1
|
Song J, Liu J, Guo D, Li H, Fan S. Chidamide maintenance therapy after allo-HSCT in SET-NUP214 fusion positive T-ALL patients: A report of two cases. Transpl Immunol 2024; 87:102119. [PMID: 39233093 DOI: 10.1016/j.trim.2024.102119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a highly invasive hematological malignancy originated from T-lineage progenitor cells. The clonal proliferation and aggregation of primordial cells in bone marrow inhibit normal hematopoietic function, resulting in a series of hematocytopenia and infiltration symptoms. SET-NUP214 fusion is a recurrent event that is common in adult male T-ALL patients. It originates from chromosome del(9)(q34.11; q34.13) or t(9; 9)(q34; q34). Hematopoietic stem cell transplantation (HSCT) can significantly improve the survival rate of these patients. Due to the poor prognosis of patients and high relapse rate after remission, more effective strategies need to be proposed to improve prognosis and prevent relapse. Chidamide is a novel oral benzamide histone deacetylase inhibitor (HDACi) that can exert anti-tumor effects through multiple mechanisms. Here we report chidamide maintenance therapy after allo-HSCT in patients with SET-NUP214 fusion positive T-ALL. Both patients improved effectively during follow-up, confirming the efficacy of chidamide in improving the condition of these patients and may provide valuable clinical information for the treatment of this rare and understudied disease.
Collapse
Affiliation(s)
- Jingyu Song
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Jie Liu
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Dan Guo
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Huibo Li
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China.
| | - Shengjin Fan
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China; NHC Key Laboratory of Cell Transplantation, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
2
|
Tonozuka Y, Tanaka H, Nomura K, Sakaguchi K, Soeda J, Kakimoto Y. The combination of brentuximab vedotin and chidamide synergistically suppresses the proliferation of T-cell lymphoma cells through the enhancement of apoptosis. Cancer Chemother Pharmacol 2024; 93:137-149. [PMID: 37921901 PMCID: PMC10853311 DOI: 10.1007/s00280-023-04609-5] [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: 11/16/2022] [Accepted: 10/14/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE Peripheral T-cell lymphoma (PTCL) is an aggressive disease with a poor prognosis. Brentuximab vedotin (BV), an anti-CD30 monoclonal antibody linked to a microtubule-disrupting agent, has been approved for the treatment of PTCL. We evaluated a new effective combination partner of BV using non-clinical approaches that could potentially identify agents capable of improving survival times for patients with PTCL. METHODS A high-throughput screening test was used to select the most synergistic partner of BV from 14 candidate drugs that were under development or available in clinical practice for PTCL. HH cells, originating from an aggressive cutaneous T-cell lymphoma, were used as an experimental model of PTCL. Apoptotic effects of the synergistic partner of BV were further investigated in vitro and in vivo using HH-cell xenograft mice. RESULTS Chidamide (tucidinostat), a novel histone deacetylase inhibitor, was found to have the greatest synergistic effect with BV on HH cells. The combined effects of chidamide and BV were demonstrated in a study of HH-cell xenograft mice; mean tumor size following combined treatment was 22% of that observed in the control group, compared with 71% and 58% following chidamide and BV monotherapy, respectively. Further investigations in vitro and in vivo revealed that the levels of an anti-apoptotic protein, Bcl-2, and a rate-limiting factor of DNA replication, CDC45, were reduced in HH cells treated with chidamide combined with BV compared with the control group. CONCLUSION The use of chidamide in conjunction with BV may positively affect and enhance T-cellular apoptotic pathways without offsetting each other.
