1
|
Budillon A, Leone A, Passaro E, Silvestro L, Foschini F, Iannelli F, Roca MS, Macchini M, Bruzzese F, Garcia Bermejo ML, Rodriguez Garrote M, Tortora G, Milella M, Reni M, Fuchs C, Hewitt E, Kubiak C, Di Gennaro E, Giannarelli D, Avallone A. Randomized phase 2 study of valproic acid combined with simvastatin and gemcitabine/nab-paclitaxel-based regimens in untreated metastatic pancreatic adenocarcinoma patients: the VESPA trial study protocol. BMC Cancer 2024; 24:1167. [PMID: 39300376 PMCID: PMC11414294 DOI: 10.1186/s12885-024-12936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Metastatic pancreatic ductal adenocarcinoma (mPDAC) patients have very poor prognosis highlighting the urgent need of novel treatments. In this regard, repurposing non-oncology already-approved drugs might be an attractive strategy to offer more-effective treatment easily tested in clinical trials. Accumulating evidence suggests that epigenetic deregulation is a hallmark of cancer contributing to treatment resistance in several solid tumors, including PDAC. Histone deacetylase inhibitors (HDACi) are epigenetic drugs we have investigated preclinically and clinically as anticancer agents. Valproic acid (VPA) is a generic low-cost anticonvulsant and mood stabilizer with HDAC inhibitory activity, and anticancer properties also demonstrated in PDAC models. Statins use was reported to be associated with lower mortality risk in patients with pancreatic cancer and statins have been shown to have a direct antitumor effect when used alone or in combination therapy. We recently showed capability of VPA/Simvastatin (SIM) combination to potentiate the antitumor activity of gemcitabine/nab-paclitaxel in vitro and in vivo PDAC preclinical models. METHODS/DESIGN VESPA is a patient-centric open label randomized multicenter phase-II investigator-initiated trial, evaluating the feasibility, safety, and efficacy of VPA/SIM plus first line gemcitabine/nab-paclitaxel-based regimens (AG or PAXG) (experimental arm) versus chemotherapy alone (standard arm) in mPDAC patients. The study involves Italian and Spanish oncology centers and includes an initial 6-patients safety run-in-phase. A sample size of 240 patients (120 for each arm) was calculated under the hypothesis that the addition of VPA/SIM to gemcitabine and nab-paclitaxel-based regimens may extend progression free survival from 6 to 9 months in the experimental arm. Secondary endpoints are overall survival, response rate, disease control rate, duration of response, CA 19.9 reduction, toxicity, and quality of life. The study includes a patient engagement plan and complementary biomarkers studies on tumor and blood samples. CONCLUSIONS VESPA is the first trial evaluating efficacy and safety of two repurposed drugs in oncology such as VPA and SIM, in combination with standard chemotherapy, with the aim of improving mPDAC survival. The study is ongoing. Enrollment started in June 2023 and a total of 63 patients have been enrolled as of June 2024. TRIAL REGISTRATION EudraCT number: 2022-004154-63; ClinicalTrials.gov identifier NCT05821556, posted 2023/04/20.
Collapse
Affiliation(s)
- Alfredo Budillon
- Scientific Directorate, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy.
| | - Alessandra Leone
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Eugenia Passaro
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Lucrezia Silvestro
- Experimental Clinical Abdominal Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Francesca Foschini
- Experimental Clinical Abdominal Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Federica Iannelli
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Marina Macchini
- Department of Medical Oncology, University "Vita-Salute San Raffaele", IRCCS- Ospedale San Raffaele, Milan, Italy
| | - Francesca Bruzzese
- Animal Facility Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Maria Laura Garcia Bermejo
- Biomarkers and Therapeutic Targets Group, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Mercedes Rodriguez Garrote
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Giampaolo Tortora
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Michele Milella
- Section of Innovation Biomedicines-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Michele Reni
- Department of Medical Oncology, University "Vita-Salute San Raffaele", IRCCS- Ospedale San Raffaele, Milan, Italy
| | | | - Eve Hewitt
- Beacon: for rare diseases, Cambridge, UK
| | - Christine Kubiak
- ECRIN - European Clinical Research Infrastructure Network-European Research Infrastructure Consortium, Paris, France
| | - Elena Di Gennaro
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Diana Giannarelli
- Facility of Epidemiology and Biostatistics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy.
| |
Collapse
|
2
|
Hunek G, Zembala J, Januszewski J, Bełżek A, Syty K, Jabiry-Zieniewicz Z, Ludwin A, Flieger J, Baj J. Micro- and Macronutrients in Endometrial Cancer-From Metallomic Analysis to Improvements in Treatment Strategies. Int J Mol Sci 2024; 25:9918. [PMID: 39337406 PMCID: PMC11432114 DOI: 10.3390/ijms25189918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
Endometrial cancer is reported to be one of the most prevalent cancers of the female reproductive organs worldwide, with increasing incidence and mortality rates over the past decade. Early diagnosis is critical for effective treatment. Recently, there has been a growing focus on the role of nutrition and micronutrient and macronutrient status in patients with gynecologic cancers, including endometrial cancer. In the following paper, we have conducted an in-depth narrative literature review with the aim of evaluating the results of metallomic studies specifically concerning the micro- and macronutrient status of patients with endometrial cancer. The main objective of the paper was to analyze the results regarding the nutritional status of endometrial cancer patients and describe the role of chosen elements in the onset and progression of endometrial carcinogenesis. Further, we have focused on the evaluation of the usage of the described elements in the potential treatment of the abovementioned cancer, as well as the possible prevention of cancer considering proper supplementation of chosen elements in healthy individuals. Calcium supplementation has been proposed to reduce the risk of endometrial cancer, although some studies offer conflicting evidence. Deficiencies in phosphorus, selenium, and zinc have been inversely associated with endometrial cancer risk, suggesting they may play a protective role, whereas excessive levels of iron, copper, and cadmium have been positively correlated with increased risk. However, the molecular mechanisms by which these elements affect endometrial carcinogenesis are not fully understood, and current findings are often contradictory. Further research is needed to clarify these relationships and to evaluate the potential of nutritional interventions for the prevention and treatment of endometrial cancer.
Collapse
Affiliation(s)
- Gabriela Hunek
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Julita Zembala
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Jacek Januszewski
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Aleksandra Bełżek
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Kinga Syty
- Institute of Health Sciences, John Paul the II Catholic University of Lublin, Konstantynów 1G, 20-708 Lublin, Poland
| | - Zoulikha Jabiry-Zieniewicz
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Artur Ludwin
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| |
Collapse
|
3
|
Salamanca-Ortiz H, Domínguez-Gomez G, Chávez-Blanco A, Ortega-Bernal D, Díaz-Chávez J, González-Fierro A, Candelaria-Hernández M, Dueñas-González A. The inhibitory and transcriptional effects of the epigenetic repurposed drugs hydralazine and valproate in lymphoma cells. Am J Cancer Res 2024; 14:3068-3082. [PMID: 39005694 PMCID: PMC11236763 DOI: 10.62347/idkg8587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/27/2024] [Indexed: 07/16/2024] Open
Abstract
Lymphoma is a disease that affects countless lives each year. In order to combat this disease, researchers have been exploring the potential of DNMTi and HDACi drugs. These drugs target the cellular processes that contribute to lymphomagenesis and treatment resistance. Our research evaluated the effectiveness of a combination of two such drugs, hydralazine (DNMTi) and valproate (HDACi), in B-cell and T-cell lymphoma cell lines. Here we show that the combination of hydralazine and valproate decreased the viability of cells over time, leading to the arrest of cell-cycle and apoptosis in both B and T-cells. This combination of drugs proved to be synergistic, with each drug showing significant growth inhibition individually. Microarray analyses of HuT 78 and Raji cells showed that the combination of hydralazine and valproate resulted in the up-regulation of 562 and 850 genes, respectively, while down-regulating 152 and 650 genes. Several proapoptotic and cell cycle-related genes were found to be up-regulated. Notably, three and five of the ten most up-regulated genes in HuT 78 and Raji cells, respectively, were related to immune function. In summary, our study suggests that the combination of hydralazine and valproate is an effective treatment option for both B- and T-lymphomas. These findings are highly encouraging, and we urge further clinical evaluation to validate our research and potentially improve lymphoma treatment.
Collapse
Affiliation(s)
- Harold Salamanca-Ortiz
- Subdirection of Basic Research, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
| | - Guadalupe Domínguez-Gomez
- Subdirección de Investigación Clínica, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
| | - Alma Chávez-Blanco
- Subdirection of Basic Research, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
| | - Daniel Ortega-Bernal
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autónoma MetropolitanaCoyoacan, Mexico City 05348, Mexico
- Department of Sciences, Universidad Autónoma MetropolitanaCoyoacan, Mexico City 04960, Mexico
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana XochimilcoCoyoacan, Mexico City 04960, Mexico
| | - José Díaz-Chávez
- Subdirection of Basic Research, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
| | - Aurora González-Fierro
- Subdirection of Basic Research, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
| | - Myrna Candelaria-Hernández
- Subdirección de Investigación Clínica, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
| | - Alfonso Dueñas-González
- Subdirection of Basic Research, Instituto Nacional de Cancerología (INCan)Tlalpan, Mexico City 14080, Mexico
- Department of Genomic Medicine and Environmental Toxicology, Institute of Biomedical Research, Universidad Nacional Autónoma de Mexico (UNAM), Av. Universidad 3004, Copilco UniversidadCoyoacan, Mexico City 04510, Mexico
| |
Collapse
|
4
|
Psilopatis I, Garmpis N, Garmpi A, Vrettou K, Sarantis P, Koustas E, Antoniou EA, Dimitroulis D, Kouraklis G, Karamouzis MV, Marinos G, Kontzoglou K, Nonni A, Nikolettos K, Fleckenstein FN, Zoumpouli C, Damaskos C. The Emerging Role of Histone Deacetylase Inhibitors in Cervical Cancer Therapy. Cancers (Basel) 2023; 15:cancers15082222. [PMID: 37190151 DOI: 10.3390/cancers15082222] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 05/17/2023] Open
Abstract
Cervical carcinoma is one of the most common cancers among women globally. Histone deacetylase inhibitors (HDACIs) constitute anticancer drugs that, by increasing the histone acetylation level in various cell types, induce differentiation, cell cycle arrest, and apoptosis. The aim of the current review is to study the role of HDACIs in the treatment of cervical cancer. A literature review was conducted using the MEDLINE and LIVIVO databases with a view to identifying relevant studies. By employing the search terms "histone deacetylase" and "cervical cancer", we managed to identify 95 studies published between 2001 and 2023. The present work embodies the most up-to-date, comprehensive review of the literature centering on the particular role of HDACIs as treatment agents for cervical cancer. Both well-established and novel HDACIs seem to represent modern, efficacious anticancer drugs, which, alone or in combination with other treatments, may successfully inhibit cervical cancer cell growth, induce cell cycle arrest, and provoke apoptosis. In summary, histone deacetylases seem to represent promising future treatment targets in cervical cancer.
