1
|
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
|
2
|
Kong FC, Qi L, Zhou YL, Yu M, Huang WF, Li F. Chidamide, Decitabine, Cytarabine, Aclarubicin, and Granulocyte Colony-stimulating Factor Therapy for Patients with Relapsed/Refractory Acute Myeloid Leukemia: A Retrospective Study from a Single-Center. Curr Med Sci 2023; 43:1151-1161. [PMID: 38057538 DOI: 10.1007/s11596-023-2805-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 09/19/2023] [Indexed: 12/08/2023]
Abstract
OBJECTIVE Preclinical evidence and clinical trials have suggested synergistic effects of epigenetic modifiers in combination with cytotoxic agents for the treatment of leukemia. However, their efficacy in patients with relapsed/refractory acute myeloid leukemia (R/R AML) remains unclear. METHODS Clinical data of R/R AML patients who received a CDCAG regimen (chidamide, decitabine, cytarabine, aclarubicin, and granulocyte colony-stimulating factor) from July 1, 2018 to October 31, 2021 at our center were retrospectively assessed, and the safety and efficacy of the CDCAG regimen were evaluated. Patients were followed up until November 30, 2021, with a median follow-up of 21.6 months (95% CI: 10.0-33.2 months). RESULTS A total of 67 patients were enrolled. Two patients died within 3 weeks after the initiation, and therefore only 65 patients underwent the assement for clinical response and survival. It was found that 56.9% patients achieved complete remission with a median overall survival (OS) of 9.6 months. The median OS of responders was 25.9 months, while that of non-responders was 5.0 months (P<0.0001). Patients with gene mutations had a superior overall response rate (ORR) (80.4% vs. 45.5%, P=0.043) compared to those without gene mutations. The presence of DNA methyltransferase 3 A (DNMT3A), ten-eleven translocation-2 (TET2), and isocitrate dehydrogenase 1/2 (IDH1/2) mutations did not affect the response rate (88.2% vs. 68.9%, P=0.220) and reflected a better OS (not attained vs. 9.0 months, P=0.05). The most common non-hematologic adverse events were pulmonary infection (73.1%), followed by febrile neutropenia (23.9%) and sepsis (19.4%). CONCLUSIONS The CDCAG regimen was effective and well-tolerated in R/R AML patients, increasing the potential for allogeneic hematopoietic stem cell transplantation. Moreover, patients with DNMT3A, TET2, and IDH1/2 mutations might benefit from this regimen.
Collapse
Affiliation(s)
- Fan-Cong Kong
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Clinical Research Center for Hematologic Diseases, Nanchang, 330006, China
| | - Ling Qi
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Clinical Research Center for Hematologic Diseases, Nanchang, 330006, China
| | - Yu-Lan Zhou
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Clinical Research Center for Hematologic Diseases, Nanchang, 330006, China
| | - Min Yu
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Clinical Research Center for Hematologic Diseases, Nanchang, 330006, China
| | - Wen-Feng Huang
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fei Li
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Jiangxi Clinical Research Center for Hematologic Diseases, Nanchang, 330006, China.
| |
Collapse
|
3
|
Wei Y, Wang L, Zhu C, Li H, Bo J, Zhang R, Lu N, Wu Y, Gao X, Dou L, Liu D, Gao C. A phase II study of chidamide, cytarabine, aclarubicin, granulocyte colony-stimulating factor, and donor lymphocyte infusion for relapsed acute myeloid leukemia and myelodysplastic syndrome after allogeneic hematopoietic stem cell transplantation. Med Oncol 2023; 40:77. [PMID: 36625951 PMCID: PMC9832090 DOI: 10.1007/s12032-022-01911-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/22/2022] [Indexed: 01/11/2023]
Abstract
Chemotherapy followed by donor lymphocyte infusion (DLI) is a promising treatment for relapsed acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the best strategy for administering this therapy is still unclear. This study sought to explore the efficacy and safety of chidamide and CAG (cytarabine, aclarubicin, and granulocyte colony-stimulating factor) (CCAG) regimen followed by DLI in relapsed AML/MDS after allo-HSCT. This was a single-arm, phase II trial in patients with relapsed AML/MDS after allo-HSCT. CCAG regimen followed by DLI was given according to the inclusion and exclusion criteria. Twenty adult patients were enrolled. The median follow-up time was 12 months. The complete remission (CR) rate was 45% and the partial remission (PR) rate was 5%. The 1-year overall survival (OS) was 56.7% (95% confidence interval (95% CI), 31.6-75.6%), and the median OS was 19 months. The 1-year relapse-free survival (RFS) was 83.3% (95% CI, 27.3-97.5%). Patients relapsing more than 6 months after HSCT and achieving CR/PR after CCAG plus DLI regimen attained significantly higher survival rates. The cumulative incidence of grade III-IV acute graft-versus-host disease (aGVHD) was 9.4%. There was no treatment-related mortality (TRM). These data suggest that CCAG plus DLI regimen is safe and induces durable remission and superior survival in patients with relapsed AML/MDS after allo-HSCT. Trial registration number: ChiCTR.org identifier: ChiCTR1800017740 and date of registration: August 12, 2018.