Collapse
Affiliation(s)
- Yukio Tonozuka
- Japan Medical Affairs, Japan Oncology Business Unit, Takeda Pharmaceutical Company Limited, 1-1 Nihonbashi Honcho 2-chome, Chuo-ku, Tokyo, 103-8668, Japan.
| | - Hiroshi Tanaka
- Integrated Biology, Integrated & Translational Science, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome Fujisawa, Kanagawa, 251-0012, Japan
| | - Kazumi Nomura
- Japan Medical Affairs, Japan Oncology Business Unit, Takeda Pharmaceutical Company Limited, 1-1 Nihonbashi Honcho 2-chome, Chuo-ku, Tokyo, 103-8668, Japan
| | - Kazuya Sakaguchi
- Frontier Technology, Integrated & Translational Science, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome Fujisawa, Kanagawa, 251-0012, Japan
| | - Junpei Soeda
- Japan Medical Affairs, Japan Oncology Business Unit, Takeda Pharmaceutical Company Limited, 1-1 Nihonbashi Honcho 2-chome, Chuo-ku, Tokyo, 103-8668, Japan
| | - Yoshihide Kakimoto
- Japan Medical Affairs, Japan Oncology Business Unit, Takeda Pharmaceutical Company Limited, 1-1 Nihonbashi Honcho 2-chome, Chuo-ku, Tokyo, 103-8668, Japan
| |
Collapse
|
3
|
Song W, Chen Z, Shi C, Gao Y, Feng X, Li H, Li Z, Zhang M. Synergistic anticancer effect of a combination of chidamide and etoposide against NK/T cell lymphoma. Hematol Oncol 2023; 41:257-266. [PMID: 34854108 DOI: 10.1002/hon.2954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
Abstract
Natural killer/T cell lymphoma (NKTCL) is a highly aggressive hematological malignancy. However, there is currently no consensus on therapies for refractory/relapsed patients. In this study, we investigated the synergistic anticancer effect and potential mechanism of combining chidamide, a histone deacetylases (HDACs) inhibitor, and etoposide, a DNA-damaging agent, in NKTCL. We demonstrated that chidamide or etoposide alone dose- and time-dependently inhibited the cell viability of NKTCL cell lines, YT, NKYS and KHYG-1. Functional experiments suggested that combined chidamide and etoposide treatment exerted synergistic antiproliferation effect and enhanced cell apoptotic death in vitro and in vivo. Furthermore, the expression of DNA damage related proteins was detected and we also examined the alternations in histone acetylation, cell cycle progression, and mitochondrial membrane potential (MMP). The results suggested that increased histone acetylation, cell cycle arrest at the G2/M phase and loss of MMP, converging to greater DNA damage, might account for the synergism of the combination of chidamide and etoposide in NKTCL. Taken together, our study provides an evident for possible application on combining HDACs inhibitors and DNA-damaging agents for the treatment of NKTCL.
Collapse
Grants
- 182102310114 Department of Science & Technology of Henan province
- 81970184 National Natural Science Foundation of China
- 82070209 National Natural Science Foundation of China
- 82170183 National Natural Science Foundation of China
- U1904139 National Natural Science Foundation of China
- Oncology Department, State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research, Translational Medical Center, Department of Nephrology of the First Affiliated Hospital of Zhengzhou University, and the Medical Sciences Academy and Research Institute of Nephrology of Zhengzhou University, and Core Unit of National Clinical Medical Research Center of Kidney Disease in Zhengzhou
- 182102310114 Department of Science & Technology of Henan province
- Oncology Department, State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research, Translational Medical Center, Department of Nephrology of the First Affiliated Hospital of Zhengzhou University, and the Medical Sciences Academy and Research Institute of Nephrology of Zhengzhou University, and Core Unit of National Clinical Medical Research Center of Kidney Disease in Zhengzhou
Collapse
Affiliation(s)
- Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhan Chen
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan, China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, Henan, China
| | - Cunzhen Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuyang Gao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyan Feng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongwen Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Song Y, Chen S, Liu C, Chen L, Wang W, Wu B, Liang Y. Chemo-free maintenance therapy in adult T-cell acute lymphoblastic leukemia: A case report and literature review. Front Pharmacol 2023; 14:1051305. [PMID: 36873995 PMCID: PMC9981645 DOI: 10.3389/fphar.2023.1051305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/07/2023] [Indexed: 02/19/2023] Open
Abstract
