1
|
Li Y, Quan J, Song H, Li D, Ma E, Wang Y, Ma C. Novel pyrrolo[2,1-c][1,4]benzodiazepine-3,11-dione (PBD) derivatives as selective HDAC6 inhibitors to suppress tumor metastasis and invasion in vitro and in vivo. Bioorg Chem 2021; 114:105081. [PMID: 34153811 DOI: 10.1016/j.bioorg.2021.105081] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 11/30/2022]
Abstract
Selective inhibition of histone deacetylase 6 (HDAC6) has been emerged as a promising approach to cancer treatment. As a pivotal strategy for drug discovery,molecular hybridization was introduced in this study and a series of pyrrolo[2,1-c][1,4] benzodiazepine-3,11-diones (PBDs) based hydroxamic acids was rationally designed and synthesizedas novel selective HDAC6 inhibitors. Preliminary in vitro enzyme inhibition assay and structure-activity relationship (SAR) discussion confirmed our design strategy and met the expectation. Several of the compounds showed high potent against HDAC6 enzyme in vitro, and compound A7 with a long aliphatic linker was revealed to have the similar activity as the positive control tubastatin A. Further in vitro characterization of A7 demonstrates the metastasis inhibitory potency in MDA-MB-231 cell line and western blotting showed that A7 could induce the upregulation of Ac-α-tubulin, but not induce the excessive acetylation of histone H3, which indicated that the compound had HDAC6 targeting effect in MDA-MB-231 cells. In vivo study revealed that compound A7 has satisfactory inhibitory effects onliver and lung metastasis of breast cancer in mice. Molecular docking released that A7 could fit well with the receptor and interact with some key residues, which lays a foundation for further structural modifications to elucidate the interaction mode between compounds and target protein. This pharmacological investigation workflow provided a reasonable and reference methodto examine the pharmacological effects of inhibiting HDAC6 with a single molecule, either in vitro or in vivo. All of these results suggested that A7 is a promising lead compound that could lead to the further development of novel selective HDAC6 inhibitors for the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Yanchun Li
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Jishun Quan
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Haoxuan Song
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Dongzhu Li
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Enlong Ma
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yanjuan Wang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| |
Collapse
|
2
|
Ghiboub M, Elfiky AMI, de Winther MPJ, Harker NR, Tough DF, de Jonge WJ. Selective Targeting of Epigenetic Readers and Histone Deacetylases in Autoimmune and Inflammatory Diseases: Recent Advances and Future Perspectives. J Pers Med 2021; 11:336. [PMID: 33922725 PMCID: PMC8145108 DOI: 10.3390/jpm11050336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Histone deacetylases (HDACs) and bromodomain-containing proteins (BCPs) play a key role in chromatin remodeling. Based on their ability to regulate inducible gene expression in the context of inflammation and cancer, HDACs and BCPs have been the focus of drug discovery efforts, and numerous small-molecule inhibitors have been developed. However, dose-limiting toxicities of the first generation of inhibitors, which typically target multiple HDACs or BCPs, have limited translation to the clinic. Over the last decade, an increasing effort has been dedicated to designing class-, isoform-, or domain-specific HDAC or BCP inhibitors, as well as developing strategies for cell-specific targeted drug delivery. Selective inhibition of the epigenetic modulators is helping to elucidate the functions of individual epigenetic proteins and has the potential to yield better and safer therapeutic strategies. In accordance with this idea, several in vitro and in vivo studies have reported the ability of more selective HDAC/BCP inhibitors to recapitulate the beneficial effects of pan-inhibitors with less unwanted adverse events. In this review, we summarize the most recent advances with these strategies, discussing advantages and limitations of these approaches as well as some therapeutic perspectives, focusing on autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Centers, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (M.G.); (A.M.I.E.)
- Adaptive Immunity Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage SG1 2NY, UK; (N.R.H.); (D.F.T.)
| | - Ahmed M. I. Elfiky
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Centers, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (M.G.); (A.M.I.E.)
- Adaptive Immunity Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage SG1 2NY, UK; (N.R.H.); (D.F.T.)
| | - Menno P. J. de Winther
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Department of Medicine, Institute for Cardiovascular Prevention (IPEK), 80336 Munich, Germany
| | - Nicola R. Harker
- Adaptive Immunity Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage SG1 2NY, UK; (N.R.H.); (D.F.T.)
| | - David F. Tough
- Adaptive Immunity Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage SG1 2NY, UK; (N.R.H.); (D.F.T.)
