1
|
Dow J, Krysztofiak A, Liu Y, Colon-Rios DA, Rogers FA, Glazer PM. Vulnerability of IDH1-Mutant Cancers to Histone Deacetylase Inhibition via Orthogonal Suppression of DNA Repair. Mol Cancer Res 2021; 19:2057-2067. [PMID: 34535560 PMCID: PMC8642278 DOI: 10.1158/1541-7786.mcr-21-0456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022]
Abstract
Exploitation of DNA repair defects has enabled major advances in treating specific cancers. Recent work discovered that the oncometabolite 2-hydroxyglutarate (2-HG), produced by neomorphic isocitrate dehydrogenase 1/2 (IDH1/2) mutations, confers a homology-directed repair (HDR) defect through 2-HG-induced histone hypermethylation masking HDR signaling. Here, we report that IDH1-mutant cancer cells are profoundly sensitive to the histone deacetylase inhibitor (HDACi) vorinostat, by further suppressing the residual HDR in 2-HG-producing cells. Vorinostat downregulates repair factors BRCA1 and RAD51 via disrupted E2F-factor regulation, causing increased DNA double-strand breaks, reduced DNA repair factor foci, and functional HDR deficiency even beyond 2-HG's effects. This results in greater cell death of IDH1-mutant cells and confers synergy with radiation and PARPi, both against cells in culture and patient-derived tumor xenografts. Our work identifies HDACi's utility against IDH1-mutant cancers, and presents IDH1/2 mutations as potential biomarkers to guide trials testing HDACi in gliomas and other malignancies. IMPLICATIONS: IDH1-mutant cells show profound vulnerability to HDACi treatment, alone and with PARPi and radiation, via HDR suppression, presenting IDH1/2 mutations as biomarkers for HDACi use in gliomas and other malignancies.
Collapse
Affiliation(s)
- Jonathan Dow
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
- Department of Genetics, Yale University School of Medicine. New Haven, Connecticut
| | - Adam Krysztofiak
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
- Department of Genetics, Yale University School of Medicine. New Haven, Connecticut
| | - Daniel A Colon-Rios
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
| | - Faye A Rogers
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut.
- Department of Genetics, Yale University School of Medicine. New Haven, Connecticut
| |
Collapse
|
2
|
Inhibiting homologous recombination by targeting RAD51 protein. Biochim Biophys Acta Rev Cancer 2021; 1876:188597. [PMID: 34332021 DOI: 10.1016/j.bbcan.2021.188597] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/09/2021] [Accepted: 07/24/2021] [Indexed: 02/06/2023]
Abstract
Homologous recombination (HR) is involved in repairing DNA double-strand breaks (DSB), the most harmful for the cell. Regulating HR is essential for maintaining genomic stability. In many forms of cancer, overactivation of HR increases tumor resistance to DNA-damaging treatments. RAD51, HR's core protein, is very often over-expressed in these cancers and plays a critical role in cancer cell development and survival. Targeting RAD51 directly to reduce its activity and its expression is therefore one strategy to sensitize and overcome resistance cancer cells to existing DNA-damaging therapies which remains the limiting factor for the success of targeted therapy. This review describes the structure and biological roles of RAD51, summarizes the different targeted sites of RAD51 and its inhibitory compounds discovered and described in the last decade.
Collapse
|
3
|
Wu K, Chen X, Feng J, Zhang S, Xu Y, Zhang J, Wu Q, You M, Xia B, Ma S. Capilliposide C from Lysimachia capillipes Restores Radiosensitivity in Ionizing Radiation-Resistant Lung Cancer Cells Through Regulation of ERRFI1/EGFR/STAT3 Signaling Pathway. Front Oncol 2021; 11:644117. [PMID: 33869036 PMCID: PMC8047471 DOI: 10.3389/fonc.2021.644117] [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: 12/20/2020] [Accepted: 03/12/2021] [Indexed: 12/25/2022] Open
Abstract
Aims Radiation therapy is used as the primary treatment for lung cancer. Unfortunately, radiation resistance remains to be the major clinic problem for lung cancer patients. Lysimachia capillipes capilliposide C (LC-C), an extract from LC Hemsl, has demonstrated multiple anti-cancer effects in several types of cancer. Here, we investigated the potential therapeutic impacts of LC-C on radiosensitivity in lung cancer cells and their underlying mechanisms. Methods Non-small cell lung cancer cell lines were initially irradiated to generate ionizing radiation (IR)-resistant lung cancer cell lines. RNA-seq analysis was used to examine the whole-transcriptome alteration in IR-resistant lung cancer cells treated with or without LC-C, and the differentially expressed genes with most significance were verified by RT-qPCR. Colony formation assays were performed to determine the effect of LC-C and the target gene ErbB receptor feedback inhibitor 1 (ERRFI1) on radiosensitivity of IR-resistant lung cancer cells. In addition, effects of ERRFI1 on cell cycle distribution, DNA damage repair activity were assessed by flow cytometry and γ-H2AX immunofluorescence staining respectively. Western blotting was performed to identify the activation of related signaling pathways. Tumor xenograft experiments were conducted to observe the effect of LC-C and ERRFI1 on radiosensitivity of IR-resistant lung cancer cells in vivo. Results Compared with parental cells, IR-resistant lung cancer cells were more resistant to radiation. LC-C significantly enhanced the effect of radiation in IR-resistant lung cancer cells both in vitro and in vivo and validated ERRFI1 as a candidate downstream gene by RNA-seq. Forced expression of ERRFI1 alone could significantly increase the radiosensitivity of IR-resistant lung cancer cells, while silencing of ERRFI1 attenuated the radiosensitizing function of LC-C. Accordingly, LC-C and ERRFI1 effectively inhibited IR-induced DNA damage repair, and ERRFI1 significantly induced G2/M checkpoint arrest. Additional investigations revealed that down-regulation of EGFR/STAT3 pathway played an important role in radiosensitization between ERRFI1 and LC-C. Furthermore, the high expression level of ERRFI1 was associated with high overall survival rates in lung cancer patients. Conclusions Treatment of LC-C may serve as a promising therapeutic strategy to overcome the radiation resistance and ERRFI1 may be a potential therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Kan Wu
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Xueqin Chen
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China.,Department of Thoracic Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Jianguo Feng
- Zhejiang Cancer Research Institute, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shirong Zhang
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Yasi Xu
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Jingjing Zhang
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Qiong Wu
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Mingliang You
- Hangzhou Cancer Institute, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Bing Xia
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China.,Department of Oncology, Jiande Second People's Hospital, Hangzhou, China
| | - Shenglin Ma
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
4
|
Liao Y, Wang Y, Cheng M, Huang C, Fan X. Weighted Gene Coexpression Network Analysis of Features That Control Cancer Stem Cells Reveals Prognostic Biomarkers in Lung Adenocarcinoma. Front Genet 2020; 11:311. [PMID: 32391047 PMCID: PMC7192063 DOI: 10.3389/fgene.2020.00311] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/16/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose We aimed to identify new prognostic biomarkers of lung adenocarcinoma (LUAD) based on cancer stem cell theory. Materials and Methods: RNA-seq and microarray data were obtained with clinical information downloaded from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Weighted gene coexpression network analysis (WGCNA) was applied to identify significant module and hub genes. The hub genes were validated via microarray data from GEO, and a prognostic signature with prognostic hub genes was constructed. Results LUAD patients enrolled from TCGA had a higher mRNA expression-based stemness index (mRNAsi) in tumor tissue than in adjacent normal tissue. Some clinical features and prognoses were found to be highly correlated with mRNAsi. WGCNA found that the green module and blue module were the most significant modules related to mRNAsi; 50 key genes were identified in the green module and were enriched mostly in the cell cycle, chromosome segregation, chromosomal region and microtubule binding. Six hub genes were revealed through the protein-protein interaction (PPI) network and Molecular Complex Detection (MCODE) plugin of Cytoscape software. Based on external verification with the GEO database, these six genes are not only expressed at different levels in LUAD and normal tissues but also associated with different clinical features. In addition, the construction of a prognostic signature with three hub genes showed high predictive value. Conclusion mRNAsi-related biomarkers may suggest a new potential treatment strategy for LUAD.
Collapse
Affiliation(s)
- Yi Liao
- Department of Respiratory and Critical Care Medicine II, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yulei Wang
- Department of Respiratory and Critical Care Medicine II, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mengqing Cheng
- Department of Respiratory and Critical Care Medicine II, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chengliang Huang
- Department of Respiratory and Critical Care Medicine II, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine II, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Igaz N, Szőke K, Kovács D, Buhala A, Varga Z, Bélteky P, Rázga Z, Tiszlavicz L, Vizler C, Hideghéty K, Kónya Z, Kiricsi M. Synergistic Radiosensitization by Gold Nanoparticles and the Histone Deacetylase Inhibitor SAHA in 2D and 3D Cancer Cell Cultures. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:158. [PMID: 31963267 PMCID: PMC7023030 DOI: 10.3390/nano10010158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/04/2020] [Accepted: 01/13/2020] [Indexed: 12/24/2022]
Abstract
Radiosensitizing agents are capable of augmenting the damage of ionizing radiation preferentially on cancer cells, thereby increasing the potency and the specificity of radiotherapy. Metal-based nanoparticles have recently gathered ground in radio-enhancement applications, owing to their exceptional competence in amplifying the cell-killing effects of irradiation. Our aim was to examine the radiosensitizing performance of gold nanoparticles (AuNPs) and the chromatin-modifying histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) alone and in combination. We observed that the colony-forming capability of cancer cells decreased significantly and the DNA damage, detected by γH2AX immunostaining, was substantially greater after combinational treatments than upon individual drug exposures followed by irradiation. Synergistic radiosensitizing effects of AuNPs and SAHA were proven on various cell lines, including radioresistant A549 and DU-145 cancer cells. 3D cultures often manifest radio- and drug-resistance, nevertheless, AuNPs in combination with SAHA could effectively enhance the potency of irradiation as the number of viable cells decreased significantly when spheroids received AuNP + SAHA prior to radiotherapy. Our results imply that a relaxed chromatin structure induced by SAHA renders the DNA of cancerous cells more susceptible to the damaging effects of irradiation-triggered, AuNP-released reactive electrons. This feature of AuNPs should be exploited in multimodal treatment approaches.