Maintenance therapy in adult T-cell acute lymphoblastic leukemia (T-ALL) is the longest phase but with limited option. The classic drugs used in the maintenance phase such as 6-mercaptopurine, methotrexate, corticosteroid and vincristine have potentially serious toxicities. Optimizing therapy in the modern age, chemo-free maintenance therapy regimens for patients with T-ALL may dramatically improve the maintenance therapeutic landscape. We report here the combination of Anti-programmed cell death protein 1 antibody and histone deacetylase inhibitor as chemo-free maintenance treatment in a T-ALL patient with literature review, thus providing a unique perspective in addition to valuable information which may inform novel therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | - Bingyi Wu
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yang Liang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
5
|
Gu Z, Yang J, Yang M, Deng Y, Jiao Y. Immunomodulatory effects of decitabine in pearl oyster Pinctada fucata martensii. FISH & SHELLFISH IMMUNOLOGY 2022; 129:191-198. [PMID: 36029945 DOI: 10.1016/j.fsi.2022.08.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Decitabine (DAC), an inhibitor of DNA methyltransferase, is typically used to reverse DNA methylation and is considered an epigenetic modifying drug. DNA methylation is crucial to the regulation of gene expression without altering genetic information. Our previous research showed that the DNA methylation levels of many immune-related genes changed after the pre-grafting condition in pearl production. In the present study, we evaluated the DNA methylation level and analyzed transcriptome, enzyme, and antimicrobial activities after DAC treatment to evaluate the effect of DAC on DNA methylation and immune system of pearl oyster Pinctada fucata martensii. Results showed that DAC significantly decreased the level of global DNA methylation in the hemocytes of the pearl oysters. Transcriptome analysis obtained 577 differentially expressed genes (DEGs) between the control and DAC treatment group. The DEGs were mainly enriched in the following pathways: "Relaxin signaling pathway," "Cytosolic DNA-sensing pathway," "Platelet activation," and "Peroxisome," and related genes were overexpressed after DAC treatment. DAC treatment resulted in a substantial increase in the levels of serum superoxide dismutase, interleukin-17, phenol oxidase, tumor necrosis factor, and antimicrobial activity, compared with the control. These results suggested that DAC can alter DNA methylation level, activate immune-related genes, and improve the level of humoral immunity in pearl oysters, thereby increasing our understanding of the mechanism underlying DNA methylation in immune regulation.
Collapse
Affiliation(s)
- Zefeng Gu
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Jingmiao Yang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Min Yang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Yuewen Deng
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, 524088, China
| | - Yu Jiao
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China.
| |
Collapse
|
6
|
Therapeutic Interaction of Apatinib and Chidamide in T-Cell Acute Lymphoblastic Leukemia through Interference with Mitochondria Associated Biogenesis and Intrinsic Apoptosis. J Pers Med 2021; 11:jpm11100977. [PMID: 34683119 PMCID: PMC8540063 DOI: 10.3390/jpm11100977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/28/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) shows poor clinical outcome and has limited therapeutic options, indicating that new treatment approaches for this disease are urgently required. Our previous study demonstrated that apatinib, an orally selective VEGFR-2 antagonist, is highly effective in T-ALL. Additionally, chidamide, a histone deacetylase inhibitor, has proven to be cytotoxic against T-ALL in preclinical and clinical settings. However, whether the therapeutic interaction of apatinib and chidamide in T-ALL remains unknown. In this study, apatinib and chidamide acted additively to decrease cell viability and induce apoptosis in T-ALL in vitro. Notably, compared with apatinib or chidamide alone, the combinational regimen was more efficient in abrogating the leukemia burden in the spleen and bone marrow of T-ALL patient-derived xenograft (PDX) models. Mechanistically, the additive antileukemia effect of apatinib and chidamide was associated with suppression of mitochondrial respiration and downregulation of the abundance levels of several rate-limiting enzymes that are involved in the citric acid cycle and oxidative phosphorylation (OXPHOS). In addition, apatinib enhanced the antileukemia effect of chidamide on T-ALL via activation of the mitochondria-mediated apoptosis pathway and impediment of mitochondrial biogenesis. Taken together, the study provides a potential role for apatinib in combination with chidamide in the management of T-ALL and warrants further clinical evaluations of this combination in patients with T-ALL.