Collapse
Affiliation(s)
- Iason Psilopatis
- Department of Gynecology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nikolaos Garmpis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Nikolaos Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anna Garmpi
- First Department of Propedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Kleio Vrettou
- Department of Cytopathology, Sismanogleio General Hospital, 15126 Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efstathios A Antoniou
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Nikolaos Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Dimitroulis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Nikolaos Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Gregory Kouraklis
- Department of Surgery, Evgenideio Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Michail V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgios Marinos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Kontzoglou
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Nikolaos Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Afroditi Nonni
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Nikolettos
- Obstetric and Gynecologic Clinic, Medical School, Democritus University of Thrace, 68110 Alexandroupolis, Greece
| | - Florian N Fleckenstein
- Department of Diagnostic and Interventional Radiology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, 13353 Berlin, Germany
| | - Christina Zoumpouli
- Department of Pathology, Sismanogleio General Hospital, 15126 Athens, Greece
| | - Christos Damaskos
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Renal Transplantation Unit, Laiko General Hospital, 11527 Athens, Greece
| |
Collapse
|
5
|
Ullrich M, Richter S, Liers J, Drukewitz S, Friedemann M, Kotzerke J, Ziegler CG, Nölting S, Kopka K, Pietzsch J. Epigenetic drugs in somatostatin type 2 receptor radionuclide theranostics and radiation transcriptomics in mouse pheochromocytoma models. Theranostics 2023; 13:278-294. [PMID: 36593963 PMCID: PMC9800739 DOI: 10.7150/thno.77918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Pheochromocytomas and paragangliomas (PCCs/PGLs) are catecholamine-producing tumors. In inoperable and metastatic cases, somatostatin type 2 receptor (SSTR2) expression allows for peptide receptor radionuclide therapy with [177Lu]Lu-DOTA-TATE. Insufficient receptor levels, however, limit treatment efficacy. This study evaluates whether the epigenetic drugs valproic acid (VPA) and 5-Aza-2'-deoxycytidine (DAC) modulate SSTR2 levels and sensitivity to [177Lu]Lu-DOTA-TATE in two mouse PCC models (MPC and MTT). Methods: Drug-effects on Sstr2/SSTR2 were investigated in terms of promoter methylation, mRNA and protein levels, and radiotracer binding. Radiotracer uptake was measured in subcutaneous allografts in mice using PET and SPECT imaging. Tumor growth and gene expression (RNAseq) were characterized after drug treatments. Results: DAC alone and in combination with VPA increased SSTR2 levels along with radiotracer uptake in vitro in MPC (high-SSTR2) and MTT cells (low-SSTR2). MTT but not MPC allografts responded to DAC and VPA combination with significantly elevated radiotracer uptake, although activity concentrations remained far below those in MPC tumors. In both models, combination of DAC, VPA and [177Lu]Lu-DOTA-TATE was associated with additive effects on tumor growth delay and specific transcriptional responses in gene sets involved in cancer and treatment resistance. Effects of epigenetic drugs were unrelated to CpG island methylation of the Sstr2 promoter. Conclusion: This study demonstrates that SSTR2 induction in mouse pheochromocytoma models has some therapeutic benefit that occurs via yet unknown mechanisms. Transcriptional changes in tumor allografts associated with epigenetic treatment and [177Lu]Lu-DOTA-TATE provide first insights into genetic responses of PCCs/PGLs, potentially useful for developing additional strategies to prevent tumor recurrence.
Collapse
Affiliation(s)
- Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany.,✉ Corresponding author: Dr. Martin Ullrich, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany. Phone: +49-351-2604046, Fax: +49-351-26012622, E-mail:
| | - Susan Richter
- University Hospital Carl Gustav Carus at the Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Josephine Liers
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany.,University Hospital Carl Gustav Carus at the Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Stephan Drukewitz
- National Center for Tumor Diseases/University Cancer Center Dresden, Core Unit for Molecular Tumor Diagnostics, Dresden, Germany.,University of Leipzig Medical Center, Institute of Human Genetics, Leipzig, Germany
| | - Markus Friedemann
- University Hospital Carl Gustav Carus at the Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Jörg Kotzerke
- University Hospital Carl Gustav Carus at the Technische Universität Dresden, Klinik und Poliklinik für Nuklearmedizin, Dresden, Germany
| | - Christian G. Ziegler
- University Hospital Carl Gustav Carus at the Technische Universität Dresden, Department of Medicine III, Dresden, Germany
| | - Svenja Nölting
- University Hospital Zurich (USZ) and University of Zurich (UZH), Department of Endocrinology, Diabetology and Clinical Nutrition, Zurich, Switzerland.,University Hospital, LMU Munich, Department of Medicine IV, Munich, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, University Cancer Center (UCC), Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
6
|
Fraga da Silva E, Roberto dos Santos P, Helen Antunes K, Marinho Franceschina C, Nascimento de Freitas D, Konrad P, Fernandes Zanin R, Machado P, Moura S, de Souza APD. Anti-tumor effects of valproate zinc complexes on a lung cancer cell line. Polyhedron 2021. [DOI: 10.1016/j.poly.2021.115415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
7
|
Gama RR, Arantes LMRB, Sorroche BP, De Marchi P, Melendez ME, Carvalho RS, de Lima MA, Vettore AL, Carvalho AL. Evaluation of acetylation and methylation in oral rinse of patients with head and neck cancer history exposed to valproic acid. Sci Rep 2021; 11:16415. [PMID: 34385507 PMCID: PMC8361187 DOI: 10.1038/s41598-021-95845-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/26/2021] [Indexed: 01/18/2023] Open
Abstract
Evaluate the biological action of valproic acid in the acetylation of histones and in the methylation of tumor suppressor genes via oral rinse in patients with a previous history of head and neck squamous cell carcinoma (HNSCC). Forty-two active or former smokers were included in this randomized, double-blind, placebo-controlled trial. Oral rinse samples were collected prior to treatment with valproic acid or placebo and after 90 days of treatment. The methylation status of five tumor suppressor genes and histone acetylation were evaluated by pyrosequencing and ELISA techniques, respectively. Differences between the 90-day and baseline oral rinse acetylation and methylation results were analyzed by comparing groups. Thirty-four patients were considered for analysis. The mean percentage adherence in the valproic and placebo groups was 93.4 and 93.0, respectively (p = 0.718). There was no statistically significant difference between groups when comparing the medians of the histone acetylation ratio and the methylation ratio for most of the studied genes. A significant reduction in the DCC methylation pattern was observed in the valproic group (p = 0.023). The use of valproic acid was safe and accompanied by good therapeutic adherence. DCC methylation was lower in the valproic acid group than in the placebo group.
Collapse
Affiliation(s)
- Ricardo Ribeiro Gama
- Head and Neck Surgery Department, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Bairro Dr. Paulo Prata, Barretos, SP, 14784-400, Brazil.
- Postgraduation Program, São Paulo Federal University, São Paulo, SP, Brazil.
| | | | | | - Pedro De Marchi
- Clinical Oncology Department, Barretos Cancer Hospital, Barretos, SP, Brazil
- Oncoclínicas, Rio de Janeiro, RJ, Brazil
| | | | - Raiany Santos Carvalho
- Researcher Support Center, Teaching and Research Institute, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Marcos Alves de Lima
- Biostatistics Center, Teaching and Research Institute, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - André Luiz Vettore
- Cancer Molecular Biology Laboratory, São Paulo Federal University, Diadema, SP, Brazil
| | - André Lopes Carvalho
- Head and Neck Surgery Department, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Bairro Dr. Paulo Prata, Barretos, SP, 14784-400, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
- Senior Researcher, International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
8
|
Lourenço de Freitas N, Deberaldini MG, Gomes D, Pavan AR, Sousa Â, Dos Santos JL, Soares CP. Histone Deacetylase Inhibitors as Therapeutic Interventions on Cervical Cancer Induced by Human Papillomavirus. Front Cell Dev Biol 2021; 8:592868. [PMID: 33634093 PMCID: PMC7901962 DOI: 10.3389/fcell.2020.592868] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
The role of epigenetic modifications on the carcinogenesis process has received a lot of attention in the last years. Among those, histone acetylation is a process regulated by histone deacetylases (HDAC) and histone acetyltransferases (HAT), and it plays an important role in epigenetic regulation, allowing the control of the gene expression. HDAC inhibitors (HDACi) induce cancer cell cycle arrest, differentiation, and cell death and reduce angiogenesis and other cellular events. Human papillomaviruses (HPVs) are small, non-enveloped double-stranded DNA viruses. They are major human carcinogens, being intricately linked to the development of cancer in 4.5% of the patients diagnosed with cancer worldwide. Long-term infection of high-risk (HR) HPV types, mainly HPV16 and HPV18, is one of the major risk factors responsible for promoting cervical cancer development. In vitro and in vivo assays have demonstrated that HDACi could be a promising therapy to HPV-related cervical cancer. Regardless of some controversial studies, the therapy with HDACi could target several cellular targets which HR-HPV oncoproteins could be able to deregulate. This review article describes the role of HDACi as a possible intervention in cervical cancer treatment induced by HPV, highlighting the main advances reached in the last years and providing insights for further investigations regarding those agents against cervical cancer.
Collapse
Affiliation(s)
- Natália Lourenço de Freitas
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Maria Gabriela Deberaldini
- Drugs and Medicines Department, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, Brazil
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Diana Gomes
- CICS-UBI – Health Science Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Aline Renata Pavan
- Drugs and Medicines Department, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, Brazil
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Ângela Sousa
- CICS-UBI – Health Science Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Jean Leandro Dos Santos
- Drugs and Medicines Department, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, Brazil
| | - Christiane P. Soares
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
9
|
Mak MP, Pasini FS, Diao L, Garcia FOT, Takahashi TK, Nakazato D, Martins RE, Almeida CM, Kulcsar MAV, Lamounier VA, Nunes EM, de Souza IC, Garcia MRT, Amadio AV, Siqueira SAC, Snitcovsky IML, Sichero L, Wang J, de Castro G. Valproic acid combined with cisplatin-based chemoradiation in locally advanced head and neck squamous cell carcinoma patients and associated biomarkers. Ecancermedicalscience 2020; 14:1155. [PMID: 33574900 PMCID: PMC7864693 DOI: 10.3332/ecancer.2020.1155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
Background Cisplatin-based chemoradiation (CCRT) offers locally advanced head and neck squamous cell carcinoma (LAHNSCC) patients high local control rate, however, relapses are frequent. Our goal was to evaluate if association of valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, with CCRT improved response rate (RR) and associated biomarkers. Methods This phase II trial included patients with unresectable locally advanced (LA) oropharynx (OP) squamous cell carcinoma. CCRT began after 2 weeks of VPA (P1). Primary goal was RR at 8 weeks after chemoradiation (CRT)+VPA (P2). Biomarkers included microRNA (miR) polymerase chain reaction (PCR)-array profiling in plasma compared to healthy controls by two-sample t-test. Distribution of p-values was analysed by beta-uniform mixture. Findings were validated by real-time PCR quantitative polymerase chain reaction (qPCR) for selected miRs in plasma and saliva. p16, HDAC2 and RAD23 Homolog B, Nucleotide Excision Repair Protein (HR23B) tumour immunohistochemistry were evaluated. Results Given significant toxicities, accrual was interrupted after inclusion of ten LA p16 negative OP patients. All were male, smokers/ex-smokers, aged 41–65 and with previous moderate/high alcohol intake. Nine evaluable patients yielded a RR of 88%. At false discovery rate of 5%, 169 miRs were differentially expressed between patients and controls, including lower expression of tumour suppressors (TSs) such as miR-31, -222, -let-7a/b/e and -145. miR-let-7a/e expression was validated by qPCR using saliva. A HDAC2 H-score above 170 was 90% accurate in predicting 6-month disease-free survival. Conclusions VPA and CRT offered high RR; however, with prohibitive toxicities, which led to early trial termination. Patients and controls had a distinct pattern of miR expression, mainly with low levels of TS miRs targeting Tumor protein P53 (TP53). miR-let-7a/e levels were lower in patients compared to controls, which reinforces the aggressive nature of such tumours (NCT01695122).