Collapse
Affiliation(s)
- Yan Wei
- Medical School of Chinese PLA, Beijing, China
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Lijun Wang
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Chengying Zhu
- School of Medicine, Nankai University, Tianjin, China
| | - Honghua Li
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Jian Bo
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Ran Zhang
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Ning Lu
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Yongli Wu
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Xiaoning Gao
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Liping Dou
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Daihong Liu
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Chunji Gao
- Department of Hematology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
4
|
Luo C, Yu T, Young KH, Yu L. HDAC inhibitor chidamide synergizes with venetoclax to inhibit the growth of diffuse large B-cell lymphoma via down-regulation of MYC, BCL2, and TP53 expression. J Zhejiang Univ Sci B 2022; 23:666-681. [PMID: 35953760 DOI: 10.1631/jzus.b2200016] [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] [Indexed: 11/11/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive type of non-Hodgkin's lymphoma. A total of 10%‒15% of DLBCL cases are associated with myelocytomatosis viral oncogene homolog(MYC) and/or B-cell lymphoma-2 (BCL2) translocation or amplification. BCL2 inhibitors have potent anti-tumor effects in DLBCL; however, resistance can be acquired through up-regulation of alternative anti-apoptotic proteins. The histone deacetylase (HDAC) inhibitor chidamide can induce BIM expression, leading to apoptosis of lymphoma cells with good efficacy in refractory recurrent DLBCL. In this study, the synergistic mechanism of chidamide and venetoclax in DLBCL was determined through in vitro and in vivo models. We found that combination therapy significantly reduced the protein levels of MYC, TP53, and BCL2 in activated apoptotic-related pathways in DLBCL cells by increasing BIM levels and inducing cell apoptosis. Moreover, combination therapy regulated expression of multiple transcriptomes in DLBCL cells, involving apoptosis, cell cycle, phosphorylation, and other biological processes, and significantly inhibited tumor growth in DLBCL-bearing xenograft mice. Taken together, these findings verify the in vivo therapeutic potential of chidamide and venetoclax combination therapy in DLBCL, warranting pre-clinical trials for patients with DLBCL.
Collapse
Affiliation(s)
- Cancan Luo
- Department of Hematology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Province Key Laboratory of Hematology, Nanchang 330006, China
| | - Tiantian Yu
- Department of Hematology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Province Key Laboratory of Hematology, Nanchang 330006, China
| | - Ken H Young
- Department of Hematopathology, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Li Yu
- Department of Hematology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China. .,Jiangxi Province Key Laboratory of Hematology, Nanchang 330006, China.
| |
Collapse
|
5
|
Li J, Liao D, Wang F, Wang Z, Li Y, Xiong Y, Niu T. RIPK1 inhibition enhances the therapeutic efficacy of chidamide in FLT3-ITD positive AML, both in vitro and in vivo. Leuk Lymphoma 2021; 63:1167-1179. [PMID: 34865571 DOI: 10.1080/10428194.2021.2010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Acute myeloid leukemia (AML) with FLT3-ITD mutation accounts for a large proportion of relapsed/refractory AML with poor prognosis. RIPK1 is a known key regulator of necroptosis and RIPK1 inhibition shows anti-AML effects in vitro. Chidamide is a histone deacetylase inhibitor (HDACi) with proven ability to induce apoptosis in FLT3-ITD positive AML cells. In the present study, we evaluated the effects of the combination of 22b, a novel RIPK1 inhibitor, and chidamide on proliferation and apoptosis in FLT3-ITD positive AML cell lines and primary cells. The results showed that 22b could significantly enhance the anti-leukemia effect of low-dose chidamide both on cell lines and primary cells. In a subcutaneous xenograft AML model, the combination of 22b and chidamide exhibited obviously elevated anti-tumor activity. In conclusion, our results support that the combination of RIPK1 inhibitor 22b and chidamide may be a novel therapeutic avenue for FLT3-ITD positive AML patients.
