1
|
Yin T, Liu Y, Li C, Feng X, Lin Y, Qu Z. Characteristic analysis of adverse reactions of histone deacetylase inhibitors based on WHO-VigiAccess. Front Pharmacol 2025; 16:1563797. [PMID: 40170733 PMCID: PMC11959061 DOI: 10.3389/fphar.2025.1563797] [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: 01/20/2025] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Background This study assessed the adverse drug reactions (ADRs) associated with HDAC inhibitors using the VigiAccess database maintained by the World Health Organization (WHO). Furthermore, it compared the ADR profiles of three different drugs to identify the one with the lowest individualized risk for patients. Materials and methods Data on adverse events of HDAC Inhibitors was retrieved from WHO-VigiAccess on 6 January 2025. We obtained data on age, gender, reporting year, and continent. Descriptive data related were calculated using Excel 2021. In this study, we used Excel software to analyze the characteristics of those who were harmed due to adverse reactions. For each drug, the reporting rate of adverse reactions was calculated by dividing the number of adverse reaction symptoms of this drug by the total number of adverse reaction reports. We listed the top 20 most frequent adverse reaction symptoms as common adverse reactions. By counting the frequency and proportion of these common adverse reactions, we conducted a comparative analysis of the adverse reaction situations of different drugs and classified them according to different types. Result The WHO-VigiAccess database received 796, 1254, and 1658 ADR reports for Chidamide, Romidepsin, and Vorinostat respectively by 2024, with a total of 3,708. Gender distribution was relatively balanced (male:female ratio 0.81:1), and the 45-64 age group had the highest reporting rates, mostly from the Americas. Chidamide had higher rates in certain disorders, Romidepsin in others, and Vorinostat in specific ones. Common ADRs included thrombocytopenia etc., with some differences in rates among drugs. Serious ADR proportions were 0% for Chidamide, 2.27% for Romidepsin, and 1.02% for Vorinostat. 37 common signals were found, with Investigations having the most. Each drug had different ADR preferred terms (PTs) in renal/urinary and metabolism/nutrition disorders, with varying numbers of distinctive symptoms. Conclusion Current comparative observational studies of these inhibitors indicate that there are both common and specific adverse reactions reported in the ADR data received by the WHO for these medications. Clinicians should enhance the rational use of these drugs by considering the characteristics of the reported ADRs.
Collapse
Affiliation(s)
- Tongnan Yin
- Central Laboratory, Nanyang Central Hospital, Nanyang, China
| | - Yuyu Liu
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenwen Li
- Department of Dermatology, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Xinran Feng
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Qu
- Department of Oncology, Nanyang Central Hospital, Nanyang, China
| |
Collapse
|
2
|
He Y, Chen D, Yi Y, Zeng S, Liu S, Li P, Xie H, Yu P, Jiang G, Liu H. Histone Deacetylase Inhibitor Sensitizes ERCC1-High Non-small-Cell Lung Cancer Cells to Cisplatin via Regulating miR-149. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:448-459. [PMID: 32478168 PMCID: PMC7251316 DOI: 10.1016/j.omto.2020.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
Resistance to platinum-based chemotherapy becomes a major obstacle in non-small-cell lung cancer (NSCLC) treatment. Overexpression of the excision repair cross-complementing 1 (ERCC1) gene is reported to negatively influence the effectiveness of cisplatin-based therapy for NSCLC cells. In this study, we confirm that high ERCC1 expression correlates with cisplatin resistance in NSCLC cells. Importantly, histone deacetylase inhibitors (HDACis) re-sensitize ERCC1-high NSCLC cells to cisplatin both in vitro and in vivo. Mechanistically, the HDACi induces the expression of miR-149 by acetylation and activation of E2F1, which directly targets ERCC1 and inhibits ERCC1 expression. Inhibition of miR-149 reverses the promotion effect of HDACis on cisplatin-induced DNA damage and cell apoptosis in ERCC1-high NSCLC cells. In conclusion, this study reveals a novel mechanism by which HDACis re-sensitizes ERCC1-high NSCLC cells to cisplatin via regulation of the E2F1/miR-149/ERCC1 axis, and we propose that combination of HDACis and cisplatin might hold promise to be a more effective therapeutic paradigm for ERCC1-high NSCLCs.