Collapse
Affiliation(s)
- Nóra Igaz
- Department of Biochemistry and Molecular Biology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary; (N.I.); (K.S.); (D.K.)
- Doctoral School of Biology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Krisztina Szőke
- Department of Biochemistry and Molecular Biology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary; (N.I.); (K.S.); (D.K.)
| | - Dávid Kovács
- Department of Biochemistry and Molecular Biology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary; (N.I.); (K.S.); (D.K.)
| | - Andrea Buhala
- Institute of Biochemistry, Biological Research Centre, Szeged, Temesvári krt. 62, H-6726 Szeged, Hungary; (A.B.); (C.V.)
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, H-6720 Szeged, Hungary; (Z.V.); (K.H.)
| | - Péter Bélteky
- Department of Applied and Environmental Chemistry, University of Szeged, Rerrich Béla tér 1, H-6720 Szeged, Hungary; (P.B.); (Z.K.)
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, Állomás utca 2, H-6720 Szeged, Hungary; (Z.R.); (L.T.)
| | - László Tiszlavicz
- Department of Pathology, University of Szeged, Állomás utca 2, H-6720 Szeged, Hungary; (Z.R.); (L.T.)
| | - Csaba Vizler
- Institute of Biochemistry, Biological Research Centre, Szeged, Temesvári krt. 62, H-6726 Szeged, Hungary; (A.B.); (C.V.)
| | - Katalin Hideghéty
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, H-6720 Szeged, Hungary; (Z.V.); (K.H.)
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Rerrich Béla tér 1, H-6720 Szeged, Hungary; (P.B.); (Z.K.)
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary; (N.I.); (K.S.); (D.K.)
| |
Collapse
|
6
|
Deycmar S, Faccin E, Kazimova T, Knobel PA, Telarovic I, Tschanz F, Waller V, Winkler R, Yong C, Zingariello D, Pruschy M. The relative biological effectiveness of proton irradiation in dependence of DNA damage repair. Br J Radiol 2019; 93:20190494. [PMID: 31687835 DOI: 10.1259/bjr.20190494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Clinical parameters and empirical evidence are the primary determinants for current treatment planning in radiation oncology. Personalized medicine in radiation oncology is only at the very beginning to take the genetic background of a tumor entity into consideration to define an individual treatment regimen, the total dose or the combination with a specific anticancer agent. Likewise, stratification of patients towards proton radiotherapy is linked to its physical advantageous energy deposition at the tumor site with minimal healthy tissue being co-irradiated distal to the target volume. Hence, the fact that photon and proton irradiation also induce different qualities of DNA damages, which require differential DNA damage repair mechanisms has been completely neglected so far. These subtle differences could be efficiently exploited in a personalized treatment approach and could be integrated into personalized treatment planning. A differential requirement of the two major DNA double-strand break repair pathways, homologous recombination and non-homologous end joining, was recently identified in response to proton and photon irradiation, respectively, and subsequently influence the mode of ionizing radiation-induced cell death and susceptibility of tumor cells with defects in DNA repair machineries to either quality of ionizing radiation.This review focuses on the differential DNA-damage responses and subsequent biological processes induced by photon and proton irradiation in dependence of the genetic background and discusses their impact on the unicellular level and in the tumor microenvironment and their implications for combined treatment modalities.
Collapse
Affiliation(s)
- Simon Deycmar
- Laboratory for Applied Radiobiology Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Molecular imaging HDACs class IIa expression-activity and pharmacologic inhibition in intracerebral glioma models in rats using PET/CT/(MRI) with [ 18F]TFAHA. Sci Rep 2019; 9:3595. [PMID: 30837601 PMCID: PMC6401080 DOI: 10.1038/s41598-019-40054-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022] Open
Abstract
HDAC class IIa enzymes (HDAC4, 5, 7, 9) are important for glioma progression, invasion, responses to TMZ and radiotherapy, and prognosis. In this study, we demonstrated the efficacy of PET/CT/(MRI) with [18F]TFAHA for non-invasive and quantitative imaging of HDAC class IIa expression-activity in intracerebral 9L and U87-MG gliomas in rats. Increased accumulation of [18F]TFAHA in 9L and U87-MG tumors was observed at 20 min post radiotracer administration with SUV of 1.45 ± 0.05 and 1.08 ± 0.05, respectively, and tumor-to-cortex SUV ratios of 1.74 ± 0.07 and 1.44 ± 0.03, respectively. [18F]TFAHA accumulation was also observed in normal brain structures known to overexpress HDACs class IIa: hippocampus, n.accumbens, PAG, and cerebellum. These results were confirmed by immunohistochemical staining of brain tissue sections revealing the upregulation of HDACs 4, 5, and 9, and HIF-1α, hypoacetylation of H2AK5ac, H2BK5ac, H3K9ac, H4K8ac, and downregulation of KLF4. Significant reduction in [18F]TFAHA accumulation in 9L tumors was observed after administration of HDACs class IIa specific inhibitor MC1568, but not the SIRT1 specific inhibitor EX-527. Thus, PET/CT/(MRI) with [18F]TFAHA can facilitate studies to elucidate the roles of HDAC class IIa enzymes in gliomagenesis and progression and to optimize therapeutic doses of novel HDACs class IIa inhibitors in gliomas.
Collapse
|
8
|
Teknos TN, Grecula J, Agrawal A, Old MO, Ozer E, Carrau R, Kang S, Rocco J, Blakaj D, Diavolitsis V, Kumar B, Kumar P, Pan Q, Palettas M, Wei L, Baiocchi R, Savvides P. A phase 1 trial of Vorinostat in combination with concurrent chemoradiation therapy in the treatment of advanced staged head and neck squamous cell carcinoma. Invest New Drugs 2018; 37:702-710. [PMID: 30569244 DOI: 10.1007/s10637-018-0696-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022]
Abstract
Purpose Vorinostat is a potent HDAC inhibitor that sensitizes head and neck squamous cell carcinoma (HNSCC) to cytotoxic therapy while sparing normal epithelium. The primary objective of this Phase I study was to determine the maximally tolerated dose (MTD) and safety of Vorinostat in combination with standard chemoradiation therapy treatment in HNSCC. Patients and Methods Eligible patients had pathologically confirmed Stage III, IVa, IVb HNSCC, that was unresectable or borderline resectable involving the larynx, hypopharynx, nasopharynx, and oropharynx. Vorinostat was administered at the assigned dosage level (100-400 mg, three times weekly) in a standard 3 + 3 dose escalation design. Vorinostat therapy began 1 week prior to initiation of standard, concurrent chemoradiation therapy and continued during the entire course of therapy. Results Twenty six patients met eligibility criteria and completed the entire protocol. The primary tumor sites included tonsil (12), base of tongue (9), posterior pharyngeal wall (1), larynx (4) and hypopharynx (3). Of the 26 patients, 17 were HPV-positive and 9 were HPV-negative. The MTD of Vorinostat was 300 mg administered every other day. Anemia (n = 23/26) and leukopenia (n = 20/26) were the most commonly identified toxicities. The most common Grade3/4 events included leukopenia (n = 11) and lymphopenia (n = 17). No patient had Grade IV mucositis, dermatitis or xerostomia. The median follow time was 33.8 months (range 1.6-82.9 months). Twenty four of 26 (96.2%) patients had a complete response to therapy. Conclusion Vorinostat in combination with concurrent chemoradiation therapy is a safe and highly effective treatment regimen in HNSCC. There was a high rate of complete response to therapy with toxicity rates comparable, if not favorable to existing therapies. Further investigation in Phase II and III trials is strongly recommended.
