for the treatment of these cancers. Quinones represent one of the most important structures in anticancer drug discovery. We have previously identified a series of quinone-based compounds (ABQ-1-17) as anti-CML agents. In the current work, ABQ-3 was taken to the National Cancer Institute (NCI) for screening to determine its in vitro antiproliferative effects against a large panel of human tumor cell lines at five doses. ABQ-3 revealed significant growth inhibition against HCT-116 CRC and MCF-7 breast cancer cells with 2.00 µM and 2.35 µM GI50 values, respectively. The MTT test also showed that ABQ-3 possessed anticancer effects towards HCT-116 and MCF-7 cells with IC50 values of 5.22 ± 2.41 μM and 7.46 ± 2.76 μM, respectively. Further experiments indicated that ABQ-3 induced apoptosis in both cell lines, and molecular docking studies explicitly suggested that ABQ-3 exhibited DNA binding in a similar fashion to previously reported compounds. Based on in silico pharmacokinetic prediction, ABQ-3 might display drug-like features enabling this compound to become a lead molecule for future studies.
Collapse
|
8
|
Pathobiology and Therapeutic Relevance of GSK-3 in Chronic Hematological Malignancies. Cells 2022; 11:cells11111812. [PMID: 35681507 PMCID: PMC9180032 DOI: 10.3390/cells11111812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 12/10/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is an evolutionarily conserved, ubiquitously expressed, multifunctional serine/threonine protein kinase involved in the regulation of a variety of physiological processes. GSK-3 comprises two isoforms (α and β) which were originally discovered in 1980 as enzymes involved in glucose metabolism via inhibitory phosphorylation of glycogen synthase. Differently from other proteins kinases, GSK-3 isoforms are constitutively active in resting cells, and their modulation mainly involves inhibition through upstream regulatory networks. In the early 1990s, GSK-3 isoforms were implicated as key players in cancer cell pathobiology. Active GSK-3 facilitates the destruction of multiple oncogenic proteins which include β-catenin and Master regulator of cell cycle entry and proliferative metabolism (c-Myc). Therefore, GSK-3 was initially considered to be a tumor suppressor. Consistently, GSK-3 is often inactivated in cancer cells through dysregulated upstream signaling pathways. However, over the past 10–15 years, a growing number of studies highlighted that in some cancer settings GSK-3 isoforms inhibit tumor suppressing pathways and therefore act as tumor promoters. In this article, we will discuss the multiple and often enigmatic roles played by GSK-3 isoforms in some chronic hematological malignancies (chronic myelogenous leukemia, chronic lymphocytic leukemia, multiple myeloma, and B-cell non-Hodgkin’s lymphomas) which are among the most common blood cancer cell types. We will also summarize possible novel strategies targeting GSK-3 for innovative therapies of these disorders.
Collapse
|