Collapse
Affiliation(s)
- Yuwen He
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Danyang Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Yanmei Yi
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shanshan Zeng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Shuang Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Pan Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Hui Xie
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Pengjiu Yu
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 528000, Guangdong, China
- Corresponding author: Guanmin Jiang, Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 528000, Guangdong, China.
| | - Hao Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
- Corresponding author: Hao Liu, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” No. 78 Engzhigang Road, Guangzhou 510095, Guangdong, China.
| |
Collapse
|
3
|
Choi SA, Kwak PA, Park CK, Wang KC, Phi JH, Lee JY, Lee CS, Lee JH, Kim SK. A novel histone deacetylase inhibitor, CKD5, has potent anti-cancer effects in glioblastoma. Oncotarget 2018; 8:9123-9133. [PMID: 27852054 PMCID: PMC5354719 DOI: 10.18632/oncotarget.13265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 11/01/2016] [Indexed: 01/09/2023] Open
Abstract
There have been extensive efforts to improve the outcome of glioblastoma, but the prognosis of this disease has not been significantly altered to date. Histone deacetylase inhibitors (HDACIs) have been evaluated as promising anti-cancer drugs and regulate cell growth, cell cycle arrest and apoptosis in glioblastoma. Here, we demonstrated the therapeutic efficacy of a novel pan-HDACI, 7-ureido-N-hydroxyheptanamide derivative (CKD5), compared with traditional pan-HDACIs, such as suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA), in vitro and in vivo. Compared with SAHA and TSA, CKD5 had improved cytotoxic effects and induced apoptosis, anti-proliferative activity and cell cycle arrest at G2/M phase. Furthermore, CKD5 significantly reduced tumor volume and prolonged the survival in vivo compared with TSA, suggesting improved anti-cancer efficacy among HDACIs. Our results demonstrate that the novel HDACI CKD5 is a promising therapeutic candidate for glioblastoma.
Collapse
Affiliation(s)
- Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea.,Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, Korea
| | - Pil Ae Kwak
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea.,Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
| | - Kyu-Chang Wang
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea.,Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
| | - Ji Hoon Phi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea.,Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, Korea.,Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
| | - Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea.,Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea.,Department of Anatomy, Seoul National University Hospital, Seoul, Korea
| | - Chang Sik Lee
- Chong Kun Dang Research Institute, CKD Pharmaceuticals, Gyeonggi-do, Korea
| | - Ju-Hee Lee
- Chong Kun Dang Research Institute, CKD Pharmaceuticals, Gyeonggi-do, Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea.,Adolescent Cancer Center, Seoul National University Cancer Hospital, Seoul, Korea.,Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
4
|
Whittle JR, Desai J. Histone deacetylase inhibitors in cancer: What have we learned? Cancer 2014; 121:1164-7. [DOI: 10.1002/cncr.29177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 11/10/2014] [Indexed: 12/11/2022]
Affiliation(s)
- James R. Whittle
- Department of Medical Oncology; Royal Melbourne Hospital; Parkville Victoria Australia
| | - Jayesh Desai
- Department of Medical Oncology; Royal Melbourne Hospital; Parkville Victoria Australia
- The Walter and Eliza Hall Institute of Medical Research; Parkville Victoria Australia
- Peter MacCallum Cancer Center, Cancer Medicine; East Melbourne Victoria Australia
| |
Collapse
|
5
|