Collapse
Affiliation(s)
- Theodoros N Teknos
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA. .,Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| | - J Grecula
- Radiation Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - A Agrawal
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - M O Old
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - E Ozer
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - R Carrau
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - S Kang
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - J Rocco
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - D Blakaj
- Radiation Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - V Diavolitsis
- Radiation Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - B Kumar
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - P Kumar
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - Q Pan
- Otolaryngology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - M Palettas
- Center for Biostatistics, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - L Wei
- Center for Biostatistics, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - R Baiocchi
- Hematology-Medical Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| | - P Savvides
- Hematology-Medical Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute (OSUCCC - James), Columbus, OH, USA
| |
Collapse
|
9
|
Wang S, Gao P, Li N, Chen P, Wang J, He N, Ji K, Du L, Liu Q. Autocrine secretions enhance radioresistance in an exosome‑independent manner in NSCLC cells. Int J Oncol 2018; 54:229-238. [PMID: 30387839 DOI: 10.3892/ijo.2018.4620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/01/2018] [Indexed: 11/05/2022] Open
Abstract
Radiotherapy resistance in patient with non‑small cell lung cancer (NSCLC) reduces patient survival and remains a significant challenge for the treatment of NSCLC. Radiation resistance has been demonstrated to be affected by secreted factors, yet it remains unclear how autocrine secretions affect the radioresistance of NSCLC cells. In the present study, the NSCLC cell line, NCI‑H460, was irradiated with γ‑rays (4 Gy) and then cultured in medium from H460 cells or normal medium to examine the potential influence of cell secretions on the radiation resistance of H460 cells. Cell viability, accumulation of reactive oxygen species and DNA repair capacity were all markedly improved in the irradiated H460 cells that were cultured in conditioned medium (CM), compared with those cells cultured in normal medium. In addition, G2/M cell cycle arrest and upregulation of homologous recombination repair proteins were observed in the CM‑treated cells, while exosomes secreted by H460 cells had no influence on the radiation resistance of H460 cells. Taken together, these results indicate that autocrine secretions enhance the radiation resistance of γ‑irradiated H460 cells and that these secretions mainly affect the DNA repair process.
Collapse
Affiliation(s)
- Shuang Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Piaoyang Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Na Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Ping Chen
- Department of Neurology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010017, P.R. China
| | - Jinhan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P.R. China
| |
Collapse
|
10
|
Zhang D, Dong Y, Zhao Y, Zhou C, Qian Y, Hegde ML, Wang H, Han S. Sinomenine hydrochloride sensitizes cervical cancer cells to ionizing radiation by impairing DNA damage response. Oncol Rep 2018; 40:2886-2895. [PMID: 30226618 PMCID: PMC6151895 DOI: 10.3892/or.2018.6693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/03/2018] [Indexed: 02/01/2023] Open
Abstract
The use of plant-based compounds derived from traditional medicine to improve human diseases has been gaining momentum, due to their high bioavailability and moderate adverse effects. Sinomenine is one such biomonomer alkali compound derived from Sinomenium acutum and is known for its anti-inflammatory and antitumor effects. However, the molecular mechanism(s) of its antitumor properties are not fully characterized. In the present study, we evaluated the radiosensitizing effects of the water-soluble sinomenine, sinomenine hydrochloride (SH) in human cervical cancer cell line (HeLa). SH sensitized HeLa cells to ionizing radiation (IR) by promoting accumulation of IR-induced DNA double-strand breaks (DSBs) and also by interfering with DNA damage checkpoint activation. We then investigated the molecular mechanisms underlying the SH-mediated cellular sensitization to IR and found that SH inhibited the expression of DNA damage response (DDR) factors Ku80 and Rad51 at the transcription level. Finally, the radiosensitizing activity of SH was confirmed in a cervical cancer mouse xenograft model. The combinatorial treatment of SH and IR significantly slowed the tumor growth rate compared with IR alone. Collectively, our study not only provides molecular insights into the novel role of SH in cellular response to IR, but also suggests a therapeutic potential of SH as a radiosensitizer in cervical cancer therapy.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yiping Dong
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ying Zhao
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Congya Zhou
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuanjie Qian
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Muralidhar L Hegde
- Department of Radiation Oncology, Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Haibo Wang
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Suxia Han
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
11
|
Lee Y, Okayasu R. Strategies to Enhance Radiosensitivity to Heavy Ion Radiation Therapy. Int J Part Ther 2018; 5:114-121. [DOI: 10.14338/ijpt-18-00014.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/16/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Younghyun Lee
- Center for Radiological Research, Columbia University Medical Center, New York, NY, USA
| | - Ryuichi Okayasu
- Department of Basic Medical Sciences for Radiation Damages, National Institutes for Quantum and Radiological Science and Technology/National Institute of Radiological Sciences, Japan
| |
Collapse
|
12
|
Deycmar S, Pruschy M. Combined Treatment Modalities for High-Energy Proton Irradiation: Exploiting Specific DNA Repair Dependencies. Int J Part Ther 2018; 5:133-139. [DOI: 10.14338/ijpt-18-00020.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/05/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Simon Deycmar
- Department of Radiation Oncology, Laboratory for Applied Radiobiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Pruschy
- Department of Radiation Oncology, Laboratory for Applied Radiobiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Tsuboi K. Advantages and Limitations in the Use of Combination Therapies with Charged Particle Radiation Therapy. Int J Part Ther 2018; 5:122-132. [PMID: 31773024 DOI: 10.14338/ijpt-18-00019.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/21/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose Studies are currently underway to help provide basic and clinical evidence for combination particle beam radiation therapy, on which there are few published reports. The purpose of this article is to summarize the current status in the use of particle beams combined with other modalities. Results Following from experiences in x-ray radiation therapy, combination therapy with proton beams (PBT) has been attempted, and several clinical studies have reported improved survival rates for patients with non-small cell lung cancer, pancreatic cancers, esophageal cancers, and glioblastomas. Recently, basic studies combining PBT with PARP inhibitors and histone deacetylase inhibitors have also reported promising results. In the area of carbon ion therapy (CIT), there are few clinical reports on combination therapy; however, the number of basic research reports exceeds that for PBT. So far, the combined use of cytotoxic drugs with CIT yields independent additive effects. In addition, it is notable that combination therapy with CIT is effective against radioresistant cancer stem-like cells. Recent evidence also suggests that local radiation therapy can induce an effective antitumor immune response. There has been an increased use of combination immune-modulating agents and cytokines with particle beams, especially CIT. The field of radiation therapy is evolving from a strong reliance on local-regional treatment to a growing reliance on systemic immunotherapy. Conclusions The combined use of anticancer agents with particle radiation therapy has a considerable potential effect. Future research in molecular targeting therapy and immunotherapy may help identify the most efficacious approach for combination therapy with protons and carbon ions.
Collapse
Affiliation(s)
- Koji Tsuboi
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Japan
| |
Collapse
|
14
|
Histone Deacetylase Inhibitor Induced Radiation Sensitization Effects on Human Cancer Cells after Photon and Hadron Radiation Exposure. Int J Mol Sci 2018; 19:ijms19020496. [PMID: 29414878 PMCID: PMC5855718 DOI: 10.3390/ijms19020496] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 12/25/2022] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase inhibitor, which has been widely utilized throughout the cancer research field. SAHA-induced radiosensitization in normal human fibroblasts AG1522 and lung carcinoma cells A549 were evaluated with a combination of γ-rays, proton, and carbon ion exposure. Growth delay was observed in both cell lines during SAHA treatment; 2 μM SAHA treatment decreased clonogenicity and induced cell cycle block in G1 phase but 0.2 μM SAHA treatment did not show either of them. Low LET (Linear Energy Transfer) irradiated A549 cells showed radiosensitization effects on cell killing in cycling and G1 phase with 0.2 or 2 μM SAHA pretreatment. In contrast, minimal sensitization was observed in normal human cells after low and high LET radiation exposure. The potentially lethal damage repair was not affected by SAHA treatment. SAHA treatment reduced the rate of γ-H2AX foci disappearance and suppressed RAD51 and RPA (Replication Protein A) focus formation. Suppression of DNA double strand break repair by SAHA did not result in the differences of SAHA-induced radiosensitization between human cancer cells and normal cells. In conclusion, our results suggest SAHA treatment will sensitize cancer cells to low and high LET radiation with minimum effects to normal cells.
Collapse
|
15
|
Tsai CL, Liu WL, Hsu FM, Yang PS, Yen RF, Tzen KY, Cheng AL, Chen PJ, Cheng JCH. Targeting histone deacetylase 4/ubiquitin-conjugating enzyme 9 impairs DNA repair for radiosensitization of hepatocellular carcinoma cells in mice. Hepatology 2018. [PMID: 28646552 DOI: 10.1002/hep.29328] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several strategies to improve the efficacy of radiation therapy against hepatocellular carcinoma (HCC) have been investigated. One approach is to develop radiosensitizing compounds. Because histone deacetylase 4 (HDAC4) is highly expressed in liver cancer and known to regulate oncogenesis through chromatin structure remodeling and controlling protein access to DNA, we postulated that HDAC4 inhibition might enhance radiation's effect on HCC cells. HCC cell lines (Huh7 and PLC5) and an ectopic xenograft were pretreated with HDAC inhibitor or short hairpin RNA to knock down expression of HDAC4 and then irradiated (2.5-10.0 Gy). We evaluated cell survival by a clonogenic assay; apoptosis by Annexin V immunofluorescence; γH2AX, Rad51, and HDAC4 by immunofluorescence staining; HDAC4, Rad51, and ubiquitin-conjugating enzyme 9 (Ubc9) in HCC cell nuclei by cell fractionation and confocal microscopy; physical interaction between HDAC4/Rad51/Ubc9 by immunoprecipitation; and the downstream targets of HDAC4 knockdown by immunoblotting. Both HDAC4 knockdown and HDAC inhibitor enhanced radiation-induced cell death and reduced homologous recombination repair of DNA double-strand breaks and protein kinase B activation, leading to increased apoptosis. HDAC4 knockdown with or without an HDAC inhibitor significantly delayed tumor growth in a radiation-treated xenograft model. Radiation stimulated nuclear translocation of Rad51 in an HDAC4-dependent manner and the binding of Ubc9 directly to HDAC4, which led to Ubc9 acetylation. Moreover, these effects were accompanied by HDAC4/Ubc9/Rad51 complex dissociation through inhibiting nuclear translocation. Conclusion: HDAC4 signaling blockade enhances radiation-induced lethality in HCC cells and xenografts. These findings raise the possibility that HDAC4/Ubc9/Rad51 complex in DNA repair may be a target for radiosensitization of HCC. (Hepatology 2018;67:586-599).