Collapse
Affiliation(s)
- Milena Perez Mak
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Fatima Solange Pasini
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas, MD Anderson Cancer Center, 1400 Pressler St. Floor 4, FCT4.6000, Houston, Texas, USA
| | - Fabyane O Teixeira Garcia
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Tiago Kenji Takahashi
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Denyei Nakazato
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Renata Eiras Martins
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Cristiane Maria Almeida
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Marco Aurelio Vamondes Kulcsar
- Head and Neck Surgery Department, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Valdelania Aparecida Lamounier
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Emily Montosa Nunes
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Isabela Cristina de Souza
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Marcio Ricardo Taveira Garcia
- Department of Radiology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Alex Vieira Amadio
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Sheila Aparecida C Siqueira
- Department of Pathology, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Eneas de Carvalho Aguiar, 255, CEP 05403-000, Sao Paulo, SP, Brazil
| | - Igor Moysés Longo Snitcovsky
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Laura Sichero
- Center for Translational Investigation in Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Jing Wang
- Head and Neck Surgery Department, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| | - Gilberto de Castro
- Department of Medical Oncology, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Av Dr Arnaldo, 251 12th floor, CEP 01246-000, Sao Paulo, SP, Brazil
| |
Collapse
|
10
|
Avallone A, Piccirillo MC, Di Gennaro E, Romano C, Calabrese F, Roca MS, Tatangelo F, Granata V, Cassata A, Cavalcanti E, Maurea N, Maiolino P, Silvestro L, De Stefano A, Giuliani F, Rosati G, Tamburini E, Aprea P, Vicario V, Nappi A, Vitagliano C, Casaretti R, Leone A, Petrillo A, Botti G, Delrio P, Izzo F, Perrone F, Budillon A. Randomized phase II study of valproic acid in combination with bevacizumab and oxaliplatin/fluoropyrimidine regimens in patients with RAS-mutated metastatic colorectal cancer: the REVOLUTION study protocol. Ther Adv Med Oncol 2020; 12:1758835920929589. [PMID: 32849914 PMCID: PMC7425244 DOI: 10.1177/1758835920929589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/04/2020] [Indexed: 01/30/2023] Open
Abstract
Background Despite effective treatments, metastatic colorectal cancer (mCRC) prognosis is still poor, mostly in RAS-mutated tumors, thus suggesting the need for novel combinatorial therapies. Epigenetic alterations play an important role in initiation and progression of cancers, including CRC. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with chemotherapy in the treatment of solid tumors. Owing to its HDACi activity and its safe use for epileptic disorders, valproic acid (VPA) is a good candidate for anticancer therapy that we have largely explored preclinically translating our findings in currently ongoing clinical studies. We have shown in CRC models that HDACi, including VPA, induces synergistic antitumor effects in combination with fluoropyrimidines. Furthermore, unpublished results from our group demonstrated that VPA induces differentiation and sensitization of CRC stem cells to oxaliplatin. Moreover, preclinical and clinical data suggest that HDACi may prevent/reverse anti-angiogenic resistance. Methods/Design A randomized, open-label, two-arm, multicenter phase-II study will be performed to explore whether the addition of VPA to first line bevacizumab/oxaliplatin/fluoropyrimidine regimens (mFOLFOX-6/mOXXEL) might improve progression-free survival (PFS) in RAS-mutated mCRC patients. A sample size of 200 patients was calculated under the hypothesis that the addition of VPA to chemotherapy/bevacizumab can improve PFS from 9 to 12 months, with one-sided alpha of 0.20 and a power of 0.80. Secondary endpoints are overall survival, objective response rate, metastases resection rate, toxicity, and quality of life. Moreover, the study will explore several prognostic and predictive biomarkers on blood samples, primary tumors, and on resected metastases. Discussion The "Revolution" study aims to improve the treatment efficacy of RAS-mutated mCRC through an attractive strategy evaluating the combination of VPA with standard cancer treatment. Correlative studies could identify novel biomarkers and could add new insight in the mechanism of interaction between VPA, fluoropyrimidine, oxaliplatin, and bevacizumab. Trial Registration EudraCT: 2018-001414-15; ClinicalTrials.gov identifier: NCT04310176.
Collapse
Affiliation(s)
- Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy
| | | | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Carmela Romano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Filomena Calabrese
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Antonio Cassata
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Nicola Maurea
- Cardiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Lucrezia Silvestro
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alfonso De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | | | - Gerardo Rosati
- Medical Oncology Unit, S. Carlo Hospital, Potenza, Italy
| | - Emiliano Tamburini
- Dipartimento di Oncologia e Cure Palliative, Azienda Ospedaliera Cardinale G. Panico, Tricase-Lecce, Italy
| | - Pasquale Aprea
- Vascular Access Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Valeria Vicario
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Anna Nappi
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Rossana Casaretti
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Paolo Delrio
- Colorectal Oncological Surgery, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy
| |
Collapse
|
11
|
Fan HC, Wang SY, Peng YJ, Lee HS. Valproic Acid Impacts the Growth of Growth Plate Chondrocytes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E3675. [PMID: 32456093 PMCID: PMC7277424 DOI: 10.3390/ijerph17103675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/13/2020] [Accepted: 05/21/2020] [Indexed: 12/24/2022]
Abstract
A range of bone abnormalities including short stature have been reported to be associated with the use of antiepileptic drugs (AEDs) in children. Exactly how AEDs impact skeletal growth, however, is not clear. In the present study, rat growth plate chondrocytes were cultured to study the effects of AEDs, including valproic acid (VPA), oxcarbazepine (OXA), levetiracetam (LEV), lamotrigine (LTG), and topiramate (TPM) on the skeletal growth. VPA markedly reduced the number of chondrocytes by apoptosiswhile other AEDs had no effect. The apoptosis associated noncleaved and cleaved caspase 3, and caspases were increased by exposure to VPA, which up-regulated cyclooxygenase 2 (COX-2) mRNA and protein levels likely through histone acetylation. The COX-2 inhibitor NS-398 attenuated the effects of VPA up-regulating COX-2 expression and decreased VPA-induced caspase 3 expression. The use of VPA in children should be closely monitored or replaced, where appropriate, by AEDs which do not apparently affect the growth plate chondrocytes.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Taichung 435, Taiwan;
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung 435, Taiwan
- Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35053, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Shih-Yu Wang
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (S.-Y.W.); (Y.-J.P.)
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (S.-Y.W.); (Y.-J.P.)
| | - Herng-Sheng Lee
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (S.-Y.W.); (Y.-J.P.)
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| |
Collapse
|
12
|
Valproic acid as an adjunctive therapeutic agent for the treatment of breast cancer. Eur J Pharmacol 2018; 835:61-74. [DOI: 10.1016/j.ejphar.2018.07.057] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023]
|
13
|
Oppermann H, Alvanos A, Seidel C, Meixensberger J, Gaunitz F. Carnosine influences transcription via epigenetic regulation as demonstrated by enhanced histone acetylation of the pyruvate dehydrogenase kinase 4 promoter in glioblastoma cells. Amino Acids 2018; 51:61-71. [DOI: 10.1007/s00726-018-2619-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/12/2018] [Indexed: 01/11/2023]
|
14
|
Monga V, Swami U, Tanas M, Bossler A, Mott SL, Smith BJ, Milhem M. A Phase I/II Study Targeting Angiogenesis Using Bevacizumab Combined with Chemotherapy and a Histone Deacetylase Inhibitor (Valproic Acid) in Advanced Sarcomas. Cancers (Basel) 2018; 10:E53. [PMID: 29462961 PMCID: PMC5836085 DOI: 10.3390/cancers10020053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/18/2022] Open
Abstract
Epigenetic events and genetic alterations under the control of the tumor microenvironment potentially mediate tumor induced angiogenesis involved in soft tissue sarcoma (STS) metastasis. Addition of antiangiogenic agent, such as bevacizumab, to standard chemotherapy in treatment of sarcoma has been studied in clinical trials, but most of the findings have not supported its use. We hypothesized the existence of an epigenetically mediated "angiogenic switch", and the tumor microenvironment, prevents bevacizumab from truly blocking angiogenesis. The addition of valproic acid (VPA), a weak histone deacetylase inhibitor, and bevacizumab, a monoclonal antibody against vascular endothelial growth factor, together with the cytotoxic effects of gemcitabine and docetaxel, may enhance responses and alter chemoresistance. This was designed as a phase I/II trial with primary endpoints including safety of the treatment combination and tumor response. Unresectable or metastatic sarcoma patients >18 years of age, irrespective of number of prior treatments, received VPA 40 mg/kg orally for 5 days prior to day 1, bevacizumab at 15 mg/kg IV on day 1, gemcitabine 900 mg/m² (day 1, day 8), and docetaxel 75 mg/m² (day 8). Cycles were of 28 day duration. Bevacizumab and VPA were continued as maintenance after 6 cycles, until disease progression. A standard 3 + 3 phase I dose de-escalation design was utilized to evaluate safety. Gain of function p53 gene mutation testing was performed on available archival tissue specimens. A total of 46 patients (30 female, 16 male) with median age of 60 (range 24-81) years were enrolled; 34 (73.9%) patients received prior chemotherapy, 14 (30%) of which received prior gemcitabine and docetaxel. Patients received a median of 5.5 cycles (range 0-24 of treatment (min 0, one patient died prior to completing the first cycle; max: 24, one patient received 6 cycles and 18 maintenance cycles before progressing). Seventeen patients underwent dose reduction, of which VPA was reduced in 6 patients. Forty-one patients were evaluable for response. There was a confirmed complete response in 1 (epithelioid sarcoma), and a partial response (PR) in 6 (1 carcinosarcoma, 2 extrauterine leiomyosarcoma (LMS), 2 undifferentiated pleomorphic sarcoma, and 1 uterine LMS) patients. Stable disease (SD) was seen in 21 patients for at least 2 months. One subject with prior gemcitabine and docetaxel had PR, and 7 had SD. Median progression-free survival (PFS) was 5.7 months (95% CI: 2.1-8.0), and overall survival (OS) was 12.9 months (95% CI: 8.3-14.5). Three patients died due to tumor progression while on the study. The combination of VPA, bevacizumab, gemcitabine, and docetaxel appears to be moderately safe and well tolerated. Given that there are very limited options for patients with relapsed refractory STS, this drug combination may be an important therapy to consider. This combination treatment deserves further investigation in epithelioid and carcinosarcoma subtypes.