Locatelli SL, Cleris L, Stirparo GG, Tartari S, Saba E, Pierdominici M, Malorni W, Carbone A, Anichini A, Carlo-Stella C. BIM upregulation and ROS-dependent necroptosis mediate the antitumor effects of the HDACi Givinostat and Sorafenib in Hodgkin lymphoma cell line xenografts. Leukemia 2014; 28:1861-71. [PMID: 24561519 DOI: 10.1038/leu.2014.81] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 01/07/2023]
Abstract
Relapsed/refractory Hodgkin's lymphoma (HL) is an unmet medical need requiring new therapeutic options. Interactions between the histone deacetylase inhibitor Givinostat and the RAF/MEK/ERK inhibitor Sorafenib were examined in HDLM-2 and L-540 HL cell lines. Exposure to Givinostat/Sorafenib induced a synergistic inhibition of cell growth (range, 70-80%) and a marked increase in cell death (up to 96%) due to increased H3 and H4 acetylation and strong mitochondrial injury. Gene expression profiling indicated that the synergistic effects of Givinostat/Sorafenib treatment are associated with the modulation of cell cycle and cell death pathways. Exposure to Givinostat/Sorafenib resulted in sustained production of reactive oxygen species (ROS) and activation of necroptotic cell death. The necroptosis inhibitor Necrostatin-1 prevented Givinostat/Sorafenib-induced ROS production, mitochondrial injury, activation of BH3-only protein BIM and cell death. Knockdown experiments identified BIM as a key signaling molecule that mediates Givinostat/Sorafenib-induced oxidative death of HL cells. Furthermore, in vivo xenograft studies demonstrated a 50% reduction in tumor burden (P<0.0001), a 5- to 15-fold increase in BIM expression (P < 0.0001) and a fourfold increase in tumor necrosis in Givinostat/Sorafenib-treated animals compared with mice that received single agents. These results provide a rationale for exploring Givinostat/Sorafenib combination in relapsed/refractory HL.
Collapse
Affiliation(s)
- S L Locatelli
- 1] Department of Oncology and Hematology, Humanitas Cancer Center - Humanitas Clinical and Research Center, Milano, Italy [2] Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| | - L Cleris
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - G G Stirparo
- 1] Department of Oncology and Hematology, Humanitas Cancer Center - Humanitas Clinical and Research Center, Milano, Italy [2] Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| | - S Tartari
- Department of Oncology and Hematology, Humanitas Cancer Center - Humanitas Clinical and Research Center, Milano, Italy
| | - E Saba
- Department of Oncology and Hematology, Humanitas Cancer Center - Humanitas Clinical and Research Center, Milano, Italy
| | - M Pierdominici
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - W Malorni
- 1] Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Rome, Italy [2] Istituto San Raffaele Sulmona, Sulmona, Italy
| | - A Carbone
- Pathology Department, CRO Aviano, Aviano, Italy
| | - A Anichini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - C Carlo-Stella
- 1] Department of Oncology and Hematology, Humanitas Cancer Center - Humanitas Clinical and Research Center, Milano, Italy [2] Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| |
Collapse
|
6
|
Li B, Ye Z. Epigenetic alterations in osteosarcoma: promising targets. Mol Biol Rep 2014; 41:3303-15. [PMID: 24500341 DOI: 10.1007/s11033-014-3193-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 01/22/2014] [Indexed: 01/10/2023]
Abstract
Cancer is being reinterpreted due to recent discoveries related to epigenetic regulation during development, and the importance of epigenetic mechanisms in initiation and progression of cancer has been further highlighted by the recent explosion in medical information. Osteosarcoma is highly genetically unstable, and current therapeutic regimens are subject to chemoresistance and tumor relapse. Understanding the epigenetic mechanisms in the pathogenesis of osteosarcoma will provide novel avenues for cancer therapy. In this review, we examine the epigenetic alterations in gene expression in osteosarcoma, and discuss the utilization of epigenetic regulation therapy in treatment against osteosarcoma.