Collapse
Affiliation(s)
- Jun Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Liao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hematology, The Third Hospital of Mianyang, Mianyang, China
| | - Fujue Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hematology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhongwang Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yueshan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Xiong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Zhao L, Lv C, Sun L, Li Q, Wang Y, Wu M, Wang Y, Guo Z, Bian S, Kong D, Lin L, Wang Y, Zhou J, Li Y. Histone deacetylase inhibitor chidamide regulates the Wnt/β-catenin pathway by MYCN/DKK3 in B-ALL. Invest New Drugs 2021; 39:961-970. [PMID: 33566253 DOI: 10.1007/s10637-021-01079-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/31/2021] [Indexed: 10/25/2022]
Abstract
Our previous studies revealed that MYCN downregulates the expression of DKK3, activates the Wnt/β-catenin signalling pathway at the transcriptional level, and thereby promotes the development of B cell acute lymphocytic leukaemia (B-ALL) but does not affect the methylation of the DKK3 promoter. Some studies have shown that MYCN is associated with histone acetylation. We speculate that histone deacetylase inhibitors (HDACis) can inhibit the Wnt/β-catenin signalling pathway by inhibiting MYCN and increasing the expression of DKK3. Based on previous experiments, we tested this hypothesis by analysing the changes in MYCN, DKK3 and the Wnt/β-catenin signalling pathways in B-ALL cells after treatment with the selective HDACi chidamide. The in vitro and in vivo experiments confirmed that chidamide inhibited the expression of MYCN and increased the expression of DKK3 by inhibiting the activity of histone deacetylase, and these effects resulted in inhibition of the Wnt/β-catenin signalling pathway and the proliferation of B-ALL cells. These findings indicate that chidamide might be used alone or in combination with other chemotherapy regimens for patients with B-ALL and thus provide a new approach to the treatment of B-ALL.
Collapse
Affiliation(s)
- Linlin Zhao
- Department of Blood Transfusion, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chengfang Lv
- Department of Hematology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Lili Sun
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qi Li
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuhuang Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Min Wu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuying Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhibo Guo
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Sicheng Bian
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Desheng Kong
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Leilei Lin
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yu Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jin Zhou
- Department of Hematology, Southern University of Science and Technology Hospital, Shenzhen, China.
| | - Yinghua Li
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
7
|
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: 0.2] [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
|
8
|
Bioinformatics analysis of the network of histone H3 lysine 9 trimethylation in acute myeloid leukaemia. Oncol Rep 2020; 44:543-554. [PMID: 32468066 PMCID: PMC7336454 DOI: 10.3892/or.2020.7627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Changes in histone H3 lysine 9 trimethylation (H3K9me3) may be related to the development of drug-resistant acute myeloid leukaemia (AML); insights into the network of H3K9me3 may improve patient prognosis. Patient data were derived from the Gene Expression Omnibus (GEO) database and data from AML cells treated with chidamide, a novel benzamide chemical class of histone deacetylase inhibitor (HDACi), in vitro were derived from ChIP-seq. Patients and AML cell data were analysed using GEO2R, GOseq, KOBAS, the STRING database and Cytoscape 3.5.1. We identified several genes related to the upregulation or downregulation of H3K9me3 in AML patients; some of these genes were related to apoptosis, autophagy, and the pathway of cell longevity. AML cells treated with chidamide in vitro showed the same gene changes. The protein interactions in the network did not have significantly more interactions than expected, suggesting the need for more research to identify these interactions. One compelling result from the protein interaction study was that sirtuin 1 (SIRT1) may have an indirect interaction with lysine-specific demethylase 4A (KDM4A). These results help explain alterations of H3K9me3 in AML that may direct further studies aimed at improving patient prognosis. These results may also provide a basis for chidamide as a treatment strategy for AML patients in the future.