Collapse
Affiliation(s)
- Varun Monga
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Umang Swami
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Munir Tanas
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Aaron Bossler
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Sarah L Mott
- Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA.
| | - Brian J Smith
- Department of Biostatistics, University of Iowa College of Public Health & Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA.
| | - Mohammed Milhem
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
15
|
Goda K, Saito K, Muta K, Kobayashi A, Saito Y, Sugai S. Ether-phosphatidylcholine characterized by consolidated plasma and liver lipidomics is a predictive biomarker for valproic acid-induced hepatic steatosis. J Toxicol Sci 2018; 43:395-405. [DOI: 10.2131/jts.43.395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Keisuke Goda
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO Inc
| | - Kosuke Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences
| | - Kyotaka Muta
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO Inc
| | - Akio Kobayashi
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO Inc
| | - Yoshiro Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences
| | - Shoichiro Sugai
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO Inc
| |
Collapse
|
16
|
Yao ZG, Li WH, Hua F, Cheng HX, Zhao MQ, Sun XC, Qin YJ, Li JM. LBH589 Inhibits Glioblastoma Growth and Angiogenesis Through Suppression of HIF-1α Expression. J Neuropathol Exp Neurol 2017; 76:1000-1007. [PMID: 29136455 DOI: 10.1093/jnen/nlx088] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma (GBM) is an angiogenic malignancy with a highly unfavorable prognosis. Angiogenesis in GBM represents an adaptation to a hypoxic microenvironment and is correlated with tumor growth, invasion, clinical recurrence, and lethality. LBH589 (also called panobinostat) is a histone deacetylase (HDAC) inhibitor with potent antitumor activity. In the current study, we investigated the mechanism and effects of LBH589 on GBM growth and hypoxia-induced angiogenesis in vitro and in vivo. To determine the antitumor and angiogenesis activity and mechanism of LBH589, we used cell proliferations in vitro and GBM xenografts in vivo. To clarify mechanisms of LBH589 on angiogenesis, HDAC assay, RT-PCR, Western blot, and co-immunoprecipitation assays were performed. We found LBH589 displayed significant antitumor effects on GBM as demonstrated by inhibited cell proliferation, slower tumor growth, and decreased microvessel density of subcutaneous xenografts. These actions of LBH589 resulted from the disruption of heat shock protein 90/HDAC6 complex, increased HIF-1α instability and degradation, and decreased VEGF expression. Our results indicate the potential antiangiogenic activity of LBH589 in human GBM and provide some preclinical data to warrant further exploration of HDAC inhibitors for the treatment of advanced glioma. Moreover, our study supports the role of HDAC inhibitors as a therapeutic strategy to target tumor angiogenesis.
Collapse
Affiliation(s)
- Zhi-Gang Yao
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Huan Li
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Hua
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Xia Cheng
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao-Qing Zhao
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi-Chao Sun
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye-Jun Qin
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Mei Li
- Department of Pathology and Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; and Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Espinoza-Zamora JR, Labardini-Méndez J, Sosa-Espinoza A, López-González C, Vieyra-García M, Candelaria M, Lozano-Zavaleta V, Toledano-Cuevas DV, Zapata-Canto N, Cervera E, Dueñas-González A. Efficacy of hydralazine and valproate in cutaneous T-cell lymphoma, a phase II study. Expert Opin Investig Drugs 2017; 26:481-487. [PMID: 28277033 DOI: 10.1080/13543784.2017.1291630] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To evaluate the activity and safety of hydralazine and valproate (Transkrip) in cutaneous T-cell lymphoma (CTCL). METHODS Previously untreated and progressive/refractory CTCL patients received hydralazine at 83 mg or 182 mg/day for slow and rapid acetylators respectively plus magnesium valproate at a total dose of 30 mg/Kg t.i.d daily in continuous 28-day cycles in this phase II study. The primary objective was overall response rate (ORR) measured by the modified severity weighted assessment tool (m-SWAT), secondary end-points were time to response (TTR), time to progression (TTP), duration of response (DOR), progression-free survival (PFS), overall survival (OS) and safety. RESULTS Fourteen patients were enrolled (7 untreated and 7 pretreated). ORR was 71% with 50% complete and 21% partial. Two had stable disease and two progressed. At a median follow-up of 36 months (5-52), median TTR was 2 months (1-4); median DOR was 28 months (5-45); median PFS 36 and not reached for OS. There were no differences in median TTR, DOR, and PFS between treated and pretreated patients. Pruritus relieve was complete in 13 out of 14 patients. No grade 3 or 4 toxicities were observed. CONCLUSION The combination of hydralazine and valproate is safe, very well tolerated and effective in CTCL.
Collapse
Affiliation(s)
| | - Juan Labardini-Méndez
- a Department of Hematology , Instituto Nacional de Cancerología , Mexico City , Mexico
| | | | - Celia López-González
- a Department of Hematology , Instituto Nacional de Cancerología , Mexico City , Mexico
| | | | - Myrna Candelaria
- b Division of Clinical Research , Instituto Nacional de Cancerología , Mexico City , Mexico
| | | | | | - Nidia Zapata-Canto
- a Department of Hematology , Instituto Nacional de Cancerología , Mexico City , Mexico
| | - Eduardo Cervera
- a Department of Hematology , Instituto Nacional de Cancerología , Mexico City , Mexico
| | - Alfonso Dueñas-González
- c Unidad de Investigación Biomédica en Cáncer , Instituto de Investigaciones Biomédicas UNAM/Instituto Nacional de Cancerología , Mexico City , Mexico
| |
Collapse
|
18
|
Caponigro F, Di Gennaro E, Ionna F, Longo F, Aversa C, Pavone E, Maglione MG, Di Marzo M, Muto P, Cavalcanti E, Petrillo A, Sandomenico F, Maiolino P, D'Aniello R, Botti G, De Cecio R, Losito NS, Scala S, Trotta A, Zotti AI, Bruzzese F, Daponte A, Calogero E, Montano M, Pontone M, De Feo G, Perri F, Budillon A. Phase II clinical study of valproic acid plus cisplatin and cetuximab in recurrent and/or metastatic squamous cell carcinoma of Head and Neck-V-CHANCE trial. BMC Cancer 2016; 16:918. [PMID: 27884140 PMCID: PMC5123351 DOI: 10.1186/s12885-016-2957-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 11/20/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Recurrent/metastatic squamous cell carcinoma of the head and neck (SCCHN) has a poor prognosis and the combination of cisplatin and cetuximab, with or without 5-fluorouracil, is the gold standard treatment in this stage. Thus, the concomitant use of novel compounds represents a critical strategy to improve treatment results. Histone deacetylase inhibitors (HDACi) enhance the activity of several anticancer drugs including cisplatin and anti-Epidermal Growth Factor Receptor (anti-EGFR) compounds. Preclinical studies in models have shown that vorinostat is able to down regulate Epidermal Growth Factor Receptor (EGFR) expression and to revert epithelial to mesenchimal transition (EMT). Due to its histone deacetylase (HDAC) inhibiting activity and its safe use as a chronic therapy for epileptic disorders, valproic acid (VPA) has been considered a good candidate for anticancer therapy. A reasonable option may be to employ the combination of cisplatin, cetuximab and VPA in recurrent/metastatic SCCHN taking advantage of the possible positive interaction between histone deacetylase inhibitors, cisplatin and/or anti-EGFR. METHOD/DESIGN V-CHANCE is a phase 2 clinical trial evaluating, in patients with recurrent/metastatic squamous cell carcinoma of the head and neck never treated with first-line chemotherapy, the concomitant standard administration of cisplatin (on day 1, every 3 weeks) and cetuximab (on day 1, weekly), in combination with oral VPA given daily from day -14 with a titration strategy in each patient (target serum level of 50-100 μg/ml). Primary end point is the objective response rate measured according to Response Evaluation Criteria in Solid Tumors (RECIST). Sample size, calculated according to Simon 2 stage minimax design will include 21 patients in the first stage with upper limit for rejection being 8 responses, and 39 patients in the second stage, with upper limit for rejection being 18 responses. Secondary endpoints are time to progression, duration of response, overall survival, safety. Objectives of the translational study are the evaluation on tumor samples of markers of treatment efficacy/resistance (i.e. γH2AX, p21/WAF, RAD51, XRCC1, EGFR, p-EGFR, Ki-67) and specific markers of VPA HDAC inhibitory activity (histones and proteins acetylation, Histone deacetylase isoforms) as well as valproate test, histones and proteins acetylation of peripheral blood mononuclear cell, tested on blood samples at baseline and at different time points during treatment. DISCUSSION Overall, this study could provide a less toxic and more effective first-line chemotherapy regimen in patients with recurrent/metastatic squamous cell carcinoma of the head and neck by demonstrating the feasibility and efficacy of cisplatin/cetuximab plus valproic acid. Moreover, correlative studies could help to identify responder patients, and will add insights in the mechanism of the synergistic interaction between these agents. EUDRACT NUMBER 2014-001523-69 TRIAL REGISTRATION: ClinicalTrials.gov number, NCT02624128.
Collapse
Affiliation(s)
- Francesco Caponigro
- Head and Neck Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy.
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Franco Ionna
- Head and neck Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Francesco Longo
- Head and neck Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Corrado Aversa
- Head and neck Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Ettore Pavone
- Head and neck Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Maria Grazia Maglione
- Head and neck Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Massimiliano Di Marzo
- Melanoma and soft tissue Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Paolo Muto
- Radiotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Ernesta Cavalcanti
- Clinical Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Fabio Sandomenico
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Roberta D'Aniello
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Nunzia Simona Losito
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Stefania Scala
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Annamaria Trotta
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Andrea Ilaria Zotti
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Antonio Daponte
- Head and Neck Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Ester Calogero
- Head and Neck Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Massimo Montano
- Head and Neck Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Monica Pontone
- Head and Neck Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Gianfranco De Feo
- Scientific Direction, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Francesco Perri
- Head and Neck Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy.,Present Address: Medical Oncology Unit, POC SS Annunziata, Taranto, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale," IRCCS, Naples, Italy.
| |
Collapse
|
19
|
Human precision-cut liver slices as a model to test antifibrotic drugs in the early onset of liver fibrosis. Toxicol In Vitro 2016; 35:77-85. [DOI: 10.1016/j.tiv.2016.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/13/2016] [Accepted: 05/24/2016] [Indexed: 01/11/2023]
|
20
|
Gandhi AK. Novel agents and treatment techniques to enhance radiotherapeutic outcomes in carcinoma of the uterine cervix. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:49. [PMID: 26904571 DOI: 10.3978/j.issn.2305-5839.2015.10.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND Survival of patients with locally advanced carcinoma cervix (LACC) using the current standard of concurrent chemo-radiotherapy (CCRT) has reached a plateau over the last two decades. Loco-regional failure in first two years of treatment completion and distant metastasis in the subsequent years has put the survival curves at a halt. Strategies of induction and adjuvant chemotherapy have yielded little as has any advancement in techniques of delivery of radiation therapy. This article aims at discussing the current existing literature as well as promising novel strategies to enhance radiotherapeutic outcomes in carcinoma of the uterine cervix. METHODS The review of English literature included phase I-III trials evaluating either a novel agent, novel application/modifications of an existing treatment regimen or an innovative treatment technique. The studies have been divided in to subsections with summary of most important findings at the end of each section. RESULTS Despite CCRT being the 'gold standard' treatment, several issues like optimum drug combination, schedule of drug delivery, combination with molecular targeted agents etc. remain undefined. Taxane, topoisomerase and gemcitabine based regimen needs to be further explored and compared with cisplatin based CCRT regimen. Several approaches like local delivery of cytotoxic agents, use of nano-medicine with CCRT are appearing on horizon with promises for the future. Therapies need to be designed based on the human papillomavirus titers of the patients and incorporation of radiosensitizers as an effective way of palliation with short course of radiotherapy may further enhance the radiotherapeutic outcomes. CONCLUSIONS The results of the studies with novel agents and treatment techniques appear promising. Further research in this arena including incorporation of cost-effectiveness analysis and quality of life issues in future trial designs are warranted.