Collapse
Affiliation(s)
- Binghao Li
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310008, China
| | | |
Collapse
|
7
|
Bhatt S, Ashlock BM, Toomey NL, Diaz LA, Mesri EA, Lossos IS, Ramos JC. Efficacious proteasome/HDAC inhibitor combination therapy for primary effusion lymphoma. J Clin Invest 2013; 123:2616-28. [PMID: 23635777 DOI: 10.1172/jci64503] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 02/28/2013] [Indexed: 12/26/2022] Open
Abstract
Primary effusion lymphoma (PEL) is a rare form of aggressive B cell lymphoma caused by Kaposi's sarcoma-associated herpesvirus (KSHV). Current chemotherapy approaches result in dismal outcomes, and there is an urgent need for new PEL therapies. Previously, we established, in a direct xenograft model of PEL-bearing immune-compromised mice, that treatment with the proteasome inhibitor, bortezomib (Btz), increased survival relative to that after treatment with doxorubicin. Herein, we demonstrate that the combination of Btz with the histone deacetylase (HDAC) inhibitor suberoylanilidehydroxamic acid (SAHA, also known as vorinostat) potently reactivates KSHV lytic replication and induces PEL cell death, resulting in significantly prolonged survival of PEL-bearing mice. Importantly, Btz blocked KSHV late lytic gene expression, terminally inhibiting the full lytic cascade and production of infectious virus in vivo. Btz treatment led to caspase activation and induced DNA damage, as evidenced by the accumulation of phosphorylated γH2AX and p53. The addition of SAHA to Btz treatment was synergistic, as SAHA induced early acetylation of p53 and reduced interaction with its negative regulator MDM2, augmenting the effects of Btz. The eradication of KSHV-infected PEL cells without increased viremia in mice provides a strong rationale for using the proteasome/HDAC inhibitor combination therapy in PEL.
Collapse
Affiliation(s)
- Shruti Bhatt
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | | | | | | | | |
Collapse
|
8
|
Zuch D, Giang AH, Shapovalov Y, Schwarz E, Rosier R, O'Keefe R, Eliseev RA. Targeting radioresistant osteosarcoma cells with parthenolide. J Cell Biochem 2012; 113:1282-91. [PMID: 22109788 DOI: 10.1002/jcb.24002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteosarcoma is a devastating tumor of bone, primarily affecting adolescents. Osteosarcoma tumors are notoriously radioresistant. Radioresistant cancers, including osteosarcoma, typically exhibit a considerable potential for relapse and development of metastases following treatment. Relapse and metastatic potential can, in part, be due to a specific radioresistant subpopulation of cells with stem-like characteristics, cancer stem cells, which maintain the capacity to regenerate entire tumors. In the current study, we have investigated whether in vitro treatments with parthenolide, a naturally occurring small molecule that interferes with NF-κB signaling and has various other effects, will re-sensitize cancer stem cells and the entire cell population to radiotherapy in osteosarcoma. Our results indicate that parthenolide and ionizing radiation synergistically induce cell death in LM7 osteosarcoma cells. Importantly, the combination treatment results in a significant reduction in the viability of both the overall population of osteosarcoma cells and the cancer stem cell subpopulation. This effect is dependent on the ability of parthenolide to induce oxidative stress. Therefore, as a supplement to current multimodal therapy, parthenolide may sensitize osteosarcoma tumors to radiation and greatly reduce the prevalence of relapse and metastatic progression.
Collapse
Affiliation(s)
- Daniel Zuch
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, 575 Elmwood Ave., Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Thayanithy V, Park C, Sarver AL, Kartha RV, Korpela DM, Graef AJ, Steer CJ, Modiano JF, Subramanian S. Combinatorial treatment of DNA and chromatin-modifying drugs cause cell death in human and canine osteosarcoma cell lines. PLoS One 2012; 7:e43720. [PMID: 22957032 PMCID: PMC3434163 DOI: 10.1371/journal.pone.0043720] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 07/23/2012] [Indexed: 11/18/2022] Open
Abstract
Downregulation of microRNAs (miRNAs) at the 14q32 locus stabilizes the expression of cMYC, thus significantly contributing to osteosarcoma (OS) pathobiology. Here, we show that downregulation of 14q32 miRNAs is epigenetically regulated. The predicted promoter regions of miRNA clusters at 14q32 locus showed no recurrent patterns of differential methylation, but Saos2 cells showed elevated histone deacetylase (HDAC) activity. Treatment with 4-phenylbutyrate increased acetylation of histones associated with 14q32 miRNAs, but interestingly, robust restoration of 14q32 miRNA expression, attenuation of cMYC expression, and induction of apoptosis required concomitant treatment with 5-Azacytidine, an inhibitor of DNA methylation. These events were associated with genome-wide gene expression changes including induction of pro-apoptotic genes and downregulation of cell cycle genes. Comparable effects were achieved in human and canine OS cells using the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA/Vorinostat) and the DNA methylation inhibitor Zebularine (Zeb), with significantly more pronounced cytotoxicity in cells whose molecular phenotypes were indicative of aggressive biological behavior. These results suggested that the combination of these chromatin-modifying drugs may be a useful adjuvant in the treatment of rapidly progressive OS.