Collapse
Affiliation(s)
- Chiao-Ling Tsai
- Graduate Institutes of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Lin Liu
- Graduate Institutes of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Feng-Ming Hsu
- Graduate Institutes of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Sheng Yang
- Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Kai-Yuan Tzen
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of General Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ann-Lii Cheng
- Graduate Institutes of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Jer Chen
- Graduate Institutes of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jason Chia-Hsien Cheng
- Graduate Institutes of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institutes of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
López-Iglesias AA, Herrero AB, Chesi M, San-Segundo L, González-Méndez L, Hernández-García S, Misiewicz-Krzeminska I, Quwaider D, Martín-Sánchez M, Primo D, Paíno T, Bergsagel PL, Mehrling T, González-Díaz M, San-Miguel JF, Mateos MV, Gutiérrez NC, Garayoa M, Ocio EM. Preclinical anti-myeloma activity of EDO-S101, a new bendamustine-derived molecule with added HDACi activity, through potent DNA damage induction and impairment of DNA repair. J Hematol Oncol 2017. [PMID: 28633670 PMCID: PMC5477689 DOI: 10.1186/s13045-017-0495-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Despite recent advances in the treatment of multiple myeloma (MM), the prognosis of most patients remains poor, and resistance to traditional and new drugs frequently occurs. EDO-S101 is a novel therapeutic agent conceived as the fusion of a histone deacetylase inhibitor radical to bendamustine, with the aim of potentiating its alkylating activity. Methods The efficacy of EDO-S101 was evaluated in vitro, ex vivo and in vivo, alone, and in combination with standard anti-myeloma agents. The underlying mechanisms of action were also evaluated on MM cell lines, patient samples, and different murine models. Results EDO-S101 displayed potent activity in vitro in MM cell lines (IC50 1.6–4.8 μM) and ex vivo in cells isolated from MM patients, which was higher than that of bendamustine and independent of the p53 status and previous melphalan resistance. This activity was confirmed in vivo, in a CB17-SCID murine plasmacytoma model and in de novo Vk*MYC mice, leading to a significant survival improvement in both models. In addition, EDO-S101 was the only drug with single-agent activity in the multidrug resistant Vk12653 murine model. Attending to its mechanism of action, the molecule showed both, a HDACi effect (demonstrated by α-tubulin and histone hyperacetylation) and a DNA-damaging effect (shown by an increase in γH2AX); the latter being again clearly more potent than that of bendamustine. Using a reporter plasmid integrated into the genome of some MM cell lines, we demonstrate that, apart from inducing a potent DNA damage, EDO-S101 specifically inhibited the double strand break repair by the homologous recombination pathway. Moreover, EDO-S101 treatment reduced the recruitment of repair proteins such as RAD51 to DNA-damage sites identified as γH2AX foci. Finally, EDO-S101 preclinically synergized with bortezomib, both in vitro and in vivo. Conclusion These findings provide rationale for the clinical investigation of EDO-S101 in MM, either as a single agent or in combination with other anti-MM drugs, particularly proteasome inhibitors. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0495-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana-Alicia López-Iglesias
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Ana B Herrero
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic, Arizona, USA
| | - Laura San-Segundo
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Lorena González-Méndez
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Susana Hernández-García
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | | | - Dalia Quwaider
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Montserrat Martín-Sánchez
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | | | - Teresa Paíno
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain.
| | | | | | - Marcos González-Díaz
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Jesús F San-Miguel
- Center for Applied Medical Research (CIMA), IDISNA, University Clinic of Navarra, Pamplona, Spain
| | - María-Victoria Mateos
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Norma C Gutiérrez
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Mercedes Garayoa
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| | - Enrique M Ocio
- University Hospital of Salamanca (IBSAL) & Cancer Research Center (IBMCC-CSIC-USAL), Salamanca, Spain
| |
Collapse
|
17
|
Bhattacharya S, Srinivasan K, Abdisalaam S, Su F, Raj P, Dozmorov I, Mishra R, Wakeland EK, Ghose S, Mukherjee S, Asaithamby A. RAD51 interconnects between DNA replication, DNA repair and immunity. Nucleic Acids Res 2017; 45:4590-4605. [PMID: 28334891 PMCID: PMC5416901 DOI: 10.1093/nar/gkx126] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/11/2022] Open
Abstract
RAD51, a multifunctional protein, plays a central role in DNA replication and homologous recombination repair, and is known to be involved in cancer development. We identified a novel role for RAD51 in innate immune response signaling. Defects in RAD51 lead to the accumulation of self-DNA in the cytoplasm, triggering a STING-mediated innate immune response after replication stress and DNA damage. In the absence of RAD51, the unprotected newly replicated genome is degraded by the exonuclease activity of MRE11, and the fragmented nascent DNA accumulates in the cytosol, initiating an innate immune response. Our data suggest that in addition to playing roles in homologous recombination-mediated DNA double-strand break repair and replication fork processing, RAD51 is also implicated in the suppression of innate immunity. Thus, our study reveals a previously uncharacterized role of RAD51 in initiating immune signaling, placing it at the hub of new interconnections between DNA replication, DNA repair, and immunity.
Collapse
Affiliation(s)
- Souparno Bhattacharya
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kalayarasan Srinivasan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Salim Abdisalaam
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Fengtao Su
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Igor Dozmorov
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ritu Mishra
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Edward K. Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Subroto Ghose
- Department of Molecular Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shibani Mukherjee
- Department of Molecular Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aroumougame Asaithamby
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
18
|
Gong F, Chiu LY, Miller KM. Acetylation Reader Proteins: Linking Acetylation Signaling to Genome Maintenance and Cancer. PLoS Genet 2016; 12:e1006272. [PMID: 27631103 PMCID: PMC5025232 DOI: 10.1371/journal.pgen.1006272] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chromatin-based DNA damage response (DDR) pathways are fundamental for preventing genome and epigenome instability, which are prevalent in cancer. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) catalyze the addition and removal of acetyl groups on lysine residues, a post-translational modification important for the DDR. Acetylation can alter chromatin structure as well as function by providing binding signals for reader proteins containing acetyl-lysine recognition domains, including the bromodomain (BRD). Acetylation dynamics occur upon DNA damage in part to regulate chromatin and BRD protein interactions that mediate key DDR activities. In cancer, DDR and acetylation pathways are often mutated or abnormally expressed. DNA damaging agents and drugs targeting epigenetic regulators, including HATs, HDACs, and BRD proteins, are used or are being developed to treat cancer. Here, we discuss how histone acetylation pathways, with a focus on acetylation reader proteins, promote genome stability and the DDR. We analyze how acetylation signaling impacts the DDR in the context of cancer and its treatments. Understanding the relationship between epigenetic regulators, the DDR, and chromatin is integral for obtaining a mechanistic understanding of genome and epigenome maintenance pathways, information that can be leveraged for targeting acetylation signaling, and/or the DDR to treat diseases, including cancer.
Collapse
Affiliation(s)
- Fade Gong
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Li-Ya Chiu
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kyle M. Miller
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
19
|
Colis LC, Herzon SB. Synergistic potentiation of (-)-lomaiviticin A cytotoxicity by the ATR inhibitor VE-821. Bioorg Med Chem Lett 2016; 26:3122-3126. [PMID: 27177826 PMCID: PMC4899226 DOI: 10.1016/j.bmcl.2016.04.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 11/23/2022]
Abstract
(-)-Lomaiviticin A (1) is a cytotoxic bacterial metabolite that induces double-strand breaks in DNA. Here we show that the cytotoxicity of (-)-lomaiviticin A (1) is synergistically potentiated in the presence of VE-821 (7), an inhibitor of ataxia telangiectasia and Rad3-related protein (ATR). While 0.5nM 1 or 10μM 7 alone are non-lethal to K562 cells, co-incubation of the two leads to high levels of cell kill (81% and 94% after 24 and 48h, respectively). Mechanistic data indicate that cells treated with 1 and 7 suffer extensive DNA double-strand breaks and apoptosis. These data suggest combinations of 1 and 7 may be a valuable chemotherapeutic strategy.