Collapse
|
9
|
Huang H, Wenbing Y, Dong A, He Z, Yao R, Guo W. Chidamide Enhances the Cytotoxicity of Cytarabine and Sorafenib in Acute Myeloid Leukemia Cells by Modulating H3K9me3 and Autophagy Levels. Front Oncol 2019; 9:1276. [PMID: 31850196 PMCID: PMC6901797 DOI: 10.3389/fonc.2019.01276] [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: 03/11/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
Previous studies showed that chidamide enhances the cytotoxicity of drugs in acute myeloid leukemia (AML) cells. Therefore, we examined whether chidamide enhanced the cytotoxicity of drugs in AML cells by affecting H3K9me3 and autophagy levels. AML cells (THP-1 and MV4-11 cells) were treated with chidamide, cytarabine (Ara-c), or sorafenib alone or in combination. Cell proliferation and survival rates were analyzed by MTT, flow cytometry, and Western blotting assays. The results showed that a low dose of chidamide enhanced the cytotoxicity of Ara-c or sorafenib in AML cells, decreasing proliferation and increasing apoptosis. H3K9me3 levels as assessed by Western blotting were upregulated by chidamide treatment. Chromatin immunoprecipitation sequencing, which was used to investigate potential signaling pathways, indicated that the autophagy pathway might play a role in the effects of chidamide. The level of autophagy induced in AML cells upon treatment with Ara-c or sorafenib was inhibited by chidamide, and autophagy markers (LC3, P62) were tested by Western blotting. SIRT1 messenger RNA (mRNA) and protein levels were lower in AML cells treated with Ara-c or sorafenib in combination with chidamide than those in cells treated with these drugs alone. Additionally, the Integrative Genomics Viewer results indicate that the H3K9me3 changes were related to SIRT1-binding sites. Together, these results show that chidamide enhances the cytotoxicity of two chemotherapy drugs in AML cells by increasing the H3K9me3 level and inhibiting autophagy via decreasing the expression of SIRT1. Chidamide may be a potential treatment strategy for AML in the future, especially for refractory AML patients.
Collapse
Affiliation(s)
- He Huang
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yang Wenbing
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Aishu Dong
- Department of Emergency, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhewei He
- The Second Clinic College of Wenzhou Medical University, Wenzhou, China
| | - Rongxing Yao
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjian Guo
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
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: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
MESH Headings
- Aminopyridines/administration & dosage
- Aminopyridines/pharmacology
- Animals
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Benzamides/administration & dosage
- Benzamides/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- DNA Repair/drug effects
- Drug Synergism
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Histone-Lysine N-Methyltransferase/genetics
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Membrane Potential, Mitochondrial/drug effects
- Mice
- Myeloid-Lymphoid Leukemia Protein/genetics
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Proto-Oncogene Proteins/genetics
- Small Molecule Libraries/administration & dosage
- Small Molecule Libraries/pharmacology
- Xenograft Model Antitumor Assays
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
|
11
|
Wu RJ, Zheng RJ, Huang YQ, Ma XD. [Chidamide combined with chemotherapy for treatment of therapy-related acute myeloid leukemia secondary to peripheral T-cell lymphoma: a case report and literatures review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:685-687. [PMID: 31495139 PMCID: PMC7342885 DOI: 10.3760/cma.j.issn.0253-2727.2019.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Indexed: 01/15/2023]
Affiliation(s)
- R J Wu
- Department of Hematology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | | | | | | |
Collapse
|
12
|
HDAC1,2 Knock-Out and HDACi Induced Cell Apoptosis in Imatinib-Resistant K562 Cells. Int J Mol Sci 2019; 20:ijms20092271. [PMID: 31071955 PMCID: PMC6539538 DOI: 10.3390/ijms20092271] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
Since imatinib (Glivec or Gleevec) has been used to target the BCR-ABL fusion protein, chronic myeloid leukemia (CML) has become a manageable chronic disease with long-term survival. However, 15%–20% of CML patients ultimately develop resistance to imatinib and then progress to an accelerated phase and eventually to a blast crisis, limiting treatment options and resulting in a poor survival rate. Thus, we investigated whether histone deacetylase inhibitors (HDACis) could be used as a potential anticancer therapy for imatinib-resistant CML (IR-CML) patients. By applying a noninvasive apoptosis detection sensor (NIADS), we found that panobinostat significantly enhanced cell apoptosis in K562 cells. A further investigation showed that panobinostat induced apoptosis in both K562 and imatinib-resistant K562 (IR-K562) cells mainly via H3 and H4 histone acetylation, whereas panobinostat targeted cancer stem cells (CSCs) in IR-K562 cells. Using CRISPR/Cas9 genomic editing, we found that HDAC1 and HDAC2 knockout cells significantly induced cell apoptosis, indicating that the regulation of HDAC1 and HDAC2 is extremely important in maintaining K562 cell survival. All information in this study indicates that regulating HDAC activity provides therapeutic benefits against CML and IR-CML in the clinic.