Collapse
Affiliation(s)
- Venugopal Thayanithy
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - ChangWon Park
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aaron L. Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Reena V. Kartha
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Derek M. Korpela
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Ashley J. Graef
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Clifford J. Steer
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jaime F. Modiano
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Subbaya Subramanian
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
10
|
A novel series of l-2-benzyloxycarbonylamino-8-(2-pyridyl)-disulfidyloctanoic acid derivatives as histone deacetylase inhibitors: design, synthesis and molecular modeling study. Eur J Med Chem 2012; 52:111-22. [PMID: 22465091 DOI: 10.1016/j.ejmech.2012.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 03/05/2012] [Accepted: 03/06/2012] [Indexed: 11/20/2022]
Abstract
Histone deacetylases inhibitors (HDACIs) have become an attractive class of anticancer agents. In order to find some novel potent HDACIs, we designed and synthesized a series of l-2-benzyloxycarbonylamino-8-(2-pyridyl)-disulfidyloctanoic acid derivatives. All compounds exhibited potent HDAC-inhibitory activity, and two of them had similar potency to TSA. The introduction of 2-amino-4-phenylthiazole or 9-methyleneoxy-fluorenyl group at the surface recognize domain of these HDACIs could greatly increase their HDAC-inhibitory activity. Molecular modeling studies indicated that coordination of the zinc ion by these inhibitors, and formation of hydrogen bond and hydrophobic interaction between inhibitors and HDACs were essential for the HDAC-inhibitory activities of these inhibitors. Asp181, Asp269, Leu276 and Tyr308 in the active site of HDAC2 gave favorable contributions for binding with all compounds.
Collapse
|
11
|
Deb AA, Wilson SS, Rove KO, Kumar B, Koul S, Lim DD, Meacham RB, Koul HK. Potentiation of Mitomycin C Tumoricidal Activity for Transitional Cell Carcinoma by Histone Deacetylase Inhibitors In Vitro [RETRACTED]. J Urol 2011; 186:2426-33. [DOI: 10.1016/j.juro.2011.07.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Indexed: 02/08/2023]
Affiliation(s)
- Abdalla Ali Deb
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Shandra S. Wilson
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Kyle O. Rove
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Binod Kumar
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Sweaty Koul
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Douglas D. Lim
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Randall B. Meacham
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Hari K. Koul
- Program in Urosciences, Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
12
|
Abstract
Gamma-(γ)-herpes virus lymphomas comprise a heterogenous group of B-cell and T-cell neoplasms most commonly associated with Epstein-Barr virus and rarely human herpes virus-8 infection. Adult T-cell leukemia/lymphoma (ATLL) is a unique disease entity caused by the human T-cell lymphotrophic virus, type 1 (HTLV-I), the only retrovirus known to cause cancer in humans. Viral lymphomas behave aggressively and disproportionally affect immunocompromised individuals and those living in underdeveloped regions. These diseases are often difficult to treat with conventional approaches. Despite recent advancements using cytotoxic, lymphoma-specific, and adoptive therapies, the long-term outcome of patients with γ-herpesvirus lymphomas occurring in severely immunocompromised patients and ATLL continues to be poor. Lytic-inducing therapies targeting NF-κB, and viral and tumor cell epigenetic mechanisms afford the advantage of exploiting the intrinsic presence of oncogenic viruses to eradicate infected tumor cells. In this review, viral-related lymphomas and newly emerging clinical approaches targeting viral latency are discussed.