Collapse
Affiliation(s)
- Laureen C Colis
- Department of Chemistry, Yale University, New Haven, CT 06520, United States
| | - Seth B Herzon
- Department of Chemistry, Yale University, New Haven, CT 06520, United States; Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
20
|
Hegde M, Mantelingu K, Pandey M, Pavankumar CS, Rangappa KS, Raghavan SC. Combinatorial Study of a Novel Poly (ADP-ribose) Polymerase Inhibitor and an HDAC Inhibitor, SAHA, in Leukemic Cell Lines. Target Oncol 2016; 11:655-665. [DOI: 10.1007/s11523-016-0441-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
de Andrade PV, Andrade AF, de Paula Queiroz RG, Scrideli CA, Tone LG, Valera ET. The histone deacetylase inhibitor PCI-24781 as a putative radiosensitizer in pediatric glioblastoma cell lines. Cancer Cell Int 2016; 16:31. [PMID: 27095947 PMCID: PMC4835828 DOI: 10.1186/s12935-016-0306-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/07/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is considered to be one of the most aggressive tumors of the central nervous system (CNS). Even with the use of modern treatment protocols, the prognosis remains reserved, with children with GBM having a mean survival of 12-15 months. METHODS In the present study we investigated the potential radiosensitizing effect of PCI-24781, a potent pan-histone deacetylase inhibitor (HDACi), on the SF188 and KNS42 cell lines of pediatric GBM. Cell proliferation rates, clonogenicity and apoptosis were compared in the presence and absence of treatment with PCI-24781. We also compared the clonogenicity rates of the irradiated SF188 and KNS42 cell lines with or without previous treatment with PCI-24781 at the doses of 0.25-16 μM. In addition, we investigated the effects of PCI-24781 on the expression of some of the main proteins responsible for the repair of double-strand DNA breaks caused by irradiation. RESULTS The inhibitor blocked cell proliferation, induced death by apoptosis and reduced the colony forming capacity of the cell lines, both of them showing a significant decrease of colony formation at all irradiation doses. The expression of the Rad51 protein, important for the homologous recombination (HR) repair pathway, and of the DNA-PKcs, Ku70 and Ku86 proteins, important for the non-homologous end joining (NHEJ) repair pathway, was more reduced when the irradiated cell line was previously treated with PCI-24781 than when it was treated exclusively with radiotherapy. CONCLUSIONS These findings demonstrate that HDACi PCI-24781 has a radiosensitizing profile that compromises the repair of double-strand DNA breaks in cells of pediatric GBM treated with radiotherapy.
Collapse
Affiliation(s)
- Pamela Viani de Andrade
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto-USP, University of São Paulo, 7º andar. Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil
| | - Augusto Faria Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil
| | - Rosane Gomes de Paula Queiroz
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto-USP, University of São Paulo, 7º andar. Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil
| | - Carlos Alberto Scrideli
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto-USP, University of São Paulo, 7º andar. Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil
| | - Luiz Gonzaga Tone
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto-USP, University of São Paulo, 7º andar. Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil ; Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto-USP, University of São Paulo, 7º andar. Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, SP CEP 14048-900 Brazil
| |
Collapse
|
22
|
Feng J, Zhang S, Wu K, Wang B, Wong JYC, Jiang H, Xu R, Ying L, Huang H, Zheng X, Chen X, Ma S. Combined Effects of Suberoylanilide Hydroxamic Acid and Cisplatin on Radiation Sensitivity and Cancer Cell Invasion in Non-Small Cell Lung Cancer. Mol Cancer Ther 2016; 15:842-53. [PMID: 26839308 DOI: 10.1158/1535-7163.mct-15-0445] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 01/19/2016] [Indexed: 11/16/2022]
Abstract
Lung cancer is a leading cause of cancer-related mortality worldwide, and concurrent chemoradiotherapy has been explored as a therapeutic option. However, the chemotherapeutic agents cannot be administered for most patients at full doses safely with radical doses of thoracic radiation, and further optimizations of the chemotherapy regimen to be given with radiation are needed. In this study, we examined the effects of suberoylanilide hydroxamic acid (SAHA) and cisplatin on DNA damage repairs, and determined the combination effects of SAHA and cisplatin on human non-small cell lung cancer (NSCLC) cells in response to treatment of ionizing radiation (IR), and on tumor growth of lung cancer H460 xenografts receiving radiotherapy. We also investigated the potential differentiation effect of SAHA and its consequences on cancer cell invasion. Our results showed that SAHA and cisplatin compromise distinct DNA damage repair pathways, and treatment with SAHA enhanced synergistic radiosensitization effects of cisplatin in established NSCLC cell lines in a p53-independent manner, and decreased the DNA damage repair capability in cisplatin-treated primary NSCLC tumor tissues in response to IR. SAHA combined with cisplatin also significantly increased inhibitory effect of radiotherapy on tumor growth in the mouse xenograft model. In addition, SAHA can induce differentiation in stem cell-like cancer cell population, reduce tumorigenicity, and decrease invasiveness of human lung cancer cells. In conclusion, our data suggest a potential clinical impact for SAHA as a radiosensitizer and as a part of a chemoradiotherapy regimen for NSCLC. Mol Cancer Ther; 15(5); 842-53. ©2016 AACR.
Collapse
Affiliation(s)
- Jianguo Feng
- Cancer Research Institute and Key Laboratory Diagnoses and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shirong Zhang
- Department of Oncology, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China. Affiliated Hangzhou First People's Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kan Wu
- Affiliated Hangzhou First People's Hospital of Zhejiang Chinese Medical University, Hangzhou, China. Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, China
| | - Bing Wang
- Affiliated Hangzhou First People's Hospital of Zhejiang Chinese Medical University, Hangzhou, China. Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, China
| | - Jeffrey Y C Wong
- Department of Radiation Oncology, City of Hope Cancer Center, Duarte, California
| | - Hong Jiang
- Department of Oncology, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - Rujun Xu
- Department of Oncology, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - Lisha Ying
- Cancer Research Institute and Key Laboratory Diagnoses and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Haixiu Huang
- Department of Oncology, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - Xiaoliang Zheng
- Centre of Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Xufeng Chen
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California
| | - Shenglin Ma
- Department of Oncology, Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China. Affiliated Hangzhou First People's Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
23
|
Xiang J, Leung C, Zhang Z, Hu C, Geng C, Liu L, Yi L, Li Z, Berenson J, Bai X. Synthesis and Evaluation of 2-Alkylthio-4-(N-substituted sulfonamide)pyrimidine Hydroxamic Acids as Anti-myeloma Agents. Chem Biol Drug Des 2015; 87:472-7. [PMID: 26518472 DOI: 10.1111/cbdd.12678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/11/2015] [Accepted: 10/22/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Jinbao Xiang
- The Center for Combinatorial Chemistry and Drug Discovery; The School of Pharmaceutical Sciences; The College of Chemistry; Jilin University; 1266 Fujin Road Changchun Jilin 130021 China
| | - Crystal Leung
- Institute for Myeloma & Bone Cancer Research; 9201 West Sunset Blvd, Suite 300 West Hollywood CA 90069 USA
| | - Zhuoqi Zhang
- The Center for Combinatorial Chemistry and Drug Discovery; The School of Pharmaceutical Sciences; The College of Chemistry; Jilin University; 1266 Fujin Road Changchun Jilin 130021 China
| | - Cassie Hu
- Institute for Myeloma & Bone Cancer Research; 9201 West Sunset Blvd, Suite 300 West Hollywood CA 90069 USA
| | - Chao Geng
- The Center for Combinatorial Chemistry and Drug Discovery; The School of Pharmaceutical Sciences; The College of Chemistry; Jilin University; 1266 Fujin Road Changchun Jilin 130021 China
| | - Lili Liu
- Changchun Discovery Sciences, Ltd.; 750 Jinbi Street Changchun Jilin 130117 China
| | - Lang Yi
- The Center for Combinatorial Chemistry and Drug Discovery; The School of Pharmaceutical Sciences; The College of Chemistry; Jilin University; 1266 Fujin Road Changchun Jilin 130021 China
| | - Zhiwei Li
- The Center for Combinatorial Chemistry and Drug Discovery; The School of Pharmaceutical Sciences; The College of Chemistry; Jilin University; 1266 Fujin Road Changchun Jilin 130021 China
| | - James Berenson
- Institute for Myeloma & Bone Cancer Research; 9201 West Sunset Blvd, Suite 300 West Hollywood CA 90069 USA
| | - Xu Bai
- The Center for Combinatorial Chemistry and Drug Discovery; The School of Pharmaceutical Sciences; The College of Chemistry; Jilin University; 1266 Fujin Road Changchun Jilin 130021 China
| |
Collapse
|
24
|
Fontana AO, Augsburger MA, Grosse N, Guckenberger M, Lomax AJ, Sartori AA, Pruschy MN. Differential DNA repair pathway choice in cancer cells after proton- and photon-irradiation. Radiother Oncol 2015; 116:374-80. [DOI: 10.1016/j.radonc.2015.08.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/12/2015] [Accepted: 08/15/2015] [Indexed: 01/23/2023]
|
25
|
Maes K, De Smedt E, Lemaire M, De Raeve H, Menu E, Van Valckenborgh E, McClue S, Vanderkerken K, De Bruyne E. The role of DNA damage and repair in decitabine-mediated apoptosis in multiple myeloma. Oncotarget 2015; 5:3115-29. [PMID: 24833108 PMCID: PMC4102796 DOI: 10.18632/oncotarget.1821] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi) are under investigation for the treatment of cancer, including the plasma cell malignancy multiple myeloma (MM). Evidence exists that DNA damage and repair contribute to the cytotoxicity mediated by the DNMTi decitabine. Here, we investigated the DNA damage response (DDR) induced by decitabine in MM using 4 human MM cell lines and the murine 5T33MM model. In addition, we explored how the HDACi JNJ-26481585 affects this DDR. Decitabine induced DNA damage (gamma-H2AX foci formation), followed by a G0/G1- or G2/M-phase arrest and caspase-mediated apoptosis. JNJ-26481585 enhanced the anti-MM effect of decitabine both in vitro and in vivo. As JNJ-26481585 did not enhance decitabine-mediated gamma-H2AX foci formation, we investigated the DNA repair response towards decitabine and/or JNJ-26481585. Decitabine augmented RAD51 foci formation (marker for homologous recombination (HR)) and/or 53BP1 foci formation (marker for non-homologous end joining (NHEJ)). Interestingly, JNJ-26481585 negatively affected basal or decitabine-induced RAD51 foci formation. Finally, B02 (RAD51 inhibitor) enhanced decitabine-mediated apoptosis. Together, we report that decitabine-induced DNA damage stimulates HR and/or NHEJ. JNJ-26481585 negatively affects RAD51 foci formation, thereby providing an additional explanation for the combinatory effect between decitabine and JNJ-26481585.