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Centers, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (M.G.); (A.M.I.E.)
- Department of Surgery, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
3
|
Ueno M, Morizane C, Furukawa M, Sakai D, Komatsu Y, Nakai Y, Tsuda M, Ozaka M, Mizuno N, Muto M, Fukutomi A, Ikeda M, Tsuji A, Katanuma A, Moriwaki T, Kajiwara T, Ishii H, Negoro Y, Shimizu S, Nemoto N, Kobayashi S, Makino K, Furuse J. A randomized, double-blind, phase II study of oral histone deacetylase inhibitor resminostat plus S-1 versus placebo plus S-1 in biliary tract cancers previously treated with gemcitabine plus platinum-based chemotherapy. Cancer Med 2021; 10:2088-2099. [PMID: 33635605 PMCID: PMC7957161 DOI: 10.1002/cam4.3813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Effective second-line chemotherapy options are limited in treating advanced biliary tract cancers (BTCs). Resminostat is an oral histone deacetylase inhibitor. Such inhibitors increase sensitivity to fluorouracil, the active form of S-1. In the phase I study, addition of resminostat to S-1 was suggested to have promising efficacy for pre-treated BTCs. This study investigated the efficacy and safety of resminostat plus S-1 in second-line therapy for BTCs. METHODS Patients were randomly assigned to receive resminostat or placebo (200 mg orally per day; days 1-5 and 8-12) and S-1 group (80-120 mg orally per day by body surface area; days 1-14) over a 21-day cycle. The primary endpoint was progression-free survival (PFS). Secondary endpoints comprised overall survival (OS), response rate (RR), disease control rate (DCR), and safety. RESULTS Among 101 patients enrolled, 50 received resminostat+S-1 and 51 received placebo+S-1. Median PFS was 2.9 months for resminostat+S-1 vs. 3.0 months for placebo+S-1 (HR: 1.154, 95% CI: 0.759-1.757, p = 0.502); median OS was 7.8 months vs. 7.5 months, respectively (HR: 1.049, 95% CI: 0.653-1.684, p = 0.834); the RR and DCR were 6.0% vs. 9.8% and 70.0% vs. 78.4%, respectively. Treatment-related adverse events (TrAEs) of grade ≥ 3 occurring more frequently (≥10% difference) in the resminostat+S-1 than in the placebo+S-1 comprised platelet count decreased (18.0% vs. 2.0%) and decreased appetite (16.0% vs. 2.0%). CONCLUSIONS Resminostat plus S-1 therapy improved neither PFS nor OS for patients with pre-treated BTCs. Addition of resminostat to S-1 was associated with higher incidence of TrAEs, but these were manageable (JapicCTI-183883).
Collapse
Affiliation(s)
- Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology DivisionKanagawa Cancer CenterKanagawaJapan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic OncologyNational Cancer Center HospitalTokyoJapan
| | - Masayuki Furukawa
- Department of Hepato‐Biliary‐PancreatologyNational Hospital Organization Kyushu Cancer CenterFukuokaJapan
| | - Daisuke Sakai
- Department of Frontier Science for Cancer and ChemotherapyOsaka University Graduate School of MedicineOsakaJapan
| | - Yoshito Komatsu
- Division of Cancer CenterHokkaido University HospitalHokkaidoJapan
| | - Yousuke Nakai
- Department of GastroenterologyDepartment of Endoscopy and Endoscopic SurgeryGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Masahiro Tsuda
- Department of Gastroenterological OncologyHyogo Cancer CenterHyogoJapan
| | - Masato Ozaka
- Hepato‐Biliary‐Pancreatic Medicine DepartmentCancer Institute Hospital of the Japanese Foundation for Cancer ResearchTokyoJapan
| | - Nobumasa Mizuno
- Department of GastroenterologyAichi Cancer Center HospitalAichiJapan
| | - Manabu Muto
- Department of Clinical OncologyKyoto University HospitalKyotoJapan
| | - Akira Fukutomi
- Division of Gastrointestinal OncologyShizuoka Cancer CenterShizuokaJapan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic OncologyNational Cancer Center Hospital EastChibaJapan
| | - Akihito Tsuji
- Department of Clinical OncologyFaculty of medicineKagawa UniversityKagawaJapan
| | - Akio Katanuma
- Center for GastroenterologyTeine‐keijinkai hospitalHokkaidoJapan
| | - Toshikazu Moriwaki
- Department of GastroenterologyFaculty of MedicineUniversity of TsukubaIbarakiJapan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical OncologyNational Hospital Organization Shikoku Cancer CenterEhimeJapan