Collapse
Affiliation(s)
- Juan Carlos Ramos
- Department of Medicine, Division of Oncology-Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Izidore S Lossos
- Department of Medicine, Division of Oncology-Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
- Department of Molecular and Cellular Pharmacology, University of Miami, Miami, FL
| |
Collapse
|
13
|
Atadja PW. HDAC inhibitors and cancer therapy. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2011; 67:175-95. [PMID: 21141730 DOI: 10.1007/978-3-7643-8989-5_9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Maintenance of normal cell growth and differentiation is highly dependent on coordinated and tight transcriptional regulation of genes. In cancer, genes encoding growth regulators are abnormally expressed. Particularly, silencing of tumor suppressor genes under the control of chromatin modifications is a major underlying cause of unregulated cellular proliferation and transformation. Thus mechanisms, which regulate chromatin structure and gene expression, have become attractive targets for anticancer therapy. Histone deacetylases are enzymes that modify chromatin structure and contribute to aberrant gene expression in cancer. Research over the past decade has led to the development of histone deacetylase inhibitors as anticancer agents. In addition to their effect on chromatin and epigenetic mechanisms, HDAC inhibitors also modify the acetylation state of a large number of cellular proteins involved in oncogenic processes, resulting in antitumor effects. The current monograph will review the role of histone deacetylases in protumorigenic mechanisms and the current developmental status and prospects for their inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Peter W Atadja
- Novartis Institute for Biomedical Research, 898 Halei Rd, Building 8, Shanghai, China.
| |
Collapse
|
14
|
Chang H, Rha SY, Jeung HC, Jung JJ, Kim TS, Kwon HJ, Kim BS, Chung HC. Identification of genes related to a synergistic effect of taxane and suberoylanilide hydroxamic acid combination treatment in gastric cancer cells. J Cancer Res Clin Oncol 2010; 136:1901-13. [PMID: 20217129 DOI: 10.1007/s00432-010-0849-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE We evaluated the cytotoxic effects of combining suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, with taxanes in human gastric cancer cell lines and assessed the pre-treatment difference of gene expression to identify genes that could potentially mediate the cytotoxic response. METHODS Gastric cancer cell lines were treated with SAHA and paclitaxel or docetaxel, and the synergistic interaction between the drugs was evaluated in vitro using the combination index (CI) method. We performed significance analysis of microarray (SAM) to identify chemosensitivity-related genes in gastric cancer cell lines that were concomitantly treated with SAHA and taxane. We generated a correlation matrix between gene expression and CI values to identify genes whose expression correlated with a combined effect of taxanes and SAHA. RESULTS Combination treatment with taxane and SAHA had a synergistic cytotoxic effect against taxane-resistant gastric cancer cells. We identified 49 chemosensitivity-related genes via SAM analysis. Among them, nine common genes (SLIT2, REEP2, EFEMP2, CDC42SE1, FSD1, POU1F1, ZNF79, ETNK1, and DOCK5) were extracted from the subsequent correlation matrix analysis. CONCLUSIONS The combination of taxane and SAHA could be efficacious for the treatment of gastric cancer. The genes that were related to the synergistic response to taxane and SAHA could serve as surrogate biomarkers to predict the therapeutic response in gastric cancer patients.