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: 22] [Impact Index Per Article: 3.7] [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
|
Spinello I, Saulle E, Quaranta MT, Pasquini L, Pelosi E, Castelli G, Ottone T, Voso MT, Testa U, Labbaye C. The small-molecule compound AC-73 targeting CD147 inhibits leukemic cell proliferation, induces autophagy and increases the chemotherapeutic sensitivity of acute myeloid leukemia cells. Haematologica 2018; 104:973-985. [PMID: 30467201 PMCID: PMC6518905 DOI: 10.3324/haematol.2018.199661] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022] Open
Abstract
CD147 is a transmembrane glycoprotein with multiple functions in human healthy tissues and diseases, in particular in cancer. Overexpression of CD147 correlates with biological functions that promote tumor progression and confers resistance to chemotherapeutic drugs. In contrast to solid tumors, the role of CD147 has not been extensively studied in leukemia. Understanding whether CD147 represents a new hematologic target and whether its inhibitor AC-73 may be used in leukemia therapy may reveal an alternative treatment strategy in patients with acute myeloid leukemia (AML). We analyzed CD147 expression and function in hematopoietic progenitor cells from normal cord blood, in several leukemic cell lines and in primary leukemic blasts obtained from patients with AML. We investigated the effects of AC-73, used alone or in combination with arabinosylcytosine (Ara-C) and arsenic trioxide (ATO), on leukemic cell proliferation. We demonstrated that CD147 overexpression promotes leukemic cell proliferation. We showed that AC-73 exhibits a potent growth inhibitory activity in leukemic cells, by inhibiting the ERK/STAT3 activation pathway and activating autophagy. We demonstrated that AC-73 exerts an anti-proliferative effect additive to chemotherapy by enhancing leukemic cell sensitivity to Ara-C-induced cytotoxicity or to ATO-induced autophagy. We also reported CD147 expression in the fraction of leukemic blasts expressing CD371, a marker of leukemic stem cells. Altogether, our study indicates CD147 as a novel potential target in the treatment of AML and AC-73 as an anti-proliferative drug and an inducer of autophagy in leukemic cells to use in combination with chemotherapeutic agents.
Collapse
Affiliation(s)
- Isabella Spinello
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome
| | - Ernestina Saulle
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome
| | - Maria Teresa Quaranta
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome
| | | | - Elvira Pelosi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità
| | - Germana Castelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Ugo Testa
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità
| | - Catherine Labbaye
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome
| |
Collapse
|
15
|
Chen L, Mi RH, Zhu ST, Yu P, Wei XD. [Therapeutic effect of chidamide and decitabine in combiantion with CHAG priming regimen for 8 patients with relapsed/refractory acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:602-604. [PMID: 30122025 PMCID: PMC7342203 DOI: 10.3760/cma.j.issn.0253-2727.2018.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Indexed: 12/14/2022]
|
16
|
Lu Z, Xu H, Yu X, Wang Y, Huang L, Jin X, Sui D. 20(S)-Protopanaxadiol induces apoptosis in human hepatoblastoma HepG2 cells by downregulating the protein kinase B signaling pathway. Exp Ther Med 2018; 15:1277-1284. [PMID: 29434714 PMCID: PMC5776618 DOI: 10.3892/etm.2017.5594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/26/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatoblastoma is the most common primary liver tumor for children aged <5 years old. 20(S)-Protopanaxadiol (PPD) is a ginsenoside extracted from Pananx quinquefolium L., which inhibits tumor growth in several cancer cell lines. The purpose of the present study was to assess the anticancer activities of 20(S)-PPD in human hepatoblastoma HepG2 cells. The cytotoxicity of 20(S)-PPD on HepG2 cells was evaluated using an MTT assay. Apoptosis was detected using DAPI staining and flow cytometry. The expression of apoptosis-associated proteins was identified by western blotting. The results demonstrated that 20(S)-PPD inhibited the viability of HepG2 cell in a dose and time-dependent manner. The IC50 values were 81.35, 73.5, 48.79 µM at 24, 48 and 72 h, respectively. Topical morphological changes of apoptotic body formation following 20(S)-PPD treatment were detected by DAPI staining. The percentage of Annexin V-fluoroscein isothyiocyanate positive cells were 3.73, 17.61, 23.44 and 65.43% in HepG2 cells treated with 0, 40, 50 and 60 µM of 20(S)-PPD, respectively. Furthermore, 20(S)-PPD upregulated the expression of Bax and downregulated the expression of Bcl-2 and also activated caspases-3 and −9, and Poly [ADP-ribose] polymerase cleavage. In addition, 20(S)-PPD inhibited the phosphorylation of protein kinase B (Akt; Ser473). The results indicate that 20(S)-PPD inhibits the viability of HepG2 cells and induces apoptosis in HepG2 cells by inhibiting the phosphoinositide-3-kinase/Akt pathway.
Collapse
Affiliation(s)
- Zeyuan Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuchen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Long Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xin Jin
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|