| | - Hiroshi Ishii
- Clinical Research CenterChiba Cancer CenterChibaJapan
| | - Yuji Negoro
- Division of Clinical OncologyKochi Health Sciences CenterKochiJapan
| | - Satoshi Shimizu
- Department of GastroenterologySaitama Cancer CenterSaitamaJapan
| | - Noriko Nemoto
- Pharmaceutical Research & Development DepartmentYakult Honsha Co., Ltd.TokyoJapan
| | - Shingo Kobayashi
- Pharmaceutical Research & Development DepartmentYakult Honsha Co., Ltd.TokyoJapan
| | - Keigo Makino
- Pharmaceutical Research & Development DepartmentYakult Honsha Co., Ltd.TokyoJapan
| | - Junji Furuse
- Department of Medical OncologyFaculty of MedicineKyorin UniversityTokyoJapan
| |
Collapse
|
4
|
Stadler R, Scarisbrick JJ. Maintenance therapy in patients with mycosis fungoides or Sézary syndrome: A neglected topic. Eur J Cancer 2020; 142:38-47. [PMID: 33217680 DOI: 10.1016/j.ejca.2020.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 11/27/2022]
Abstract
In advanced-stage cutaneous T-cell lymphoma (CTCL), the current therapeutic options rarely provide long-lasting responses, leaving allogenic stem-cell transplantation the only potentially curative option for highly selected patients. Until today, there are no standardised strategies for maintenance therapy in patients with CTCL who achieved disease control. Moreover, evidence-based treatment options or drugs that are approved for maintenance treatment in advanced stages after remission induction are still lacking. Patients require maintenance options that provide a good safety profile, are convenient to apply and do not negatively affect their health-related quality of life. However, carrying out large-scale, controlled studies is challenging in CTCL. In addition to information on the concept of maintenance therapy, this review provides an update on current and emerging approaches that target maintenance treatment in advanced-stage CTCL. After all, the group of potentially interesting maintenance therapy candidates, especially for patients in advanced stage, includes not only immunomodulating and phototherapeutic modalities that have been used already for many decades but also newer systemic therapies, including epigenetic modifiers.
Collapse
Affiliation(s)
- Rudolf Stadler
- University Clinic for Dermatology, Johannes Wesling Medical Centre, Minden, Germany.
| | | |
Collapse
|
5
|
Arai S, Takeuchi S, Fukuda K, Tanimoto A, Nishiyama A, Konishi H, Takagi A, Takahashi H, Ong ST, Yano S. Resminostat, a histone deacetylase inhibitor, circumvents tolerance to EGFR inhibitors in EGFR-mutated lung cancer cells with BIM deletion polymorphism. THE JOURNAL OF MEDICAL INVESTIGATION 2020; 67:343-350. [PMID: 33148913 DOI: 10.2152/jmi.67.343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Drug-tolerant cells are mediators of acquired resistance. BIM-intron2 deletion polymorphism (BIM-del) is one of the mechanisms underlying the resistance to epidermal growth factor tyrosine kinase inhibitor (EGFR-TKI)-mediated apoptosis that induces drug tolerance. Here, we investigated whether resminostat, a histone deacetylase inhibitor, circumvents BIM-del-associated apoptosis resistance. The human EGFR-mutated non-small cell lung cancer (NSCLC) cell line PC-9 and its homozygous BIM-del-positive variant (PC-9 BIMi2- / -), established by editing with zinc finger nuclease, were used. In comparison with PC-9 cells, PC-9 BIMi2- / - cells were less sensitive to apoptosis mediated by EGFR-TKIs such as gefitinib and osimertinib. The combined use of resminostat and an EGFR-TKI preferentially induced the expression of the pro-apoptotic BIM transcript containing exon 4 rather than that containing exon 3, increased the level of pro-apoptotic BIM protein (BIMEL), and stimulated apoptosis in vitro. In a subcutaneous tumor model derived from PC-9 BIMi2- / - cells, gefitinib monotherapy decreased tumor size but retained residual lesions, indicative of the presence of tolerant cells in tumors. The combined use of resminostat and gefitinib increased BIMEL protein level and induced apoptosis, subsequently leading to the remarkable shrinkage of tumor. These findings suggest the potential of resminostat to circumvent tolerance to EGFR-TKIs associated with BIM deletion polymorphism. J. Med. Invest. 67 : 343-350, August, 2020.