Collapse
Affiliation(s)
- Ajeet Kumar Gandhi
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
21
|
Nervi C, De Marinis E, Codacci-Pisanelli G. Epigenetic treatment of solid tumours: a review of clinical trials. Clin Epigenetics 2015; 7:127. [PMID: 26692909 PMCID: PMC4676165 DOI: 10.1186/s13148-015-0157-2] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 11/10/2015] [Indexed: 12/12/2022] Open
Abstract
Epigenetic treatment has been approved by regulatory agencies for haematological malignancies. The success observed in cutaneous lymphomas represents a proof of principle that similar results may be obtained in solid tumours. Several agents that interfere with DNA methylation-demethylation and histones acetylation/deacetylation have been studied, and some (such as azacytidine, decitabine, valproic acid and vorinostat) are already in clinical use. The aim of this review is to provide a brief overview of the molecular events underlying the antitumour effects of epigenetic treatments and to summarise data available on clinical trials that tested the use of epigenetic agents against solid tumours. We not only list results but also try to indicate how the proper evaluation of this treatment might result in a better selection of effective agents and in a more rapid development. We divided compounds in demethylating agents and HDAC inhibitors. For each class, we report the antitumour activity and the toxic side effects. When available, we describe plasma pharmacokinetics and pharmacodynamic evaluation in tumours and in surrogate tissues (generally white blood cells). Epigenetic treatment is a reality in haematological malignancies and deserves adequate attention in solid tumours. A careful consideration of available clinical data however is required for faster drug development and possibly to re-evaluate some molecules that were perhaps discarded too early.
Collapse
Affiliation(s)
- Clara Nervi
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "la Sapienza", Corso della Repubblica, 97, 04100 Latina, Italy
| | - Elisabetta De Marinis
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "la Sapienza", Corso della Repubblica, 97, 04100 Latina, Italy
| | - Giovanni Codacci-Pisanelli
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "la Sapienza", Corso della Repubblica, 97, 04100 Latina, Italy
| |
Collapse
|
22
|
Berghmans T, Lafitte JJ, Scherpereel A, Ameye L, Paesmans M, Meert AP, Colinet B, Tulippe C, Willems L, Leclercq N, Sculier JP. VAC chemotherapy with valproic acid for refractory/relapsing small cell lung cancer: a phase II study. ERJ Open Res 2015; 1:00029-2015. [PMID: 27730152 PMCID: PMC5005117 DOI: 10.1183/23120541.00029-2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/09/2015] [Indexed: 01/13/2023] Open
Abstract
Salvage chemotherapy (CT) for relapsing or refractory small cell lung cancer (SCLC) remains disappointing. In vitro experiments showed that valproic acid increases apoptosis of SCLC cell lines exposed to doxorubicin, vindesine and bis(2-chloroethyl)amine. The primary objective of this phase II study was to determine whether epigenetic modulation with valproic acid in addition to a doxorubicin, vindesine and cyclophosphamide (VAC) regimen improves 6-month progression-free survival (PFS). Patients with pathologically proven SCLC refractory to prior platinum derivatives and etoposide were eligible. After central registration, patients received VAC plus daily oral valproic acid. 64 patients were registered, of whom six were ineligible. Seven patients did not receive any CT, leaving 51 patients assessable for the primary end-point. The objective response rate was 19.6%. Median PFS was 2.8 months (95% CI 2.5–3.6 months) and 6-month PFS was 6%. Median survival time was 5.9 months (95% CI 4.7–7.5 months). Toxicity was mainly haematological, with 88% and 26% grade 3–4 neutropenia and thrombopenia, respectively. Despite an interesting response rate, the addition of valproic acid to VAC did not translate into adequate PFS in relapsing SCLC or SCLC refractory to platinum–etoposide. Epigenetic modulation with valproic acid does not improve CT efficacy in refractory SCLC after platinum–etoposidehttp://ow.ly/R0rBt
Collapse
Affiliation(s)
- Thierry Berghmans
- Department of Intensive Care Unit and Thoracic Oncology, Institut Jules Bordet, Centre des Tumeurs de l'Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jean-Jacques Lafitte
- Department of Pulmonary and Thoracic Oncology, CHRU de Lille and University of Lille II, France
| | - Arnaud Scherpereel
- Department of Pulmonary and Thoracic Oncology, CHRU de Lille and University of Lille II, France
| | - Lieveke Ameye
- Data Centre, Institut Jules Bordet, Centre des Tumeurs de l'ULB, Brussels, Belgium
| | - Marianne Paesmans
- Data Centre, Institut Jules Bordet, Centre des Tumeurs de l'ULB, Brussels, Belgium
| | - Anne-Pascale Meert
- Department of Intensive Care Unit and Thoracic Oncology, Institut Jules Bordet, Centre des Tumeurs de l'Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Benoit Colinet
- Department of Pneumology, Grand Hôpital de Charleroi, Gilly, Belgium
| | | | - Luc Willems
- Gembloux Agro-Bio Tech and Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège, Gembloux, Belgium
| | - Nathalie Leclercq
- Department of Intensive Care Unit and Thoracic Oncology, Institut Jules Bordet, Centre des Tumeurs de l'Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jean-Paul Sculier
- Department of Intensive Care Unit and Thoracic Oncology, Institut Jules Bordet, Centre des Tumeurs de l'Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | |
Collapse
|
23
|
Renusch SR, Harshman S, Pi H, Workman E, Wehr A, Li X, Prior TW, Elsheikh BH, Swoboda KJ, Simard LR, Kissel JT, Battle D, Parthun MR, Freitas MA, Kolb SJ. Spinal Muscular Atrophy Biomarker Measurements from Blood Samples in a Clinical Trial of Valproic Acid in Ambulatory Adults. J Neuromuscul Dis 2015; 2:119-130. [PMID: 27858735 PMCID: PMC5271431 DOI: 10.3233/jnd-150081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Clinical trials of therapies for spinal muscular atrophy (SMA) that are designed to increase the expression the SMN protein ideally include careful assessment of relevant SMN biomarkers. Objective: In the SMA VALIANT trial, a recent double-blind placebo-controlled crossover study of valproic acid (VPA) in ambulatory adult subjects with SMA, we investigated relevant pharmacodynamic biomarkers in blood samples from SMA subjects by direct longitudinal measurement of histone acetylation and SMN mRNA and protein levels in the presence and absence of VPA treatment. Methods: Thirty-three subjects were randomized to either VPA or placebo for the first 6 months followed by crossover to the opposite arm for an additional 6 months. Outcome measures were compared between the two treatments (VPA and placebo) using a standard crossover analysis. Results: A significant increase in histone H4 acetylation was observed with VPA treatment (p = 0.005). There was insufficient evidence to suggest a treatment effect with either full length or truncated SMN mRNA transcript levels or SMN protein levels. Conclusions: These measures were consistent with the observed lack of change in the primary clinical outcome measure in the VALIANT trial. These results also highlight the added benefit of molecular and pharmacodynamic biomarker measurements in the interpretation of clinical trial outcomes.
Collapse
Affiliation(s)
- Samantha R Renusch
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sean Harshman
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hongyang Pi
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eileen Workman
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Allison Wehr
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaobai Li
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Thomas W Prior
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bakri H Elsheikh
- Department of Neurology, Dhahran Medical Center, Dhahran, Saudi Arabia
| | - Kathryn J Swoboda
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Louise R Simard
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John T Kissel
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel Battle
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mark R Parthun
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Michael A Freitas
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Stephen J Kolb
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
24
|
Dali-Youcef N, Froelich S, Moussallieh FM, Chibbaro S, Noël G, Namer IJ, Heikkinen S, Auwerx J. Gene expression mapping of histone deacetylases and co-factors, and correlation with survival time and 1H-HRMAS metabolomic profile in human gliomas. Sci Rep 2015; 5:9087. [PMID: 25791281 PMCID: PMC4367156 DOI: 10.1038/srep09087] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 02/19/2015] [Indexed: 01/12/2023] Open
Abstract
Primary brain tumors are presently classified based on imaging and histopathological
techniques, which remains unsatisfaying. We profiled here by quantitative real time
PCR (qRT-PCR) the transcripts of eighteen histone deacetylases (HDACs) and a subset
of transcriptional co-factors in non-tumoral brain samples from 15 patients operated
for epilepsia and in brain tumor samples from 50 patients diagnosed with grade II
oligodendrogliomas (ODII, n = 9), grade III oligodendrogliomas (ODIII, n = 22) and
glioblastomas (GL, n = 19). Co-factor transcripts were significantly different in
tumors as compared to non-tumoral samples and distinguished different molecular
subgroups of brain tumors, regardless of tumor grade. Among all patients studied,
the expression of HDAC1 and HDAC3 was inversely correlated with
survival, whereas the expression of HDAC4, HDAC5, HDAC6,
HDAC11 and SIRT1 was significantly and positively correlated with
survival time of patients with gliomas. 1H-HRMAS technology revealed
metabolomically distinct groups according to the expression of HDAC1, HDAC4 and
SIRT1, suggesting that these genes may play an important role in regulating brain
tumorigenesis and cancer progression. Our study hence identified different molecular
fingerprints for subgroups of histopathologically similar brain tumors that may
enable the prediction of outcome based on the expression level of co-factor genes
and could allow customization of treatment.
Collapse
Affiliation(s)
- Nassim Dali-Youcef
- 1] Laboratoire de Biochimie et Biologie Moléculaire, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, 1 place de l'hôpital, 67091 Strasbourg Cedex, France [2] Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)/CNRS UMR 7104/INSERM U 964/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Sébastien Froelich
- Department of Neurosurgery, Hôpitaux Universitaires de Strasbourg, avenue Molière, 67085 Strasbourg Cedex, France
| | - François-Marie Moussallieh
- Department of Biophysics and Nuclear Medicine, Hôpitaux Universitaires de Strasbourg, CHU de Hautepierre, avenue Molière, 67200 Strasbourg Cedex, France
| | - Salvatore Chibbaro
- Department of Neurosurgery, Hôpitaux Universitaires de Strasbourg, avenue Molière, 67085 Strasbourg Cedex, France
| | - Georges Noël
- Centre Paul Strauss, 3 rue de la porte de l'Hôpital 67065 Strasbourg, Cedex, France
| | - Izzie J Namer
- Department of Biophysics and Nuclear Medicine, Hôpitaux Universitaires de Strasbourg, CHU de Hautepierre, avenue Molière, 67200 Strasbourg Cedex, France
| | - Sami Heikkinen
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)/CNRS UMR 7104/INSERM U 964/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Johan Auwerx
- 1] Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)/CNRS UMR 7104/INSERM U 964/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France [2] Institut Clinique de la Souris, 1 rue Laurent Fries, 67404 Illkirch, France
| |
Collapse
|
25
|
Abstract
INTRODUCTION Advancements in epigenetic treatments are not only coming from new drugs, but also from modifications or encapsulation of the existing drugs into different formulations leading to greater stability and enhanced delivery to the target site. The epigenome is highly regulated and complex; therefore, it is important that off-target effects of epigenetic drugs be minimized. The step from in vitro to in vivo treatment of these drugs often requires development of a method of effective delivery for clinical translation. AREAS COVERED This review covers epigenetic mechanisms such as DNA methylation, chromatin remodeling and small-RNA-mediated gene regulation. There is a section in the review with examples of diseases where epigenetic alterations lead to impaired pathways, with an emphasis on cancer. Epigenetic drugs, their targets and clinical status are presented. Advantages of using a delivery method for epigenetic drugs as well as examples of current advancements and challenges are also discussed. EXPERT OPINION Epigenetic drugs have the potential to be very effective therapy against a number of diseases, especially cancers and neurological disorders. As with many chemotherapeutics, undesired side effects need to be minimized. Finding a suitable delivery method means reducing side effects and achieving a higher therapeutic index. Each drug may require a unique delivery method exploiting the drug's chemistry or other physical characteristic requiring interdisciplinary participation and would benefit from a better understanding of the mechanisms of action.