Collapse
|
7
|
Zhang G, Gao X, Zhao X, Wu H, Yan M, Li Y, Zeng H, Ji Z, Guo X. Decitabine inhibits the proliferation of human T-cell acute lymphoblastic leukemia molt4 cells and promotes apoptosis partly by regulating the PI3K/AKT/mTOR pathway. Oncol Lett 2021; 21:340. [PMID: 33747197 PMCID: PMC7967925 DOI: 10.3892/ol.2021.12601] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/11/2020] [Indexed: 11/30/2022] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is a highly aggressive hematological cancer; however, there is a lack of effective chemotherapeutic or targeted drugs for the treatment of T-ALL. Decitabine is a DNA demethylation agent but it has not been used for T-ALL treatment. Therefore, the present study aimed to assess the inhibitory effect of decitabine on T-ALL molt4 cells and determine its regulatory role in the PI3K/AKT/mTOR pathway. Molt4 cells were stimulated with decitabine in vitro, after which cell proliferation, apoptosis and cell cycle analyses were performed to assess cell viability. Subcellular morphology was observed using transmission electron microscopy. Expression levels of phosphate and tension homology (PTEN), genes involved in the PI3K/AKT/mTOR pathway and the corresponding downstream genes were analyzed using reverse transcription-quantitative PCR and western blotting. The results showed that decitabine induced apoptosis, inhibited proliferation and arrested molt4 cells in the G2 phase. Following decitabine intervention, an increase in the number of lipid droplets, autophagosomes and mitochondrial damage was observed. At concentrations of 1 and 10 µM, decitabine downregulated the expression of PI3K, AKT, mTOR, P70S6 and eukaryotic initiating factor 4E-binding protein 1, which in turn upregulated PTEN expression; however, 50 µM decitabine downregulated PTEN levels. Overall, these results demonstrated that decitabine reduced the viability of molt4 cells partly by inhibiting the PI3K/AKT/mTOR pathway via PTEN, especially at low decitabine concentrations.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaohui Gao
- Department of Pediatrics, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaoyan Zhao
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Haibing Wu
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Minchao Yan
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Yuan Li
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Hui Zeng
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Zhaoning Ji
- Department of Medical Oncology, The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Xiaojun Guo
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
8
|
Peng J, Li SJ, Fu X, Liu Y, Zhao XL. Chidamide acts on the histone deacetylase-mediated miR-34a/Bcl-2 axis to regulate NB4 cell line proliferation and apoptosis. Kaohsiung J Med Sci 2020; 36:1004-1013. [PMID: 32783381 DOI: 10.1002/kjm2.12283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/22/2020] [Accepted: 07/08/2020] [Indexed: 12/25/2022] Open
Abstract
Acute promyelocytic leukemia (APL), a biologically and clinically distinct variant of acute myelogenous leukemia, is characterized by the fusion of the N-terminus of promyelocytic leukemia protein to the C terminus of retinoic acid receptor alpha, mostly due to chromosomal translocation t(15;17). Chidamide, a synthetic analogue of MS-275 identified from a group of benzamide-type compounds, has been found to have efficient anticancer activity in basic and clinical research studies. However, the concrete role and underlying mechanism of Chidamide in the treatment of APL has not been well characterized. Our data demonstrate that Chidamide inhibited the expression of histone deacetylase (HDAC) to induce apoptosis and suppress proliferation in NB4 cells. Mechanistically, Chidamide increases the expression of miR-34a by suppressing HDAC. Furthermore, B-cell lymphoma-2 (Bcl-2) is a direct target of miR-34a, the expression of which is regulated by miR-34a. Functionally, Chidamide inhibits cell proliferation and promotes apoptosis through miR-34a/Bcl-2. Chidamide exerts its anticancer effect via the HDAC-mediated miR-34a/Bcl-2 axis, providing potential targets for APL therapy.