Collapse
Affiliation(s)
- Hyun Chang
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, #134 Shinchon-Dong, Seodaemoon-gu, Seoul 120-752, Korea
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Sha K, Winn LM. Characterization of valproic acid-initiated homologous recombination. ACTA ACUST UNITED AC 2010; 89:124-32. [PMID: 20437471 DOI: 10.1002/bdrb.20236] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Valproic acid (VPA) is a frequently used antiepileptic agent and known teratogen. Previous research suggests that inhibition of histone deacetylases (HDACs) may play a role in VPA-induced teratogenicity. We have also shown that VPA exposure leads to both an increase in reactive oxygen species (ROS) production and increased frequency of homologous recombination (HR). METHODS In the present study, we evaluated the role of HDAC inhibition in VPA-initiated HR to determine if HDAC inhibition could alter repair activity and/or cause DNA double-strand breaks (DSBs), which would then initiate repair. Histone acetylation status was assessed to determine if VPA exposure led to HDAC inhibition in CHO 33 cells. RESULTS Our results demonstrate that VPA (5 mM) exposure leads to increased acetylated histone H3 and H4 protein levels after 10 to 24 hr. Secondly, in our recombination assay where an artificial DNA DSB was induced in CHO 33 cells to assess repair activity, VPA exposure did not affect the repair activity of VPA-initiated HR. Subsequently, to determine if VPA could increase susceptibility to DNA DSBs, the number of gamma-H2AX foci was assessed using immunocytochemistry and results revealed an increase in gamma-H2AX foci after 10- to 24-hr exposure to VPA. CONCLUSIONS Although we demonstrated the protective effect of polyethylene glycol-catalase against VPA-induced HR and the generation of intracellular ROS within 24 hr, we did not observed an increase in DNA oxidation. These studies suggest that HDAC inhibition and ROS signaling may play roles in DNA maintenance and cell-cycle arrest in initiating DNA damage and repair.
Collapse
Affiliation(s)
- Kevin Sha
- Department of Pharmacology and Toxicology, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
16
|
Ascierto PA, Streicher HZ, Sznol M. Melanoma: a model for testing new agents in combination therapies. J Transl Med 2010; 8:38. [PMID: 20406483 PMCID: PMC2873374 DOI: 10.1186/1479-5876-8-38] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 04/20/2010] [Indexed: 12/15/2022] Open
Abstract
Treatment for both early and advanced melanoma has changed little since the introduction of interferon and IL-2 in the early 1990s. Recent data from trials testing targeted agents or immune modulators suggest the promise of new strategies to treat patients with advanced melanoma. These include a new generation of B-RAF inhibitors with greater selectivity for the mutant protein, c-Kit inhibitors, anti-angiogenesis agents, the immune modulators anti-CTLA4, anti-PD-1, and anti-CD40, and adoptive cellular therapies. The high success rate of mutant B-RAF and c-Kit inhibitors relies on the selection of patients with corresponding mutations. However, although response rates with small molecule inhibitors are high, most are not durable. Moreover, for a large subset of patients, reliable predictive biomarkers especially for immunologic modulators have not yet been identified. Progress may also depend on identifying additional molecular targets, which in turn depends upon a better understanding of the mechanisms leading to response or resistance. More challenging but equally important will be understanding how to optimize the treatment of individual patients using these active agents sequentially or in combination with each other, with other experimental treatment, or with traditional anticancer modalities such as chemotherapy, radiation, or surgery. Compared to the standard approach of developing new single agents for licensing in advanced disease, the identification and validation of patient specific and multi-modality treatments will require increased involvement by several stakeholders in designing trials aimed at identifying, even in early stages of drug development, the most effective way to use molecularly guided approaches to treat tumors as they evolve over time.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Medical Oncology and Innovative Therapy, National Tumor Institute, Naples, Italy.