Collapse
Affiliation(s)
- Samantha A Cramer
- Lerner Research Institute, Cleveland Clinic, Department of Biomedical Engineering/ND20 , 9500 Euclid Avenue, Cleveland, OH 44195 , USA +1 216 445 9364 ; +1 216 444 9198 ;
| | | | | |
Collapse
|
26
|
Avallone A, Piccirillo MC, Delrio P, Pecori B, Di Gennaro E, Aloj L, Tatangelo F, D’Angelo V, Granata C, Cavalcanti E, Maurea N, Maiolino P, Bianco F, Montano M, Silvestro L, Terranova Barberio M, Roca MS, Di Maio M, Marone P, Botti G, Petrillo A, Daniele G, Lastoria S, Iaffaioli VR, Romano G, Caracò C, Muto P, Gallo C, Perrone F, Budillon A. Phase 1/2 study of valproic acid and short-course radiotherapy plus capecitabine as preoperative treatment in low-moderate risk rectal cancer-V-shoRT-R3 (Valproic acid--short Radiotherapy--rectum 3rd trial). BMC Cancer 2014; 14:875. [PMID: 25421252 PMCID: PMC4289397 DOI: 10.1186/1471-2407-14-875] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/13/2014] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Locally advanced rectal cancer (LARC) is a heterogeneous group of tumors where a risk-adapted therapeutic strategy is needed. Short-course radiotherapy (SCRT) is a more convenient option for LARC patients than preoperative long-course RT plus capecitabine. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with RT and chemotherapy in the treatment of solid tumors. Valproic acid (VPA) is an anti-epileptic drug with HDACi and anticancer activity. In preclinical studies, our group showed that the addition of HDACi, including VPA, to capecitabine produces synergistic antitumour effects by up-regulating thymidine phosphorylase (TP), the key enzyme converting capecitabine to 5-FU, and by downregulating thymidylate synthase (TS), the 5-FU target. METHODS/DESIGN Two parallel phase-1 studies will assess the safety of preoperative SCRT (5 fractions each of 5 Gy, on days 1 to 5) combined with (a) capecitabine alone (increasing dose levels: 500-825 mg/m2/bid), on days 1-21, or (b) capecitabine as above plus VPA (oral daily day -14 to 21, with an intra-patient titration for a target serum level of 50-100 microg/ml) followed by surgery 8 weeks after the end of SCRT, in low-moderate risk RC patients. Also, a randomized phase-2 study will be performed to explore whether the addition of VPA and/or capecitabine to preoperative SCRT might increase pathologic complete tumor regression (TRG1) rate. A sample size of 86 patients (21-22/arm) was calculated under the hypothesis that the addition of capecitabine or VPA to SCRT can improve the TRG1 rate from 5% to 20%, with one-sided alpha = 0.10 and 80% power.Several biomarkers will be evaluated comparing normal mucosa with tumor (TP, TS, VEGF, RAD51, XRCC1, Histones/proteins acetylation, HDAC isoforms) and on blood samples (polymorphisms of DPD, TS, XRCC1, GSTP1, RAD51 and XRCC3, circulating endothelial and progenitors cells; PBMCs-Histones/proteins acetylation). Tumor metabolism will be measured by 18FDG-PET at baseline and 15 days after the beginning of SCRT. DISCUSSION This project aims to improve the efficacy of preoperative treatment of LARC and to decrease the inconvenience and the cost of standard long-course RT. Correlative studies could identify both prognostic and predictive biomarkers and could add new insight in the mechanism of interaction between VPA, capecitabine and RT.EudraCT Number: 2012-002831-28. TRIAL REGISTRATION ClinicalTrials.gov number, NCT01898104.
Collapse
Affiliation(s)
- Antonio Avallone
- />Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Maria Carmela Piccirillo
- />Clinical Trials Unit, Istituto Nazionale Tumori “Fondazione G. Pascale” – IRCCS, Via M. Semmola 80131, Napoli, Italy
| | - Paolo Delrio
- />Colorectal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Biagio Pecori
- />Radiotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Elena Di Gennaro
- />Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Luigi Aloj
- />Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Fabiana Tatangelo
- />Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Valentina D’Angelo
- />Endoscopy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Cinzia Granata
- />Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Ernesta Cavalcanti
- />Clinical Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Nicola Maurea
- />Cardiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Piera Maiolino
- />Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Franco Bianco
- />Gastrointestinal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Massimo Montano
- />Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Lucrezia Silvestro
- />Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Manuela Terranova Barberio
- />Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Maria Serena Roca
- />Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Massimo Di Maio
- />Clinical Trials Unit, Istituto Nazionale Tumori “Fondazione G. Pascale” – IRCCS, Via M. Semmola 80131, Napoli, Italy
| | - Pietro Marone
- />Endoscopy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Gerardo Botti
- />Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Antonella Petrillo
- />Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Gennaro Daniele
- />Clinical Trials Unit, Istituto Nazionale Tumori “Fondazione G. Pascale” – IRCCS, Via M. Semmola 80131, Napoli, Italy
| | - Secondo Lastoria
- />Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Vincenzo R Iaffaioli
- />Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Giovanni Romano
- />Gastrointestinal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Corradina Caracò
- />Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Paolo Muto
- />Radiotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| | - Ciro Gallo
- />Medical Statistics Unit, Second University of Naples, Naples, Italy
| | - Francesco Perrone
- />Clinical Trials Unit, Istituto Nazionale Tumori “Fondazione G. Pascale” – IRCCS, Via M. Semmola 80131, Napoli, Italy
| | - Alfredo Budillon
- />Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione Giovanni Pascale” – IRCCS, Napoli, Italy
| |
Collapse
|
27
|
Dueñas-Gonzalez A, Coronel J, Cetina L, González-Fierro A, Chavez-Blanco A, Taja-Chayeb L. Hydralazine-valproate: a repositioned drug combination for the epigenetic therapy of cancer. Expert Opin Drug Metab Toxicol 2014; 10:1433-44. [PMID: 25154405 DOI: 10.1517/17425255.2014.947263] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION DNA methylation (DNMTi) and histone deacetylase inhibitors (HDACi) are in development for cancer therapy. So far, four epigenetic drugs are approved for myelodysplastic syndrome (MDS) and cutaneous T-cell lymphoma (CTCL). The combination of hydralazine-valproate (TRANSKRIP(™)) is being repositioned as an oral DNMT and HDAC inhibitor. AREAS COVERED Brief discussion on the current status of epigenetic drugs and studies published on the preclinical and clinical development of the hydralazine-valproate combination. EXPERT OPINION Drug repositioning is a strategy for prompt and cost-efficient drug discovery. There is evidence that combining DNMTi with HDACi would be more efficacious than administering each agent on its own. Hydralazine-valproate is safe when used alone or in combination with chemotherapy or chemoradiation. The fact that both drugs are orally administered is another advantage over current epigenetic drugs. This combination is promising but larger studies are needed. Among these, the randomized Phase III trials in advanced and in locally advanced cervical cancer combined with chemotherapy and cisplatin-radiation respectively, would eventually confirm its efficacy. Studies on MDS and CTCL would also eventually prove the efficacy of hydralazine valproate so that in the coming years hydralazine-valproate could have a role in cancer epigenetic therapy.
Collapse
Affiliation(s)
- Alfonso Dueñas-Gonzalez
- Instituto de Investigaciones Biomédicas UNAM/Instituto Nacional de Cancerología Mexico, Unit of Biomedical Research on Cancer , Mexico City , Mexico
| | | | | | | | | | | |
Collapse
|
28
|
Yao ZG, Liang L, Liu Y, Zhang L, Zhu H, Huang L, Qin C. Valproate improves memory deficits in an Alzheimer's disease mouse model: investigation of possible mechanisms of action. Cell Mol Neurobiol 2014; 34:805-12. [PMID: 24939432 PMCID: PMC11488907 DOI: 10.1007/s10571-013-0012-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease (AD) is a very common progressive neurodegenerative disorder affecting the learning and memory abilities in the brain. Key findings from recent studies of epigenetic mechanisms of memory suggest chromatin remodeling disorders via histone hypoacetylation of the lysine residue contribute to the cognitive impairment in AD. Therefore, the deinhibition of histone acetylation induced by histone deacetylases (HDACs) inhibitors contributes to recovery of learning and memory. We show here that the antiepileptic drug sodium valproate (VPA) potently enhanced long-term recognition memory and spatial learning and memory in AD transgenic mice. Possible mechanisms showed VPA could significantly elevate histone acetylation through HDACs activity inhibition and increase plasticity-associated gene expression within the hippocampi of mice. Our study suggests that VPA, serving as a HDACs inhibitor, can be considered as a potential pharmaceutical agent for the improvement of cognitive function in AD.
Collapse
Affiliation(s)
- Zhi-Gang Yao
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Liang Liang
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Yu Liu
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Ling Zhang
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Hua Zhu
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Lan Huang
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Chuan Qin
- Comparative Medical Center, Institute of Laboratory Animal Science, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| |
Collapse
|
29
|
Eskander RN, Tewari KS. Beyond angiogenesis blockade: targeted therapy for advanced cervical cancer. J Gynecol Oncol 2014; 25:249-59. [PMID: 25045438 PMCID: PMC4102744 DOI: 10.3802/jgo.2014.25.3.249] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/28/2014] [Indexed: 11/30/2022] Open
Abstract
The global burden of advanced stage cervical cancer remains significant, particular in resource poor countries where effective screening programs are absent. Unfortunately, a proportion of patients will be diagnosed with advanced stage disease, and may suffer from persistent or recurrent disease despite treatment with combination chemotherapy and radiation. Patients with recurrent disease have a poor salvage rate, with an expected 5-year survival of less than 10%. Recently, significant gains have been made in the antiangiogenic arena; nonetheless the need to develop effective alternate targeted strategies is implicit. As such, a review of molecular targeted therapy in the treatment of this disease is warranted. In an era of biologics, combined therapy with cytotoxic drugs and molecular targeted agents, represents an exciting arena yet to be fully explored.