Collapse
Affiliation(s)
- Sachiko Arai
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Shinji Takeuchi
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Koji Fukuda
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Azusa Tanimoto
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Akihiro Nishiyama
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Konishi
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Akimitsu Takagi
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | | | - S Tiong Ong
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore.,Department of Haematology, Singapore General Hospital, Singapore.,Department of Medical Oncology, National Cancer Centre Singapore, Singapore.,Department of Medicine, Duke University Medical Center, Durham, NC, United States of America
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
6
|
Inhibition of histone deacetylases, topoisomerases and epidermal growth factor receptor by metal-based anticancer agents: Design & synthetic strategies and their medicinal attributes. Bioorg Chem 2020; 105:104396. [PMID: 33130345 DOI: 10.1016/j.bioorg.2020.104396] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/22/2022]
Abstract
Metal-based inhibitors of histone deacetylases (HDAC), DNA topoisomerases (Topos) and Epidermal Growth Factor Receptor (EGFR) have demonstrated their cytotoxic potential against various cancer types such as breast, lung, uterus, colon, etc. Additionally, these have proven their role in resolving the resistance issues, enhancing the affinity, lipophilicity, stability, and biocompatibility and therefore, emerged as potential candidates for molecularly targeted therapeutics. This review focusses on nature and role of metals and organic ligands in tuning the anticancer activity in multiple modes of inhibition considering HDACs, Topos or EGFR as one of the primary targets. The conceptual design and synthetic approaches of platinum and non-platinum metal complexes comprising of chiefly ruthenium, rhodium, palladium, copper, iron, nickel, cobalt, zinc metals coordinated with organic scaffolds, along with their biological activity profiles, structure-activity relationships (SARs), docking studies, possible modes of action, and their scope and limitations are discussed in detail.
Collapse
|
7
|
Abstract
Histone deacetylases (HDACs) are expressed at increased levels in cells of various malignancies, and the use of HDAC inhibitors has improved outcomes in patients with haematological malignancies (T-cell lymphomas and multiple myeloma). However, they are not as effective in solid tumours. Five agents are currently approved under various jurisdictions, namely belinostat, chidamide, panobinostat, romidepsin and vorinostat. These agents are associated with a range of class-related and agent-specific serious and/or severe adverse effects, notably myelosuppression, diarrhoea and various cardiac effects. Among the cardiac effects are ST-T segment abnormalities and QTc interval prolongation of the electrocardiogram, isolated cases of atrial fibrillation and, in rare instances, ventricular tachyarrhythmias. In order to improve the safety profile of this class of drugs as well as their efficacy in indications already approved and to further widen their indications, a large number of newer HDAC inhibitors with varying degrees of HDAC isoform selectivity have been synthesised and are currently under clinical development. Preliminary evidence from early studies suggests that they may be effective in non-haematological cancers as well when used in combination with other therapeutic modalities, but that they too appear to be associated with the above class-related adverse effects. As the database accumulates, the safety, efficacy and risk/benefit of the newer agents and their indications will become clearer.
Collapse
|
8
|
Phase I/II study of first-line combination therapy with sorafenib plus resminostat, an oral HDAC inhibitor, versus sorafenib monotherapy for advanced hepatocellular carcinoma in east Asian patients. Invest New Drugs 2018; 36:1072-1084. [PMID: 30198057 DOI: 10.1007/s10637-018-0658-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Resminostat is an oral inhibitor of class I, IIB, and IV histone deacetylases. This phase I/II study compared the safety and efficacy of resminostat plus sorafenib versus sorafenib monotherapy as first-line therapy for advanced hepatocellular carcinoma (HCC). EXPERIMENTAL DESIGN In phase I, resminostat (400 mg or 600 mg/day on days 1 to 5 every 14 days) was administered with sorafenib (800 mg/day for 14 days) to determine the recommended dose for phase II. In phase II, patients were randomized (1:1) to sorafenib monotherapy or resminostat plus sorafenib. The primary endpoint was time-to-progression (TTP). RESULTS Nine patients (3: 400 mg, 6: 600 mg) were enrolled in phase I, and the recommended dose of resminostat was determined to be 400 mg/day. Then 170 patients were enrolled in phase II. Median TTP/overall survival (OS) were 2.8/14.1 months with monotherapy versus 2.8/11.8 months with combination therapy (Hazard Ratio [HR]: 0.984, p = 0.925/HR: 1.046, p = 0.824). The overall incidence of adverse events was similar in both groups (98.8% versus 100.0%). However, thrombocytopenia ≥ Grade 3 was significantly more frequent in the combination therapy group (34.5% versus 2.4%, p < 0.001). Subgroup analysis revealed that median TTP/OS was 1.5/6.9 months for monotherapy versus 2.8/13.1 months for combination therapy (HR: 0.795, p = 0.392/HR: 0.567, p = 0.065) among patients with a normal-to-high baseline platelet count (≥ 150 × 103/mm3). CONCLUSIONS In patients with advanced HCC, first-line therapy with resminostat at the recommended dose plus sorafenib showed no significant efficacy advantage over sorafenib monotherapy.