| | | | | |
Collapse
|
17
|
The histone deacetylase inhibitor valproic acid sensitizes human and canine osteosarcoma to doxorubicin. Cancer Chemother Pharmacol 2010; 67:83-92. [PMID: 20306194 DOI: 10.1007/s00280-010-1287-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 02/12/2010] [Indexed: 12/20/2022]
Abstract
PURPOSE Osteosarcoma (OS) remains an incurable and ultimately fatal disease in many patients, and novel forms of therapy are needed. Improved models of OS that more closely mimic human disease would provide more robust information regarding the utility of novel therapies. Spontaneous OS in dogs may provide such a model. Pharmacologic inhibition of histone deacetylase (HDAC) enzymes has a variety of anti-tumor effects but may demonstrate the most utility when utilized in combination with standard cytotoxic therapies. We sought to determine the in vitro and in vivo effects of the HDAC inhibitor valproic acid (VPA) on doxorubicin (DOX) sensitivity in canine and human OS. METHODS We evaluated the in vitro anti-proliferative and apoptotic effects of VPA/DOX combination treatment, alterations in histone acetylation and nuclear DOX accumulation resulting from VPA treatment, and the in vivo efficacy of combination therapy in a xenograft model. RESULTS Treatment of canine and human OS cell lines with clinically achievable VPA concentrations resulted in increased histone acetylation but modest anti-proliferative effects. Pre-incubation with VPA followed by doxorubicin (DOX) resulted in significant growth inhibition and potentiation of apoptosis, associated with a dose-dependent increase in nuclear DOX accumulation. The combination of VPA and DOX was superior to either monotherapy in a canine OS xenograft model. CONCLUSION These results demonstrate a rationale for the addition of HDAC inhibitors to current protocols for the treatment of OS and illustrate the similarities in response to HDAC inhibitors between human and canine OS, lending further credibility to the canine OS model.
Collapse
|
18
|
Chang H, Jeung HC, Jung JJ, Kim TS, Rha SY, Chung HC. Identification of genes associated with chemosensitivity to SAHA/taxane combination treatment in taxane-resistant breast cancer cells. Breast Cancer Res Treat 2010; 125:55-63. [DOI: 10.1007/s10549-010-0825-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 02/25/2010] [Indexed: 11/24/2022]
|
19
|
Peng CY, Jiang J, Zheng HT, Liu XS. Growth-inhibiting effects of arsenic trioxide plus epigenetic therapeutic agents on leukemia cell lines. Leuk Lymphoma 2009; 51:297-303. [DOI: 10.3109/10428190903486212] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Multicenter phase II trial of the histone deacetylase inhibitor pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate in pretreated metastatic melanoma. Melanoma Res 2009; 18:274-8. [PMID: 18626312 DOI: 10.1097/cmr.0b013e328307c248] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Systemic treatment of metastatic melanoma is of low efficacy, and new therapeutic strategies are needed. Histone deacetylase inhibitors are supposed to restore the expression of tumor suppressor genes and induce tumor cell differentiation, growth arrest, and apoptosis. This study was aimed to evaluate the efficacy, safety, and pharmacokinetics of the histone deacetylase inhibitor pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate (MS-275) in patients with pretreated metastatic melanoma. Patients with unresectable AJCC stage IV melanoma refractory to at least one earlier systemic therapy were randomized to receive MS-275 3 mg biweekly (days 1+15, arm A) or 7 mg weekly (days 1+8+15, arm B), in 4-week cycles. The primary study endpoint was objective tumor response, secondary endpoints were safety and time-to-progression. On the basis of Simon's two-stage design, the study initially allowed an entry of 14 patients per arm; if there was at least one responder, additional 33 patients were to be enrolled. Among 28 patients enrolled, no objective response was detected. Four (29%) patients in arm A and three (21%) patients in arm B showed disease stabilizations. Median time-to-progression was comparable in both arms with 55.5 versus 51.5 days, respectively; median overall survival was 8.84 months. Toxicity was mild to moderate with nausea (39%) and hypophosphatemia (29%) as the most frequently reported events. No treatment-related serious adverse events occurred. Single-agent treatment with MS-275 was well-tolerated and showed long-term tumor stabilizations, but no objective responses in pretreated metastatic melanoma. Further evaluation of MS-275 in combination schedules is warranted.
Collapse
|
21
|
New sulfurated derivatives of valproic acid with enhanced histone deacetylase inhibitory activity. Bioorg Med Chem Lett 2008; 18:1893-7. [DOI: 10.1016/j.bmcl.2008.02.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 02/05/2008] [Accepted: 02/05/2008] [Indexed: 11/21/2022]
|