Collapse
Affiliation(s)
- Ramez N Eskander
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of California Irvine Medical Center, Orange, CA, USA
| | - Krishnansu S Tewari
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of California Irvine Medical Center, Orange, CA, USA
| |
Collapse
|
30
|
Nguyen HT, Tian G, Murph MM. Molecular epigenetics in the management of ovarian cancer: are we investigating a rational clinical promise? Front Oncol 2014; 4:71. [PMID: 24782983 PMCID: PMC3986558 DOI: 10.3389/fonc.2014.00071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/20/2014] [Indexed: 12/21/2022] Open
Abstract
Epigenetics is essentially a phenotypical change in gene expression without any alteration of the DNA sequence; the emergence of epigenetics in cancer research and mainstream oncology is fueling new hope. However, it is not yet known whether this knowledge will translate to improved clinical management of ovarian cancer. In this malignancy, women are still undergoing chemotherapy similar to what was approved in 1978, which to this day represents one of the biggest breakthroughs for treating ovarian cancer. Although liquid tumors are benefiting from epigenetically related therapies, solid tumors like ovarian cancer are not (yet?). Herein, we will review the science of molecular epigenetics, especially DNA methylation, histone modifications and microRNA, but also include transcription factors since they, too, are important in ovarian cancer. Pre-clinical and clinical research on the role of epigenetic modifications is also summarized. Unfortunately, ovarian cancer remains an idiopathic disease, for the most part, and there are many areas of patient management, which could benefit from improved technology. This review will also highlight the evidence suggesting that epigenetics may have pre-clinical utility in pharmacology and clinical applications for prognosis and diagnosis. Finally, drugs currently in clinical trials (i.e., histone deacetylase inhibitors) are discussed along with the promise for epigenetics in the exploitation of chemoresistance. Whether epigenetics will ultimately be the answer to better management in ovarian cancer is currently unknown; but we hope so in the future.
Collapse
Affiliation(s)
- Ha T Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy , Athens, GA , USA
| | - Geng Tian
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy , Athens, GA , USA ; Department of Obstetrics and Gynecology, The Second Hospital of Jilin University , Changchun , China
| | - Mandi M Murph
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy , Athens, GA , USA
| |
Collapse
|
31
|
Garcés-Eisele SJ, Cedillo-Carvallo B, Reyes-Núñez V, Estrada-Marín L, Vázquez-Pérez R, Juárez-Calderón M, Guzmán-García MO, Dueñas-González A, Ruiz-Argüelles A. Genetic selection of volunteers and concomitant dose adjustment leads to comparable hydralazine/valproate exposure. J Clin Pharm Ther 2014; 39:368-75. [PMID: 24702251 DOI: 10.1111/jcpt.12155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Hydralazine is an inhibitor of DNA methyltransferases, whereas valproate interferes with histone deacetylation. In combination, they show a marked synergism in reducing tumour growth as well as development of metastasis and inducing cell differentiation. Hydralazine is metabolized by the highly polymorphic N-acetyltransferase 2. The current pilot study was performed to analyse the pharmacokinetic parameters of a single dose of hydralazine in 24 h (one tablet with 83 mg for slow acetylators and one tablet with 182 mg for fast acetylators) and three fixed doses of valproate (one tablet of controlled liberation with 700 mg every 8 h) in healthy genetically selected volunteers. Selection was performed based on their NAT2 activity as deduced from their genotype. METHODS An open label non-randomized single arm study was conducted in two groups of six healthy volunteers of both genders aged 20-45 years with a body mass index 22·2-26·9 which were classified as fast or slow acetylators after genotyping 3 SNPs that cover 99·9% of the NAT2 variants in the Mexican population. Blood samples were collected predose and serially post-dose in an interval of 48 h. Hydralazine and valproate concentrations were determined by ultra-high performance liquid chromatography (UPLC) coupled to tandem mass spectroscopy (MS/MS). RESULTS AND DISCUSSION The AUC0-48 h and Cmax of hydralazine were almost identical (1410 ± 560 vs. 1446 ± 509 ng h/mL and 93·4 ± 16·7 vs. 112·5 ± 42·1 ng/mL) in both groups with NAT2 genotype-adjusted doses, whereas the multidose parameters of valproate were not significantly affected neither by the selection of the NAT2 genotype (AUC0-48 h 2064 ± 455 vs. 1896 ± 185 μg h/mL; Cmax 96·4 ± 21·1 vs. 88·8 ± 7·2 μg/mL, for the fast and slow acetylators, respectively) nor the co-administration of 83 or 182 mg of hydralazine. WHAT IS NEW AND CONCLUSION Comparable hydralazine exposures (differences in AUC0-inf of only 7%) were observed in this study with genetic selection of volunteers and concomitant dose adjustment. However, the conclusions have yet to be confirmed with a full-powered 2 × 2 crossover study.
Collapse
Affiliation(s)
- S J Garcés-Eisele
- Clínica Ruiz Laboratorios, Puebla, México; Universidad Popular Autónoma del Estado de Puebla, Puebla, México
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Combined effects of a high-fat diet and chronic valproic acid treatment on hepatic steatosis and hepatotoxicity in rats. Acta Pharmacol Sin 2014; 35:363-72. [PMID: 24442146 DOI: 10.1038/aps.2013.135] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 08/23/2013] [Indexed: 01/01/2023]
Abstract
AIM To investigate the potential interactive effects of a high-fat diet (HFD) and valproic acid (VPA) on hepatic steatosis and hepatotoxicity in rats. METHODS Male SD rats were orally administered VPA (100 or 500 mg·kg⁻¹·d⁻¹) combined with HFD or a standard diet for 8 weeks. Blood and liver samples were analyzed to determine lipid levels and hepatic function biomarkers using commercial kit assays. Low-molecular-weight compounds in serum, urine and bile samples were analyzed using a metabonomic approach based on GC/TOF-MS. RESULTS HFD alone induced extensive hepatocyte steatosis and edema in rats, while VPA alone did not cause significant liver lesions. VPA significantly aggravated HFD-induced accumulation of liver lipids, and caused additional spotty or piecemeal necrosis, accompanied by moderate infiltration of inflammatory cells in the liver. Metabonomic analysis of serum, urine and bile samples revealed that HFD significantly increased the levels of amino acids, free fatty acids (FFAs) and 3-hydroxy-butanoic acid, whereas VPA markedly decreased the levels of amino acids, FFAs and the intermediate products of the tricarboxylic acid cycle (TCA) compared with the control group. HFD aggravated VPA-induced inhibition on lipid and amino acid metabolism. CONCLUSION HFD magnifies VPA-induced impairment of mitochondrial β-oxidation of FFAs and TCA, thereby increases hepatic steatosis and hepatotoxicity. The results suggest the patients receiving VPA treatment should be advised to avoid eating HFD.
Collapse
|
33
|
|
34
|
|
35
|
Targeting of histone deacetylases to reactivate tumour suppressor genes and its therapeutic potential in a human cervical cancer xenograft model. PLoS One 2013; 8:e80657. [PMID: 24260446 PMCID: PMC3834007 DOI: 10.1371/journal.pone.0080657] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 10/06/2013] [Indexed: 12/20/2022] Open
Abstract
Aberrant histone acetylation plays an essential role in the neoplastic process via the epigenetic silencing of tumour suppressor genes (TSGs); therefore, the inhibition of histone deacetylases (HDAC) has become a promising target in cancer therapeutics. To investigate the correlation of histone acetylation with clinicopathological features and TSG expression, we examined the expression of acetylated H3 (AcH3), RARβ2, E-cadherin, and β-catenin by immunohistochemistry in 65 cervical squamous cell carcinoma patients. The results revealed that the absence of AcH3 was directly associated with poor histological differentiation and nodal metastasis as well as reduced/negative expression of RARβ2, E-cadherin, and β-catenin in clinical tumour samples. We further demonstrated that the clinically available HDAC inhibitors valproic acid (VPA) and suberoylanilide hydroxamic acid (SAHA), in combination with all-trans retinoic acid (ATRA), can overcome the epigenetic barriers to transcription of RARβ2 in human cervical cancer cells. Chromatin immunoprecipitation analysis showed that the combination treatment increased the enrichment of acetylated histone in the RARβ2-RARE promoter region. In view of these findings, we evaluated the antitumor effects induced by combined VPA and ATRA treatment in a xenograft model implanted with poorly differentiated human squamous cell carcinoma. Notably, VPA restored RARβ2 expression via epigenetic modulation. Additive antitumour effects were produced in tumour xenografts by combining VPA with ATRA treatment. Mechanistically, the combination treatment reactivated the expression of TSGs RARβ2, E-cadherin, P21 (CIP1) , and P53 and reduced the level of p-Stat3. Sequentially, upregulation of involucrin and loricrin, which indicate terminal differentiation, strongly contributed to tumour growth inhibition along with partial apoptosis. In conclusion, targeted therapy with HDAC inhibitors and RARβ2 agonists may represent a novel therapeutic approach for patients with cervical squamous cell carcinoma.
Collapse
|
36
|
Valproic acid, but not levetiracetam, selectively decreases HDAC7 and HDAC2 expression in human ovarian cancer cells. Toxicol Lett 2013; 224:225-32. [PMID: 24200999 DOI: 10.1016/j.toxlet.2013.10.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 10/24/2013] [Accepted: 10/28/2013] [Indexed: 12/20/2022]
Abstract
Histone deacetylases (HDACs) are often overexpressed in cancer cells, leading to altered expression and activity of numerous proteins involved in carcinogenesis. Recent evidence suggests that expression of class I HDACs is increased in ovarian carcinomas and plays a significant role in carcinogenesis and resistance to chemotherapeutic agents. Two compounds, valproic acid (VPA) and levetiracetam (LEV), exhibit HDAC inhibitor (HDACI) activity in various cell types, but data concerning their activity in ovarian cancer are lacking. Here we compared the effects of VPA and LEV as HDACIs, using a human ovarian cancer cell line, OVCAR-3. Cells were cultured with VPA or LEV at concentrations between 1 and 10 mM for 1-24h. HDAC activity was determined by fluorometric assay and confirmed by western blotting. Expression of HDAC genes was determined by real-time PCR and HDAC proteins expression was evaluated by western blotting. Additionally, we used high-performance liquid chromatography to determine whether OVCAR-3 cells can metabolize LEV to its major metabolite, 2-pyrrolidinone-n-butyric acid (PBA), which might exert HDACI activity. LEV, however, had no apparent effect on HDAC activity, or gene and protein expression. The OVCAR-3 cell line was able to metabolize LEV to PBA, but the effect was small. Our observations suggest that VPA should be considered as a possible adjunctive drug in ovarian cancer treatment.
Collapse
|
37
|
Evidence from clinical trials for the use of valproic acid in solid tumors: focus on prostate cancer. ACTA ACUST UNITED AC 2013. [DOI: 10.4155/cli.13.23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
38
|
Chambliss AB, Wu PH, Chen WC, Sun SX, Wirtz D. Simultaneously defining cell phenotypes, cell cycle, and chromatin modifications at single-cell resolution. FASEB J 2013; 27:2667-76. [PMID: 23538711 DOI: 10.1096/fj.12-227108] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heterogeneity of cellular phenotypes in asynchronous cell populations placed in the same biochemical and biophysical environment may depend on cell cycle and chromatin modifications; however, no current method can measure these properties at single-cell resolution simultaneously and in situ. Here, we develop, test, and validate a new microscopy assay that rapidly quantifies global acetylation on histone H3 and measures a wide range of cell and nuclear properties, including cell and nuclear morphology descriptors, cell-cycle phase, and F-actin content of thousands of cells simultaneously, without cell detachment from their substrate, at single-cell resolution. These measurements show that isogenic, isotypic cells of identical DNA content and the same cell-cycle phase can still display large variations in H3 acetylation and that these variations predict specific phenotypic variations, in particular, nuclear size and actin cytoskeleton content, but not cell shape. The dependence of cell and nuclear properties on cell-cycle phase is assessed without artifact-prone cell synchronization. To further demonstrate its versatility, this assay is used to quantify the complex interplay among cell cycle, epigenetic modifications, and phenotypic variations following pharmacological treatments affecting DNA integrity, cell cycle, and inhibiting chromatin-modifying enzymes.