Collapse
Affiliation(s)
- Ken Maes
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ward A, Khanna KK, Wiegmans AP. Targeting homologous recombination, new pre-clinical and clinical therapeutic combinations inhibiting RAD51. Cancer Treat Rev 2014; 41:35-45. [PMID: 25467108 DOI: 10.1016/j.ctrv.2014.10.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 12/22/2022]
Abstract
The DNA damage response (DDR) is essential for maintaining genomic stability and cell survival. However, when tumour cells with deficiencies in HR are faced with radio- and chemotherapies they are forced to rely on error-prone, alternative repair pathways or aberrant HR for survival; threatening genome integrity and driving further mutation. Accurate therapeutic targeting of the key drivers of DNA repair can circumvent survival pathways and avoid aggressive therapy resistant mutants. Several studies have identified that stabilization of the cancer genome in HR deficient cells can be achieved by overexpression of the recombinase RAD51. Radio- and chemotherapeutic resistance is associated with overactive HR repair mechanisms. However no clinical trials have directly targeted RAD51, despite RAD51 displaying synergy in several drug screens against multiple cancer types. Currently synthetic lethality targeting the DDR pathways and HR deficiency has had clinical success with BRCA1 functional loss and PARP inhibition. In this review we suggest that clinical outcomes could be improved by additionally targeting RAD51. We examine the latest developments in directly and indirectly targeting RAD51. We scrutinize the potential treatment efficacy and future clinical applications of RAD51 inhibitors as single agents and in combination with other therapies and consider the best therapeutic options.
Collapse
Affiliation(s)
- Ambber Ward
- Signal Transduction Laboratory, QIMR Berghofer, Herston Rd, Herston, QLD 4006, Australia.
| | - Kum Kum Khanna
- Signal Transduction Laboratory, QIMR Berghofer, Herston Rd, Herston, QLD 4006, Australia.
| | - Adrian P Wiegmans
- Signal Transduction Laboratory, QIMR Berghofer, Herston Rd, Herston, QLD 4006, Australia.
| |
Collapse
|
27
|
Alagpulinsa DA, Ayyadevara S, Shmookler Reis RJ. A Small-Molecule Inhibitor of RAD51 Reduces Homologous Recombination and Sensitizes Multiple Myeloma Cells to Doxorubicin. Front Oncol 2014; 4:289. [PMID: 25401086 PMCID: PMC4214226 DOI: 10.3389/fonc.2014.00289] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/06/2014] [Indexed: 11/22/2022] Open
Abstract
We previously reported high expression of RAD51 and increased homologous recombination (HR) rates in multiple myeloma (MM) cells, and showed that genomic instability and disease progression are commensurate with HR levels. Moreover, high RAD51 expression in vivo is associated with chemoresistance and poor patient survival. Doxorubicin (DOX) is one of the most widely used drug treatments in MM chemotherapy. DOX is cytotoxic because it induces DNA double-strand breaks, which can be repaired by RAD51-mediated HR; activation of this pathway thus contributes to resistance. To investigate the role of RAD51 in MM drug resistance, we assessed the ability of B02, a small-molecule inhibitor of RAD51, to enhance DOX sensitivity of MM cells. Combining low-toxicity doses of DOX and B02 resulted in significant synthetic lethality, observed as increased apoptosis and reduced viability compared to either agent alone, or to the product of their individual effects. In contrast, the combination did not produce significant synergy against normal human CD19+ B cells from peripheral blood. DOX induced RAD51 at both mRNA and protein levels, while arresting cells in S and G2. DOX treatment also increased the number of RAD51 foci, a marker of HR repair, so that the fraction of cells with ≥5 foci rose fourfold, whereas γH2AX foci rose far less, implying that most new breaks are repaired. When B02 treatment preceded DOX exposure, the induction of RAD51 foci was severely blunted, whereas, γH2AX foci rose significantly relative to basal levels or either agent alone. In MM cells carrying a chromosomally integrated reporter of HR repair, DOX increased HR events while B02 inhibition of RAD51 blocked the HR response. These studies demonstrate the crucial role of RAD51 in protecting MM cells from genotoxic agents such as DOX, and suggest that specific inhibition of RAD51 may be an effective means to block DNA repair in MM cells and thus to enhance the efficacy of chemotherapy.
Collapse
Affiliation(s)
- David A Alagpulinsa
- McClellan Veterans Medical Center, Central Arkansas Veterans Healthcare System , Little Rock, AR , USA ; Department of Geriatrics, University of Arkansas for Medical Science , Little Rock, AR , USA
| | - Srinivas Ayyadevara
- McClellan Veterans Medical Center, Central Arkansas Veterans Healthcare System , Little Rock, AR , USA ; Department of Geriatrics, University of Arkansas for Medical Science , Little Rock, AR , USA
| | - Robert Joseph Shmookler Reis
- McClellan Veterans Medical Center, Central Arkansas Veterans Healthcare System , Little Rock, AR , USA ; Department of Geriatrics, University of Arkansas for Medical Science , Little Rock, AR , USA
| |
Collapse
|
28
|
Pont LMEB, Naipal K, Kloezeman JJ, Venkatesan S, van den Bent M, van Gent DC, Dirven CMF, Kanaar R, Lamfers MLM, Leenstra S. DNA damage response and anti-apoptotic proteins predict radiosensitization efficacy of HDAC inhibitors SAHA and LBH589 in patient-derived glioblastoma cells. Cancer Lett 2014; 356:525-35. [PMID: 25305451 DOI: 10.1016/j.canlet.2014.09.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/27/2014] [Accepted: 09/29/2014] [Indexed: 12/15/2022]
Abstract
HDAC inhibitors have radiosensitizing effects in established cancer cell lines. This study was conducted to compare the efficacy of SAHA, LBH589, Valproic Acid (VPA), MS275 and Scriptaid in the patient-derived glioblastoma model. In more detail, SAHA and LBH589 were evaluated to determine predictors of response. Acetylated-histone-H3, γH2AX/53BP1, (p)Chek2/ATM, Bcl-2/Bcl-XL, p21(CIP1/WAF1) and caspase-3/7 were studied in relation to response. SAHA sensitized 50% of cultures, LBH589 45%, VPA and Scriptaid 40% and MS275 60%. Differences after treatment with SAHA/RTx or LBH589/RTx in a sensitive and resistant culture were increased acetylated-H3, caspase-3/7 and prolonged DNA damage repair γH2AX/53BP1 foci. pChek2 was found to be associated with both SAHA/RTx and LBH589/RTx response with a positive predictive value (PPV) of 90%. Bcl-XL had a PPV of 100% for LBH589/RTx response. Incubation with HDACi 24 and 48 hours pre-RTx resulted in the best efficacy of combination treatment. In conclusion a subset of patient-derived glioblastoma cultures were sensitive to HDACi/RTx. For SAHA and LBH589 responses were strongly associated with pChek2 and Bcl-XL, which warrant further clinical exploration. Additional information on responsiveness was obtained by DNA damage response markers and apoptosis related proteins.