Collapse
Affiliation(s)
- Jie Peng
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shu-Jun Li
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao Fu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xie-Lan Zhao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Wang XT, Guo W, Sun M, Han W, Du ZH, Wang XX, Du BB, Bai O. Effect of chidamide on treating hepatosplenic T-cell lymphoma: A case report. World J Clin Cases 2020; 8:3122-3129. [PMID: 32775395 PMCID: PMC7385602 DOI: 10.12998/wjcc.v8.i14.3122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/26/2020] [Accepted: 07/04/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Hepatosplenic T-cell lymphoma (HSTCL) is a rare subtype of non-Hodgkin’s lymphoma, which has an aggressive clinical course and an extremely poor prognosis. Chidamide is a novel, orally active, benzamide-type histone deacetylase (HDAC) inhibitor that has been used for peripheral T-cell lymphoma (PTCL) treatment. However, to date, there has been no report of the treatment and effect of the HDAC inhibitor chidamide in HSTCL, which is a special subtype of PTCL.
CASE SUMMARY A 45-year-old male patient was admitted with splenomegaly and slight bicytopenia. He was diagnosed with HSTCL via splenectomy. The patient was treated with fractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with high-dose methotrexate and cytarabine regiment as inductive therapy. Unfortunately, the disease progressed rapidly during chemotherapy before a suitable allogeneic gene transplant donor was found. The chidamide-combined chemotherapy regimen and single-drug oral maintenance regimen achieved complete remission, duration of response of 9 mo, and overall survival of 15 mo.
CONCLUSION The novel agent chidamide can be used in HSTCL to achieve deep remission and improve the duration of response and overall survival.
Collapse
Affiliation(s)
- Xing-Tong Wang
- Department of Hematology, The First Hospital of Jilin University, Jilin Provincial Hematology Research Institute, National Key Discipline, Changchun 130021, Jilin Province, China
| | - Wei Guo
- Department of Hematology, The First Hospital of Jilin University, Jilin Provincial Hematology Research Institute, National Key Discipline, Changchun 130021, Jilin Province, China
| | - Mo Sun
- Department of Pathology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Wei Han
- Department of Hematology, The First Hospital of Jilin University, Jilin Provincial Hematology Research Institute, National Key Discipline, Changchun 130021, Jilin Province, China
| | - Zhong-Hua Du
- Department of Hematology, The First Hospital of Jilin University, Jilin Provincial Hematology Research Institute, National Key Discipline, Changchun 130021, Jilin Province, China
| | - Xiu-Xiu Wang
- Department of Hematology, The First Hospital of Jilin University, Jilin Provincial Hematology Research Institute, National Key Discipline, Changchun 130021, Jilin Province, China
| | - Bei-Bei Du
- Department of Cardiology, The Third Hospital of Jilin University, Changchun, Jilin Province 130031, China
| | - Ou Bai
- Department of Hematology, The First Hospital of Jilin University, Jilin Provincial Hematology Research Institute, National Key Discipline, Changchun 130021, Jilin Province, China
| |
Collapse
|
10
|
Alves J, Dexheimer GM, Reckzigel L, Goettert M, Biolchi V, Abujamra AL. Changes in IDH2, TET2 and KDM2B Gene Expression After Treatment With Classic Chemotherapeutic Agents and Decitabine in Myelogenous Leukemia Cell Lines. J Hematol 2020; 8:89-101. [PMID: 32300452 PMCID: PMC7153660 DOI: 10.14740/jh531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/22/2019] [Indexed: 11/11/2022] Open
Abstract
Background Hematological malignancies are a heterogeneous group of tumors with increased proliferative and auto-replicative capacity. Despite treatment advances, post-treatment quality of life remains highly affected. Studies addressing the molecular mechanisms of these diseases are critical for the development of effective, rapid and selective therapies, since few therapeutic strategies succeed in being effective without triggering high-grade toxicities or debilitating late effects. Our aim of this study was to verify changes in the expression of genes involved in the malignant phenotype of hematological malignancies, by treating human cell lines in vitro with classic chemotherapeutic agents and the demethylating agent, decitabine. Methods KASUMI-1 and K-562 human myeloid leukemia cell lines were plated at a density of 3 × 104 cells/well and treated with increasing concentrations of different chemotherapeutic agents commonly used in the clinical setting. After 24 and 48 h of treatment, cell viability was tested, and RNA was extracted. Complementary DNA (cDNA) was synthesized and quantitative real-time polymerase chain reaction (qPCR) was performed to evaluate the gene expression of IDH2, TET2 and KDM2B. Results A modulation in gene expression was observed before and after treatment with classic chemotherapeutic agents. It was possible to demonstrate a difference in gene expression when cells were treated with chemotherapeutic agents or decitabine alone when compared to chemotherapeutic agents in association with decitabine. Conclusions The genes tested, and the modulation of their expression during in vitro treatments suggest that IDH2, TET2, and KDM2B should be further investigated as potential biomarkers for ongoing treatment response and follow-up for patients diagnosed with hematological malignancies of the myeloid lineage.