Collapse
|
9
|
Walewski J, Paszkiewicz-Kozik E, Borsaru G, Hellmann A, Janikova A, Warszewska A, Mais A, Ammendola A, Herz T, Krauss B, Henning SW. Resminostat in patients with relapsed or refractory Hodgkin lymphoma: results of the phase II SAPHIRE study. Leuk Lymphoma 2018; 60:675-684. [DOI: 10.1080/10428194.2018.1492122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jan Walewski
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | - Ewa Paszkiewicz-Kozik
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | | | - Andrzej Hellmann
- Department for Hematology and Transplantology, University Clinical Centre, Medical University of Gdansk, Gdansk, Poland
| | - Andrea Janikova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Agnieszka Warszewska
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | - Anna Mais
- 4SC AG, Martinsried, Planegg, Germany
| | | | | | | | | |
Collapse
|
10
|
Alaibac M. Small molecule inhibitors for cutaneous T-cell lymphomas. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1469004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Mauro Alaibac
- Unit of Dermatology, Department of Dermatology, University of Padua, Padua, Italy
| |
Collapse
|
11
|
Resminostat induces changes in epithelial plasticity of hepatocellular carcinoma cells and sensitizes them to sorafenib-induced apoptosis. Oncotarget 2017; 8:110367-110379. [PMID: 29299154 PMCID: PMC5746389 DOI: 10.18632/oncotarget.22775] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 11/17/2017] [Indexed: 01/16/2023] Open
Abstract
Resminostat, a novel class I, IIb, and IV histone deacetylase inhibitor, was studied in advanced hepatocellular carcinoma (HCC) patients after relapse to sorafenib (SHELTER study). In this phase I/II clinical trial, combination of sorafenib and resminostat was safe and showed early signs of efficacy. However, the molecular mechanisms behind this synergism have not been explored yet. In this work, we aimed to analyze whether resminostat regulates epithelial-mesenchymal and stemness phenotype as a mechanism of sensitization to sorafenib. Three HCC cell lines with differences in their epithelial/mesenchymal characteristics were treated with resminostat and sorafenib alone, or in combination. Resminostat prevented growth and induced cell death in the HCC cells, in a time and dose dependent manner. A collaborative effect between resminostat and sorafenib was detected in the mesenchymal HCC cells, which were insensitive to sorafenib-induced apoptosis. Expression of mesenchymal-related genes was decreased in resminostat-treated HCC cells, concomitant with an increase in epithelial-related gene expression, organized tight junctions and reduced invasive growth. Moreover, resminostat down-regulated CD44 expression, coincident with decreased capacity to form colonies at low cell density. CONCLUSION Resminostat shifts mesenchymal cells towards a more epithelial phenotype, lower invasive and stemness properties, which may contribute to the sensitization to sorafenib-induced apoptosis.