Collapse
Affiliation(s)
- Lotte M E Berghauser Pont
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Kishan Naipal
- Department of Genetics, Department Radiation Oncology, Cancer Genomics Netherlands, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jenneke J Kloezeman
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Subramanian Venkatesan
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Martin van den Bent
- Department of Neurology/Neuro-oncology, Brain Tumor Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Dik C van Gent
- Department of Genetics, Department Radiation Oncology, Cancer Genomics Netherlands, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Clemens M F Dirven
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Roland Kanaar
- Department of Genetics, Department Radiation Oncology, Cancer Genomics Netherlands, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Martine L M Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands; Department of Neurosurgery, Elizabeth Medical Hospital, Tilburg, The Netherlands.
| |
Collapse
|
29
|
Barazzuol L, Jeynes JCG, Merchant MJ, Wéra AC, Barry MA, Kirkby KJ, Suzuki M. Radiosensitization of glioblastoma cells using a histone deacetylase inhibitor (SAHA) comparing carbon ions with X-rays. Int J Radiat Biol 2014; 91:90-8. [DOI: 10.3109/09553002.2014.946111] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
30
|
Oike T, Ogiwara H, Amornwichet N, Nakano T, Kohno T. Chromatin-regulating proteins as targets for cancer therapy. JOURNAL OF RADIATION RESEARCH 2014; 55:613-28. [PMID: 24522270 PMCID: PMC4099987 DOI: 10.1093/jrr/rrt227] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Chromatin-regulating proteins represent a large class of novel targets for cancer therapy. In the context of radiotherapy, acetylation and deacetylation of histones by histone acetyltransferases (HATs) and histone deacetylases (HDACs) play important roles in the repair of DNA double-strand breaks generated by ionizing irradiation, and are therefore attractive targets for radiosensitization. Small-molecule inhibitors of HATs (garcinol, anacardic acid and curcumin) and HDACs (vorinostat, sodium butyrate and valproic acid) have been shown to sensitize cancer cells to ionizing irradiation in preclinical models, and some of these molecules are being tested in clinical trials, either alone or in combination with radiotherapy. Meanwhile, recent large-scale genome analyses have identified frequent mutations in genes encoding chromatin-regulating proteins, especially in those encoding subunits of the SWI/SNF chromatin-remodeling complex, in various human cancers. These observations have driven researchers toward development of targeted therapies against cancers carrying these mutations. DOT1L inhibition in MLL-rearranged leukemia, EZH2 inhibition in EZH2-mutant or MLL-rearranged hematologic malignancies and SNF5-deficient tumors, BRD4 inhibition in various hematologic malignancies, and BRM inhibition in BRG1-deficient tumors have demonstrated promising anti-tumor effects in preclinical models, and these strategies are currently awaiting clinical application. Overall, the data collected so far suggest that targeting chromatin-regulating proteins is a promising strategy for tomorrow's cancer therapy, including radiotherapy and molecularly targeted chemotherapy.
Collapse
Affiliation(s)
- Takahiro Oike
- Division of Genome Biology, National Cancer Center Research Institute, 1-1, Tsukiji 5-chome, Chuo-ku, Tokyo 104-0045, Japan Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hideaki Ogiwara
- Division of Genome Biology, National Cancer Center Research Institute, 1-1, Tsukiji 5-chome, Chuo-ku, Tokyo 104-0045, Japan
| | - Napapat Amornwichet
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan Department of Radiology, Chulalongkorn University, 1873, Rama 4 Road, Pathumwan, Bangkok 10330, Thailand
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, 1-1, Tsukiji 5-chome, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
31
|
Wiegmans AP, Al-Ejeh F, Chee N, Yap PY, Gorski JJ, Silva LD, Bolderson E, Chenevix-Trench G, Anderson R, Simpson PT, Lakhani SR, Khanna KK. Rad51 supports triple negative breast cancer metastasis. Oncotarget 2014; 5:3261-72. [PMID: 24811120 PMCID: PMC4102808 DOI: 10.18632/oncotarget.1923] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/25/2014] [Indexed: 01/05/2023] Open
Abstract
In contrast to extensive studies on familial breast cancer, it is currently unclear whether defects in DNA double strand break (DSB) repair genes play a role in sporadic breast cancer development and progression. We performed analysis of immunohistochemistry in an independent cohort of 235 were sporadic breast tumours. This analysis suggested that RAD51 expression is increased during breast cancer progression and metastasis and an oncogenic role for RAD51 when deregulated. Subsequent knockdown of RAD51 repressed cancer cell migration in vitro and reduced primary tumor growth in a syngeneic mouse model in vivo. Loss of RAD51 also inhibited associated metastasis not only in syngeneic mice but human xenografts and changed the metastatic gene expression profile of cancer cells, consistent with inhibition of distant metastasis. This demonstrates for the first time a new function of RAD51 that may underlie the proclivity of patients with RAD51 overexpression to develop distant metastasis. RAD51 is a potential biomarker and attractive drug target for metastatic triple negative breast cancer, with the capability to extend the survival of patients, which is less than 6 months.
Collapse
Affiliation(s)
- Adrian P Wiegmans
- QIMR Berghofer Medical Research Institute, Signal Transduction Laboratory, Herston Rd, Herston QLD 4006, Australia
| | - Fares Al-Ejeh
- QIMR Berghofer Medical Research Institute, Signal Transduction Laboratory, Herston Rd, Herston QLD 4006, Australia
| | - Nicole Chee
- QIMR Berghofer Medical Research Institute, Signal Transduction Laboratory, Herston Rd, Herston QLD 4006, Australia
| | - Pei-Yi Yap
- QIMR Berghofer Medical Research Institute, Signal Transduction Laboratory, Herston Rd, Herston QLD 4006, Australia
| | - Julia J Gorski
- Queens University Belfast, Dentistry and Biomedical Science, Lisburn Rd, Belfast, BT5 7BL, UK
| | - Leonard Da Silva
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, QLD 4006, Australia
- The University of Queensland, School of Medicine, Herston, Brisbane, QLD 4006, Australia
| | - Emma Bolderson
- The University of Queensland, Institute of Health and Biomedical Innovation, TRI, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Georgia Chenevix-Trench
- Cancer Genetics Laboratory, Queensland Institute of Medical Research, Herston Rd, Herston QLD 4006, Australia
| | - Robin Anderson
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne Vic 3002, Australia
- Department of Oncology, Sir Peter MacCallum Cancer Centre, The University of Melbourne, Parkville Vic 3052, Australia
| | - Peter T Simpson
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, QLD 4006, Australia
| | - Sunil R Lakhani
- The University of Queensland, UQ Centre for Clinical Research, Herston, Brisbane, QLD 4006, Australia
- The University of Queensland, School of Medicine, Herston, Brisbane, QLD 4006, Australia
- Pathology Queensland: The Royal Brisbane & Women's Hospital, Brisbane, Herston QLD 4006, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Signal Transduction Laboratory, Herston Rd, Herston QLD 4006, Australia
| |
Collapse
|
32
|
Konstantinopoulos PA, Wilson AJ, Saskowski J, Wass E, Khabele D. Suberoylanilide hydroxamic acid (SAHA) enhances olaparib activity by targeting homologous recombination DNA repair in ovarian cancer. Gynecol Oncol 2014; 133:599-606. [PMID: 24631446 DOI: 10.1016/j.ygyno.2014.03.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Approximately 50% of serous epithelial ovarian cancers (EOC) contain molecular defects in homologous recombination (HR) DNA repair pathways. Poly(ADP-ribose) polymerase inhibitors (PARPi) have efficacy in HR-deficient, but not in HR-proficient, EOC tumors as a single agent. Our goal was to determine whether the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), can sensitize HR-proficient ovarian cancer cells to the PARPi AZD-2281 (olaparib). METHODS Ovarian cancer cell lines (SKOV-3, OVCAR-8, NCI/ADR-Res, UWB1.289 BRCA1null and UWB1.289+BRCA1 wild-type) were treated with saline vehicle, olaparib, SAHA or olaparib/SAHA. Sulforhodamine B (SRB) assessed cytotoxicity and immunofluorescence and Western blot assays assessed markers of apoptosis (cleaved PARP) and DNA damage (pH2AX and RAD51). Drug effects were also tested in SKOV-3 xenografts in Nude mice. Affymetrix microarray experiments were performed in vehicle and SAHA-treated SKOV-3 cells. RESULTS In a microarray analysis, SAHA induced coordinated down-regulation of HR pathway genes, including RAD51 and BRCA1. Nuclear co-expression of RAD51 and pH2AX, a marker of efficient HR repair, was reduced approximately 40% by SAHA treatment alone and combined with olaparib. SAHA combined with olaparib induced apoptosis and pH2AX expression to a greater extent than either drug alone. Olaparib reduced cell viability at increasing concentrations and SAHA enhanced these effects in 4 of 5 cell lines, including BRCA1 null and wild-type cells, in vitro and in SKOV-3 xenografts in vivo. CONCLUSIONS These results provide preclinical rationale for targeting DNA damage response pathways by combining small molecule PARPi with HDACi as a mechanism for reducing HR efficiency in ovarian cancer.
Collapse
Affiliation(s)
- Panagiotis A Konstantinopoulos
- Department of Medical Oncology, Medical Gynecologic Oncology Program, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew J Wilson
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeanette Saskowski
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erica Wass
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dineo Khabele
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| |
Collapse
|
33
|
Treatment of nasopharyngeal carcinoma cells with the histone-deacetylase inhibitor abexinostat: cooperative effects with cis-platin and radiotherapy on patient-derived xenografts. PLoS One 2014; 9:e91325. [PMID: 24618637 PMCID: PMC3949989 DOI: 10.1371/journal.pone.0091325] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 02/09/2014] [Indexed: 01/02/2023] Open
Abstract
EBV-related nasopharyngeal carcinomas (NPCs) still raise serious therapeutic problems. The therapeutic potential of the histone-deacetylase (HDAC) inhibitor Abexinostat was investigated using 5 preclinical NPC models including 2 patient-derived xenografts (C15 and C17). The cytotoxicity of Abexinostat used either alone or in combination with cis-platin or irradiation was assessed in vitro by MTT and clonogenic assays using 2 EBV-negative (CNE1 and HONE1) and 3 EBV-positive NPC models (C15, C17 and C666-1). Subsequently, the 3 EBV-positive models were used under the form of xenografts to assess the impact of systemic treatments by Abexinostat or combinations of Abexinostat with cis-platin or irradiation. Several cell proteins known to be affected by HDAC inhibitors and the small viral non-coding RNA EBER1 were investigated in the treated tumors. Synergistic cytotoxic effects of Abexinostat combined with cis-platin or irradiation were demonstrated in vitro for each NPC model. When using xenografts, Abexinostat by itself (12.5 mg/kg, BID, 4 days a week for 3 weeks) had significant anti-tumor effects against C17. Cooperative effects with cis-platin (2 mg/kg, IP, at days 3, 10 and 17) and irradiation (1 Gy) were observed for the C15 and C17 xenografts. Simultaneously two types of biological alterations were induced in the tumor tissue, especially in the C17 model: a depletion of the DNA-repair protein RAD51 and a stronger in situ detection of the small viral RNA EBER1. Overall, these results support implementation of phase I/II clinical trials of Abexinostat for the treatment of NPC. A depletion of RAD51 is likely to contribute to the cooperation of Abexinostat with DNA damaging agents. Reduction of RAD51 combined to enhanced detection of EBER 1 might be helpful for early assessment of tumor response.