Collapse
Affiliation(s)
- Jayse Alves
- Graduate Program in Biotechnology, Univates, Lajeado, R.S., 95914-014, Brazil
| | | | - Laura Reckzigel
- Biological and Health Sciences Center, Univates, Lajeado, R.S., 95914-014, Brazil
| | - Marcia Goettert
- Graduate Program in Biotechnology, Univates, Lajeado, R.S., 95914-014, Brazil
| | - Vanderlei Biolchi
- Biological and Health Sciences Center, Univates, Lajeado, R.S., 95914-014, Brazil
| | - Ana Lucia Abujamra
- Graduate Program in Biotechnology, Univates, Lajeado, R.S., 95914-014, Brazil
| |
Collapse
|
11
|
Guan W, Jing Y, Dou L, Wang M, Xiao Y, Yu L. Chidamide in combination with chemotherapy in refractory and relapsed T lymphoblastic lymphoma/leukemia. Leuk Lymphoma 2019; 61:855-861. [PMID: 31755348 DOI: 10.1080/10428194.2019.1691195] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chidamide, a novel histone deacetylase inhibitor, has exerted effects in T-cell tumors through various mechanisms. Seventeen patients with refractory or relapsed T-cell acute lymphoblastic lymphoma/leukemia (T-LBL/ALL) received Chidamide combined with chemotherapy as salvage treatment. Historical data was analyzed as comparison as chemotherapy group. Complete response (CR) rate and overall response rate (ORR) of Chidamide + chemotherapy group were higher than that of chemotherapy group after one course. Chidamide + chemotherapy group had a better progress-free survival (PFS) compared to chemotherapy group. No difference in overall survival (OS) was observed. Grade 3/4 nonhematological adverse events (>10%) of patients in Chidamide + chemotherapy group included febrile neutropenia (64.7%), drug-induced liver failure (17.6%), decreased fibrinogen (11.8%), sepsis (11.8%), pneumonitis (11.8%), and oral mucositis (11.8%). This study demonstrates that Chidamide included regimen may be a new treatment strategy with an acceptable safety profile for refractory or relapsed T-LBL/ALL patients but requires further investigation.
Collapse
Affiliation(s)
- Wei Guan
- Department of Hematology, Chinese PLA General Hospital, Beijing, China.,Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, China
| | - Yu Jing
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Liping Dou
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Maoquan Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Yang Xiao
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Li Yu
- Department of Hematology, Chinese PLA General Hospital, Beijing, China.,Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, China.,School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
12
|
Ye J, Zha J, Shi Y, Li Y, Yuan D, Chen Q, Lin F, Fang Z, Yu Y, Dai Y, Xu B. Co-inhibition of HDAC and MLL-menin interaction targets MLL-rearranged acute myeloid leukemia cells via disruption of DNA damage checkpoint and DNA repair. Clin Epigenetics 2019; 11:137. [PMID: 31590682 PMCID: PMC6781368 DOI: 10.1186/s13148-019-0723-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/05/2019] [Indexed: 02/08/2023] Open
Abstract
While the aberrant translocation of the mixed-lineage leukemia (MLL) gene drives pathogenesis of acute myeloid leukemia (AML), it represents an independent predictor for poor prognosis of adult AML patients. Thus, small molecule inhibitors targeting menin-MLL fusion protein interaction have been emerging for the treatment of MLL-rearranged AML. As both inhibitors of histone deacetylase (HDAC) and menin-MLL interaction target the transcription-regulatory machinery involving epigenetic regulation of chromatin remodeling that governs the expression of genes involved in tumorigenesis, we hypothesized that these two classes of agents might interact to kill MLL-rearranged (MLL-r) AML cells. Here, we report that the