Collapse
|
12
|
Tambo Y, Hosomi Y, Sakai H, Nogami N, Atagi S, Sasaki Y, Kato T, Takahashi T, Seto T, Maemondo M, Nokihara H, Koyama R, Nakagawa K, Kawaguchi T, Okamura Y, Nakamura O, Nishio M, Tamura T. Phase I/II study of docetaxel combined with resminostat, an oral hydroxamic acid HDAC inhibitor, for advanced non-small cell lung cancer in patients previously treated with platinum-based chemotherapy. Invest New Drugs 2017; 35:217-226. [PMID: 28138828 DOI: 10.1007/s10637-017-0435-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/24/2017] [Indexed: 12/28/2022]
Abstract
Objectives To determine the recommended dose and efficacy/safety of docetaxel combined with resminostat (DR) in non-small cell lung cancer (NSCLC) patients with previous platinum-based chemotherapy. Materials and Methods A multicenter, open-label, phase I/II study was performed in Japanese patients with stage IIIB/IV or recurrent NSCLC and prior platinum-based chemotherapy. The recommended phase II dose was determined using a standard 3 + 3 dose design in phase I part. Resminostat was escalated from 400 to 600 mg/day and docetaxel fixed at 75 mg/m2. In phase II part, the patients were randomly assigned to docetaxel alone (75 mg/m2) or DR therapy. Docetaxel was administered on day 1 and resminostat on days 1-5 in the DR group. Treatment was repeated every 21 days until progression or unacceptable toxicity. The primary endpoint was progression-free survival (PFS). Results A total of 117 patients (phase I part, 9; phase II part, 108) were enrolled. There was no dose-limiting toxicity in phase I part; the recommended dose for resminostat was 600 mg/day with 75 mg/m2 of docetaxel. In phase II part, median PFS (95% confidence interval [CI]) was 4.2 (2.8-5.7) months with docetaxel group and 4.1 (1.5-5.4) months with DR group (hazard ratio [HR]: 1.354, 95% CI: 0.835-2.195; p = 0.209). Grade ≥ 3 adverse events significantly more common with DR group than docetaxel group were leukopenia, febrile neutropenia, thrombocytopenia, and anorexia. Conclusion In Japanese NSCLC patients previously treated with platinum-based chemotherapy, DR therapy did not improve PFS compared with docetaxel alone and increased toxicity.
Collapse
Affiliation(s)
- Yuichi Tambo
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Hiroshi Sakai
- Division of Thoracic Oncology, Saitama Cancer Center, Saitama, Japan
| | - Naoyuki Nogami
- Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Shinji Atagi
- Department of Thoracic Oncology, National Hospital Organization Kinki-chuo Chest Medical Center, Osaka, Japan
| | - Yasutsuna Sasaki
- Division of Medical Oncology, Showa University School of Medicine, Tokyo, Japan
| | - Terufumi Kato
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Kanagawa, Japan
| | | | - Takashi Seto
- Department of Thoracic Oncology, National Kyushu Cancer Center, Fukuoka, Japan
| | - Makoto Maemondo
- Department of Respiratory Medicine, Miyagi Cancer Center, Miyagi, Japan
| | - Hiroshi Nokihara
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ryo Koyama
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Respiratory Medicine, Osaka City University Hospital, Osaka, Japan
| | - Yuta Okamura
- Pharmaceutical Research and Development Department, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Osamu Nakamura
- Pharmaceutical Research and Development Department, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Tomohide Tamura
- Thoracic Center, St. Luke's International Hospital, Tokyo, Japan
| |
Collapse
|
13
|
Ellerhoff TP, Berchtold S, Venturelli S, Burkard M, Smirnow I, Wulff T, Lauer UM. Novel epi-virotherapeutic treatment of pancreatic cancer combining the oral histone deacetylase inhibitor resminostat with oncolytic measles vaccine virus. Int J Oncol 2016; 49:1931-1944. [DOI: 10.3892/ijo.2016.3675] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/22/2016] [Indexed: 11/05/2022] Open
|
14
|
Frühwald MC, Biegel JA, Bourdeaut F, Roberts CWM, Chi SN. Atypical teratoid/rhabdoid tumors-current concepts, advances in biology, and potential future therapies. Neuro Oncol 2016; 18:764-78. [PMID: 26755072 DOI: 10.1093/neuonc/nov264] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 09/27/2015] [Indexed: 01/05/2023] Open
Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is the most common malignant CNS tumor of children below 6 months of age. The majority of AT/RTs demonstrate genomic alterations in SMARCB1 (INI1, SNF5, BAF47) or, to a lesser extent, SMARCA4 (BRG1) of the SWItch/sucrose nonfermentable chromatin remodeling complex. Recent transcription and methylation profiling studies suggest the existence of molecular subgroups. Thus, at the root of these seemingly enigmatic tumors lies a network of factors related to epigenetic regulation, which is not yet completely understood. While conventional-type chemotherapy may have significant survival benefit for certain patients, it remains to be determined which patients will eventually prove resistant to chemotherapy and thus need novel therapeutic strategies. Elucidation of the molecular consequences of a disturbed epigenome has led to the identification of a series of transduction cascades, which may be targeted for therapy. Among these are the pathways of cyclin D1/cyclin-dependent kinases 4 and 6, Hedgehog/GLI1, Wnt/ß-catenin, enhancer of zeste homolog 2, and aurora kinase A, among others. Compounds specifically targeting these pathways or agents that alter the epigenetic state of the cell are currently being evaluated in preclinical settings and in experimental clinical trials for AT/RT.