Collapse
|
34
|
Carvalho JFS, Kanaar R. Targeting homologous recombination-mediated DNA repair in cancer. Expert Opin Ther Targets 2014; 18:427-58. [PMID: 24491188 DOI: 10.1517/14728222.2014.882900] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION DNA is the target of many traditional non-specific chemotherapeutic drugs. New drugs or therapeutic approaches with a more rational and targeted component are mandatory to improve the success of cancer therapy. The homologous recombination (HR) pathway is an attractive target for the development of inhibitors because cancer cells rely heavily on HR for repair of DNA double-strand breaks resulting from chemotherapeutic treatments. Additionally, the discovery that poly(ADP)ribose polymerase-1 inhibitors selectively kill cells with genetic defects in HR has spurned an even greater interest in inhibitors of HR. AREAS COVERED HR drives the repair of broken DNA via numerous protein-mediated sequential DNA manipulations. Due to extensive number of steps and proteins involved, the HR pathway provides a rich pool of potential drug targets. This review discusses the latest developments concerning the strategies being explored to inhibit HR. Particular attention is given to the identification of small molecule inhibitors of key HR proteins, including the BRCA proteins and RAD51. EXPERT OPINION Current HR inhibitors are providing the basis for pharmaceutical development of more potent and specific inhibitors to be applied in mono- or combinatorial therapy regimes, while novel targets will be uncovered by experiments aimed to gain a deeper mechanistic understanding of HR and its subpathways.
Collapse
Affiliation(s)
- João F S Carvalho
- Erasmus MC Cancer Institute, Department of Genetics, Department of Radiation Oncology, Cancer Genomics Netherlands , PO Box 2040, 3000 CA Rotterdam , The Netherlands
| | | |
Collapse
|
35
|
Chen X, Radany EH, Wong P, Ma S, Wu K, Wang B, Wong JYC. Suberoylanilide hydroxamic acid induces hypersensitivity to radiation therapy in acute myelogenous leukemia cells expressing constitutively active FLT3 mutants. PLoS One 2013; 8:e84515. [PMID: 24367670 PMCID: PMC3868602 DOI: 10.1371/journal.pone.0084515] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/14/2013] [Indexed: 01/08/2023] Open
Abstract
Histone deacetylase inhibitors (HDIs) have shown promise as candidate radiosensitizer for many types of cancers. However, the mechanisms of action are not well understood, and whether they could have clinical impact on radiotherapy for leukemia is unclear. In this study, we demonstrate that suberoylanilide hydroxamic acid (SAHA) can increase radiosensitivity of acute myeloid leukemia (AML) cells through posttranslational modification of Rad51 protein responses and selective inhibition of the homology-directed repair (HDR) pathway. Our data also showed that AML cells with mutant, constitutively active FMS-like tyrosine kinase-3 (FLT3) were more radiation sensitive, caused by compromised non-homologous end joining (NHEJ) repair. Furthermore, SAHA-induced radiosensitization were enhanced in AML cells with expression of these FLT3 mutants. The results of this study suggest that SAHA, a recently approved HDI in clinical trials, may act as a candidate component for novel conditioning regimens to improve efficacy for AML patients undergoing radiotherapy and chemotherapy.
Collapse
MESH Headings
- Cell Line, Tumor
- DNA Damage
- DNA Repair/drug effects
- DNA Repair/radiation effects
- Enzyme Activation/drug effects
- Enzyme Activation/radiation effects
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/radiation effects
- Humans
- Hydroxamic Acids/pharmacology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/radiotherapy
- Mutation
- Protein Kinase C/metabolism
- Rad51 Recombinase/metabolism
- Radiation Tolerance/drug effects
- Vorinostat
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Xufeng Chen
- Department of Radiation Oncology, City of Hope Cancer Center, Duarte, California, United States of America
- Department of Radiation Oncology, The First People′s Hospital of Hangzhou Medical Group, Hangzhou, Zhejiang, China
| | - Eric H. Radany
- Department of Radiation Oncology, City of Hope Cancer Center, Duarte, California, United States of America
| | - Patty Wong
- Department of Radiation Oncology, City of Hope Cancer Center, Duarte, California, United States of America
| | - Shenglin Ma
- Department of Radiation Oncology, The First People′s Hospital of Hangzhou Medical Group, Hangzhou, Zhejiang, China
| | - Kan Wu
- Department of Radiation Oncology, The First People′s Hospital of Hangzhou Medical Group, Hangzhou, Zhejiang, China
| | - Bing Wang
- Department of Radiation Oncology, The First People′s Hospital of Hangzhou Medical Group, Hangzhou, Zhejiang, China
| | - Jeffrey Y. C. Wong
- Department of Radiation Oncology, City of Hope Cancer Center, Duarte, California, United States of America
| |
Collapse
|
36
|
Ladd B, Ackroyd JJ, Hicks JK, Canman CE, Flanagan SA, Shewach DS. Inhibition of homologous recombination with vorinostat synergistically enhances ganciclovir cytotoxicity. DNA Repair (Amst) 2013; 12:1114-21. [PMID: 24231389 DOI: 10.1016/j.dnarep.2013.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/03/2013] [Accepted: 10/21/2013] [Indexed: 01/28/2023]
Abstract
The nucleoside analog ganciclovir (GCV) elicits cytotoxicity in tumor cells via a novel mechanism in which drug incorporation into DNA produces minimal disruption of replication, but numerous DNA double strand breaks occur during the second S-phase after drug exposure. We propose that homologous recombination (HR), a major repair pathway for DNA double strand breaks, can prevent GCV-induced DNA damage, and that inhibition of HR will enhance cytotoxicity with GCV. Survival after GCV treatment in cells expressing a herpes simplex virus thymidine kinase was strongly dependent on HR (>14-fold decrease in IC50 in HR-deficient vs. HR-proficient CHO cells). In a homologous recombination reporter assay, the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA; vorinostat), decreased HR repair events up to 85%. SAHA plus GCV produced synergistic cytotoxicity in U251tk human glioblastoma cells. Elucidation of the synergistic mechanism demonstrated that SAHA produced a concentration-dependent decrease in the HR proteins Rad51 and CtIP. GCV alone produced numerous Rad51 foci, demonstrating activation of HR. However, the addition of SAHA blocked GCV-induced Rad51 foci formation completely and increased γH2AX, a marker of DNA double strand breaks. SAHA plus GCV also produced synergistic cytotoxicity in HR-proficient CHO cells, but the combination was antagonistic or additive in HR-deficient CHO cells. Collectively, these data demonstrate that HR promotes survival with GCV and compromise of HR by SAHA results in synergistic cytotoxicity, revealing a new mechanism for enhancing anticancer activity with GCV.
Collapse
Affiliation(s)
- Brendon Ladd
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
37
|
Maes K, Menu E, Van Valckenborgh E, Van Riet I, Vanderkerken K, De Bruyne E. Epigenetic modulating agents as a new therapeutic approach in multiple myeloma. Cancers (Basel) 2013; 5:430-61. [PMID: 24216985 PMCID: PMC3730337 DOI: 10.3390/cancers5020430] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) is an incurable B-cell malignancy. Therefore, new targets and drugs are urgently needed to improve patient outcome. Epigenetic aberrations play a crucial role in development and progression in cancer, including MM. To target these aberrations, epigenetic modulating agents, such as DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi), are under intense investigation in solid and hematological cancers. A clinical benefit of the use of these agents as single agents and in combination regimens has been suggested based on numerous studies in pre-clinical tumor models, including MM models. The mechanisms of action are not yet fully understood but appear to involve a combination of true epigenetic changes and cytotoxic actions. In addition, the interactions with the BM niche are also affected by epigenetic modulating agents that will further determine the in vivo efficacy and thus patient outcome. A better understanding of the molecular events underlying the anti-tumor activity of the epigenetic drugs will lead to more rational drug combinations. This review focuses on the involvement of epigenetic changes in MM pathogenesis and how the use of DNMTi and HDACi affect the myeloma tumor itself and its interactions with the microenvironment.
Collapse
Affiliation(s)
- Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Ivan Van Riet
- Stem Cell Laboratory, Department Clinical Hematology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussel, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| |
Collapse
|
38
|
Histone deacetylase inhibitors in the treatment for multiple myeloma. Int J Hematol 2013; 97:324-32. [DOI: 10.1007/s12185-013-1290-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/06/2013] [Indexed: 10/27/2022]
|