combination treatment with subtoxic doses of the HDAC inhibitor chidamide and the menin-MLL interaction inhibitor MI-3 displayed a highly synergistic anti-tumor activity against human MLL-r AML cells in vitro and in vivo, but not those without this genetic aberration. Mechanistically, co-exposure to chidamide and MI-3 led to robust apoptosis in MLL-r AML cells, in association with loss of mitochondrial membrane potential and a sharp increase in ROS generation. Combined treatment also disrupted DNA damage checkpoint at the level of CHK1 and CHK2 kinases, rather than their upstream kinases (ATR and ATM), as well as DNA repair likely via homologous recombination (HR), but not non-homologous end joining (NHEJ). Genome-wide RNAseq revealed gene expression alterations involving several potential signaling pathways (e.g., cell cycle, DNA repair, MAPK, NF-κB) that might account for or contribute to the mechanisms of action underlying anti-leukemia activity of chidamide and MI-3 as a single agent and particularly in combination in MLL-r AML. Collectively, these findings provide a preclinical basis for further clinical investigation of this novel targeted strategy combining HDAC and Menin-MLL interaction inhibitors to improve therapeutic outcomes in a subset of patients with poor-prognostic MLL-r leukemia.
Collapse
Affiliation(s)
- Jing Ye
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yuanfei Shi
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yin Li
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Delin Yuan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Qinwei Chen
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Fusheng Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Zhihong Fang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yong Yu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
13
|
Yuan XG, Huang YR, Yu T, Jiang HW, Xu Y, Zhao XY. Chidamide, a histone deacetylase inhibitor, induces growth arrest and apoptosis in multiple myeloma cells in a caspase-dependent manner. Oncol Lett 2019; 18:411-419. [PMID: 31289512 PMCID: PMC6540238 DOI: 10.3892/ol.2019.10301] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/29/2019] [Indexed: 12/25/2022] Open
Abstract
Chidamide, a novel histone deacetylase (HDAC) inhibitor, induces antitumor effects in various types of cancer. The present study aimed to evaluate the cytotoxic effect of chidamide on multiple myeloma and the underlying mechanisms involved. Viability of multiple myeloma cells upon chidamide treatment was determined by the Cell Counting Kit-8 assay. Apoptosis induction and cell cycle alteration were detected by flow cytometry. Specific apoptosis-associated proteins and cell cycle proteins were evaluated by western blot analysis. Chidamide suppressed cell viability in a time- and dose-dependent manner. Chidamide treatment markedly suppressed the expression of type I HDACs and further induced the acetylation of histones H3 and H4. In addition, it promoted G0/G1 arrest by decreasing cyclin D1 and c-myc expression, and increasing phosphorylated-cellular tumor antigen p53 and cyclin-dependent kinase inhibitor 1 (p21) expression in a dose-dependent manner. Treatment with chidamide induced cell apoptosis by upregulating the apoptosis regulator Bax/B-cell lymphoma 2 ratio in a caspase-dependent manner. In addition, the combination of chidamide with bortezomib, a proteasome inhibitor widely used as a therapeutic agent for multiple myeloma, resulted in enhanced inhibition of cell viability. In conclusion, chidamide induces a marked antimyeloma effect by inducing G0/G1 arrest and apoptosis via a caspase-dependent pathway. The present study provides evidence for the clinical application of chidamide in multiple myeloma.