Collapse
Affiliation(s)
- Michael C Frühwald
- Children's Hospital and Swabian Children's Cancer Center, Augsburg, Germany (M.C.F.); Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California (J.A.B.); INSERM U830, Laboratory of Genetics and Biology of Cancers, and Department of Pediatric Oncology, Curie Institute, Paris, France (F.B.); Comprehensive Cancer Center and Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee (C.W.M.R.); Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.N.C.); Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts (S.N.C.); Department of Pediatrics, Harvard Medical School, Boston, Massachusetts (S.N.C.)
| | - Jaclyn A Biegel
- Children's Hospital and Swabian Children's Cancer Center, Augsburg, Germany (M.C.F.); Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California (J.A.B.); INSERM U830, Laboratory of Genetics and Biology of Cancers, and Department of Pediatric Oncology, Curie Institute, Paris, France (F.B.); Comprehensive Cancer Center and Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee (C.W.M.R.); Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.N.C.); Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts (S.N.C.); Department of Pediatrics, Harvard Medical School, Boston, Massachusetts (S.N.C.)
| | - Franck Bourdeaut
- Children's Hospital and Swabian Children's Cancer Center, Augsburg, Germany (M.C.F.); Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California (J.A.B.); INSERM U830, Laboratory of Genetics and Biology of Cancers, and Department of Pediatric Oncology, Curie Institute, Paris, France (F.B.); Comprehensive Cancer Center and Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee (C.W.M.R.); Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.N.C.); Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts (S.N.C.); Department of Pediatrics, Harvard Medical School, Boston, Massachusetts (S.N.C.)
| | - Charles W M Roberts
- Children's Hospital and Swabian Children's Cancer Center, Augsburg, Germany (M.C.F.); Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California (J.A.B.); INSERM U830, Laboratory of Genetics and Biology of Cancers, and Department of Pediatric Oncology, Curie Institute, Paris, France (F.B.); Comprehensive Cancer Center and Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee (C.W.M.R.); Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.N.C.); Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts (S.N.C.); Department of Pediatrics, Harvard Medical School, Boston, Massachusetts (S.N.C.)
| | - Susan N Chi
- Children's Hospital and Swabian Children's Cancer Center, Augsburg, Germany (M.C.F.); Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California (J.A.B.); INSERM U830, Laboratory of Genetics and Biology of Cancers, and Department of Pediatric Oncology, Curie Institute, Paris, France (F.B.); Comprehensive Cancer Center and Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee (C.W.M.R.); Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.N.C.); Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts (S.N.C.); Department of Pediatrics, Harvard Medical School, Boston, Massachusetts (S.N.C.)
| |
Collapse
|
15
|
Shen S, Kozikowski AP. Why Hydroxamates May Not Be the Best Histone Deacetylase Inhibitors--What Some May Have Forgotten or Would Rather Forget? ChemMedChem 2015; 11:15-21. [PMID: 26603496 DOI: 10.1002/cmdc.201500486] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Indexed: 12/21/2022]
Abstract
Hydroxamate-based histone deacetylase inhibitors (HDACIs) have been approved as therapeutic agents by the US Food and Drug Administration for use in oncology applications. While the potential utility of such HDACIs in other areas of medicinal chemistry is tremendous, there are significant concerns that "pan-HDAC inhibitors" may be too broadly acting and/or toxic for clinical use beyond oncology. In addition to the isozyme selectivity challenge, the potential mutagenicity of hydroxamate-containing HDAC inhibitors represents a major hindrance in their application to other therapeutic areas. Herein we report on the mutagenicity of known hydroxamates, discuss the mechanisms responsible for their genotoxicity, and review some of the current alternatives to hydroxamates. We conclude that the hydroxamate group, while providing high-potency HDACIs, is not necessarily the best zinc-binding group for HDACI drug discovery.
Collapse
Affiliation(s)
- Sida Shen
- Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Alan P Kozikowski
- Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|