Collapse
Affiliation(s)
- Xiang-Gui Yuan
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yu-Rong Huang
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Teng Yu
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Hua-Wei Jiang
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yang Xu
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiao-Ying Zhao
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
14
|
Roolf C, Richter A, Konkolefski C, Knuebel G, Sekora A, Krohn S, Stenzel J, Krause BJ, Vollmar B, Murua Escobar H, Junghanss C. Decitabine demonstrates antileukemic activity in B cell precursor acute lymphoblastic leukemia with MLL rearrangements. J Hematol Oncol 2018; 11:62. [PMID: 29728108 PMCID: PMC5936021 DOI: 10.1186/s13045-018-0607-3] [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: 03/02/2018] [Accepted: 04/26/2018] [Indexed: 01/05/2023] Open
Abstract
Background Promotor hypermethylation of CpG islands is common in B cell precursor acute lymphoblastic leukemia (BCP-ALL) with mixed lineage leukemia (MLL) gene rearrangements. Hypomethylating agents (HMA) such as azacitidine (AZA) and decitabine (DEC) reduce DNA hypermethylation by incorporation into DNA and were successfully introduced into the clinic for the treatment of myeloid neoplasias. Methods Here, we investigated whether HMA induce comparable biological effects in MLL-positive BCP-ALL. Further, efficacy of HMA and concomitant application of cytostatic drugs (cytarabine and doxorubicin) were evaluated on established SEM and RS4;11 cell lines. In addition, promising approaches were studied on BCP-ALL cell line- and patient-derived xenograft models. Results In general, DEC effects were stronger compared to AZA on MLL-positive BCP-ALL cells. DEC significantly reduced proliferation by induction of cell cycle arrest in G0/G1 phase and apoptosis. Most sensitive to HMA were SEM cells which are characterized by a fast cell doubling time. The combination of low-dose HMA and conventional cytostatic agents revealed a heterogeneous response pattern. The strongest antiproliferative effects were observed when ALL cells were simultaneously exposed to HMA and cytostatic drugs. Most potent synergistic effects of HMA were induced with cytarabine. Finally, the therapeutic potential of DEC was evaluated on BCP-ALL xenograft models. DEC significantly delayed leukemic proliferation in xenograft models as demonstrated longitudinally by non-invasive bioluminescence as well as 18F-FDG-PET/CT imaging. Unexpectedly, in vivo concomitant application of DEC and cytarabine did not enhance the antiproliferative effect compared to DEC monotherapy. Conclusions Our data reveal that DEC is active in MLL-positive BCP-ALL and warrant clinical evaluation. Electronic supplementary material The online version of this article (10.1186/s13045-018-0607-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- C Roolf
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - A Richter
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - C Konkolefski
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - G Knuebel
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - A Sekora
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - S Krohn
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - J Stenzel
- Department of Nuclear Medicine, Rostock University Medical Center, University of Rostock, Gertrudenplatz 1, 18057, Rostock, Germany
| | - B J Krause
- Department of Nuclear Medicine, Rostock University Medical Center, University of Rostock, Gertrudenplatz 1, 18057, Rostock, Germany
| | - B Vollmar
- Institute of Experimental Surgery, Rostock University Medical Center, University of Rostock, Schillingallee 69a, 18057, Rostock, Germany
| | - H Murua Escobar
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - C Junghanss
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
15
|
Wang S, Guo W, Wan X, Teng Y, Zhou X, Bai O. Exploring the effect of chidamide on blastic plasmacytoid dendritic cell neoplasm: a case report and literature review. Ther Clin Risk Manag 2018; 14:47-51. [PMID: 29379294 PMCID: PMC5757208 DOI: 10.2147/tcrm.s156204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive hematopoietic malignancy mainly affecting elderly patients. It is highly responsive to chemotherapy, but the median event-free survival is very short and has a high rate of relapse even after performing allogeneic stem cell transplantation; thus, the discovery of novel agents for the treatment of BPDCN is urgent. Chidamide is a new oral isotype-selective histone deacetylase inhibitor (HDACi). It is proved to exert a well-characterized anticancer property in a wide range of hematological malignancies, especially lymphoma. Here, we report a 41-year-old man who used oral chidamide 30 mg twice per week for maintenance therapy after receiving complete remission. For the first time in this field, we had explored the efficiency of chidamide in the treatment of BPDCN and tried to give more choices to the therapy of this disease.
Collapse
Affiliation(s)
| | | | | | - Yongliang Teng
- Department of Pathology, The First Hospital of Jilin University, Changchun
| | - Xiaoge Zhou
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | | |
Collapse
|