Collapse
Affiliation(s)
- Allison B Chambliss
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | | | | | | | | |
Collapse
|
39
|
Witt D, Burfeind P, von Hardenberg S, Opitz L, Salinas-Riester G, Bremmer F, Schweyer S, Thelen P, Neesen J, Kaulfuss S. Valproic acid inhibits the proliferation of cancer cells by re-expressing cyclin D2. Carcinogenesis 2013; 34:1115-24. [PMID: 23349020 DOI: 10.1093/carcin/bgt019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this study, primary murine prostate cancer (PCa) cells were derived using the well-established TRAMP model. These PCa cells were treated with the histone deacetylase inhibitor, valproic acid (VPA), and we demonstrated that VPA treatment has an antimigrative, antiinvasive and antiproliferative effect on PCa cells. Using microarray analyses, we discovered several candidate genes that could contribute to the cellular effects we observed. In this study, we could demonstrate that VPA treatment of PCa cells causes the re-expression of cyclin D2, a known regulator that is frequently lost in PCa as we could show using immunohistochemical analyses on PCa specimens. We demonstrate that VPA specifically induces the re-expression of cyclin D2, one of the highly conserved D-type cyclin family members, in several cancer cell lines with weak or no cyclin D2 expression. Interestingly, VPA treatment had no effect in fibroblasts, which typically have high basal levels of cyclin D2 expression. The re-expression of cyclin D2 observed in PCa cells is activated by increased histone acetylation in the promoter region of the Ccnd2 gene and represents one underlying molecular mechanism of VPA treatment that inhibits the proliferation of cancer cells. Altogether, our results confirm that VPA is an anticancer therapeutic drug for the treatment of tumors with epigenetically repressed cyclin D2 expression.
Collapse
Affiliation(s)
- Daria Witt
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhang C, Qi X, Shi Y, Sun Y, Li S, Gao X, Yu H. Estimation of trace elements in mace (Myristica fragrans Houtt) and their effect on uterine cervix cancer induced by methylcholanthrene. Biol Trace Elem Res 2012; 149:431-4. [PMID: 22565472 DOI: 10.1007/s12011-012-9443-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 04/25/2012] [Indexed: 10/28/2022]
Abstract
In the present work, trace elemental analysis of mace (Myristica fragrans Houtt) was carried out by the atomic absorption spectrometry technique. The concentrations of various elements analyzed in this medicine were ranked in decreasing order: selenium (Se) > zinc (Zn) > magnesium (Mg) > iron (Fe) > calcium (Ca) > manganese (Mn) > lead (Pb). The concentrations of Mg, Zn, Fe, Mn, Ca, and Se were significantly decreased in serum of methylcholanthrene tumor models (P < 0.001) compared with the control and mace groups. It is consistent with the result of tumor incidence. These trace elements could be directly or indirectly responsible for the antitumor activity of mace. The inorganic elements in this folk remedy can partly account for the antitumor.
Collapse
Affiliation(s)
- Chunjing Zhang
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar 161006, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
41
|
New M, Olzscha H, La Thangue NB. HDAC inhibitor-based therapies: can we interpret the code? Mol Oncol 2012; 6:637-56. [PMID: 23141799 DOI: 10.1016/j.molonc.2012.09.003] [Citation(s) in RCA: 243] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/30/2012] [Indexed: 12/19/2022] Open
Abstract
Abnormal epigenetic control is a common early event in tumour progression, and aberrant acetylation in particular has been implicated in tumourigenesis. One of the most promising approaches towards drugs that modulate epigenetic processes has been seen in the development of inhibitors of histone deacetylases (HDACs). HDACs regulate the acetylation of histones in nucleosomes, which mediates changes in chromatin conformation, leading to regulation of gene expression. HDACs also regulate the acetylation status of a variety of other non-histone substrates, including key tumour suppressor proteins and oncogenes. Histone deacetylase inhibitors (HDIs) are potent anti-proliferative agents which modulate acetylation by targeting histone deacetylases. Interest is increasing in HDI-based therapies and so far, two HDIs, vorinostat (SAHA) and romidepsin (FK228), have been approved for treating cutaneous T-cell lymphoma (CTCL). Others are undergoing clinical trials. Treatment with HDIs prompts tumour cells to undergo apoptosis, and cell-based studies have shown a number of other outcomes to result from HDI treatment, including cell-cycle arrest, cell differentiation, anti-angiogenesis and autophagy. However, our understanding of the key pathways through which HDAC inhibitors affect tumour cell growth remains incomplete, which has hampered progress in identifying malignancies other than CTCL which are likely to respond to HDI treatment.
Collapse
Affiliation(s)
- Maria New
- Department of Oncology, Laboratory of Cancer Biology, University of Oxford, Oxford OX3 7DQ, UK
| | | | | |
Collapse
|
42
|
Yeh HH, Tian M, Hinz R, Young D, Shavrin A, Mukhapadhyay U, Flores LG, Balatoni J, Soghomonyan S, Jeong HJ, Pal A, Uthamanthil R, Jackson JN, Nishii R, Mizuma H, Onoe H, Kagawa S, Higashi T, Fukumitsu N, Alauddin M, Tong W, Herholz K, Gelovani JG. Imaging epigenetic regulation by histone deacetylases in the brain using PET/MRI with ¹⁸F-FAHA. Neuroimage 2012; 64:630-9. [PMID: 22995777 DOI: 10.1016/j.neuroimage.2012.09.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/31/2012] [Accepted: 09/05/2012] [Indexed: 01/12/2023] Open
Abstract
Epigenetic modifications mediated by histone deacetylases (HDACs) play important roles in the mechanisms of different neurologic diseases and HDAC inhibitors (HDACIs) have shown promise in therapy. However, pharmacodynamic profiles of many HDACIs in the brain remain largely unknown due to the lack of validated methods for noninvasive imaging of HDAC expression-activity. In this study, dynamic PET/CT imaging was performed in 4 rhesus macaques using [(18)F]FAHA, a novel HDAC substrate, and [(18)F]fluoroacetate, the major radio-metabolite of [(18)F]FAHA, and fused with corresponding MR images of the brain. Quantification of [(18)F]FAHA accumulation in the brain was performed using a customized dual-tracer pharmacokinetic model. Immunohistochemical analyses of brain tissue revealed the heterogeneity of expression of individual HDACs in different brain structures and cell types and confirmed that PET/CT/MRI with [(18)F]FAHA reflects the level of expression-activity of HDAC class IIa enzymes. Furthermore, PET/CT/MRI with [(18)F]FAHA enabled non-invasive, quantitative assessment of pharmacodynamics of HDAC inhibitor SAHA in the brain.
Collapse
Affiliation(s)
- Hsin-Hsien Yeh
- Department of Experimental Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tanaka T. Preclinical cancer chemoprevention studies using animal model of inflammation-associated colorectal carcinogenesis. Cancers (Basel) 2012; 4:673-700. [PMID: 24213461 PMCID: PMC3712717 DOI: 10.3390/cancers4030673] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/14/2012] [Accepted: 07/06/2012] [Indexed: 12/21/2022] Open
Abstract
Inflammation is involved in all stages of carcinogenesis. Inflammatory bowel disease, such as ulcerative colitis and Crohn’s disease is a longstanding inflammatory disease of intestine with increased risk for colorectal cancer (CRC). Several molecular events involved in chronic inflammatory process are reported to contribute to multi-step carcinogenesis of CRC in the inflamed colon. They include over-production of free radicals, reactive oxygen and nitrogen species, up-regulation of inflammatory enzymes in arachidonic acid biosynthesis pathway, up-regulation of certain cytokines, and intestinal immune system dysfunction. In this article, firstly I briefly introduce our experimental animal models where colorectal neoplasms rapidly develop in the inflamed colorectum. Secondary, data on preclinical cancer chemoprevention studies of inflammation-associated colon carcinogenesis by morin, bezafibrate, and valproic acid, using this novel inflammation-related colorectal carcinogenesis model is described.
Collapse
Affiliation(s)
- Takuji Tanaka
- Cytopatholgy Division, Tohkai Cytopathology Institute, Cancer Research and Prevention (TCI-CaRP), 5-1-2 Minami-uzura, Gifu 500-8285, Japan.
| |
Collapse
|
44
|
Soni GV, Dekker C. Detection of nucleosomal substructures using solid-state nanopores. NANO LETTERS 2012; 12:3180-3186. [PMID: 22554358 DOI: 10.1021/nl301163m] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Histone proteins assemble onto DNA into nucleosomes that control the structure and function of eukaryotic chromatin. More specifically, the structural integrity of nucleosomes regulates gene expression rates and serves as an important early marker for cell apoptosis. Nucleosomal (sub)structures are however hard to detect and characterize. Here, we show that solid-state nanopores are well suited for fast and label-free detection of nucleosomes and its histone subcomplexes. (Nucleo-)protein complexes are individually driven through the nanopore by an applied electric field, which results in characteristic conductance blockades that provide quantitative information on the molecular size of the translocating complex. We observe a systematic dependence of the conductance blockade and translocation time on the molecular weight of the nucleosomal substructures. This allows discriminating and characterizing these protein and DNA-protein complexes at the single-complex level. Finally, we demonstrate the ability to distinguish nucleosomes and dinucleosomes as a first step toward using the nanopore platform to study chromatin arrays.
Collapse
Affiliation(s)
- Gautam V Soni
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Lorentzweg 1, 2628 CJ Delft, The Netherlands
| | | |
Collapse
|
45
|
Brabec V, Griffith DM, Kisova A, Kostrhunova H, Zerzankova L, Marmion CJ, Kasparkova J. Valuable insight into the anticancer activity of the platinum-histone deacetylase inhibitor conjugate, cis-[Pt(NH3)2malSAHA-2H)]. Mol Pharm 2012; 9:1990-9. [PMID: 22591133 DOI: 10.1021/mp300038f] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
cis-[Pt(II)(NH3)2(malSAHA-2H)], a cisplatin adduct conjugated to a potent histone deacetylase inhibitor (HDACi), suberoylanilide hydroxamic acid (SAHA), was previously developed as a potential anticancer agent. This Pt-HDACi conjugate was demonstrated to have comparable cytotoxicity to cisplatin against A2780 ovarian cancer cells but significantly reduced cytotoxicity against a representative normal cell line, NHDF. Thus, with a view to (i) understanding more deeply the effects that may play an important role in the biological (pharmacological) properties of this new conjugate against cancer cells and (ii) developing the next generation of Pt-HDACi conjugates, the cytotoxicity, DNA binding, cellular accumulation and HDAC inhibitory activity of cis-[Pt(II)(NH3)2(malSAHA-2H)] were investigated and are reported herein. cis-[Pt(II)(NH3)2(malSAHA-2H)] was found to have marginally lower cytotoxicity against a panel of cancer cell lines as compared to cisplatin and SAHA. cis-[Pt(II)(NH3)2(malSAHA-2H)] was also found to accumulate better in cancer cells but bind DNA less readily as compared to cisplatin. DNA binding experiments indicated that cis-[Pt(II)(NH3)2(malSAHA-2H)] bound DNA more effectively in cellulo as compared to in cell-free media. Activation of the Pt-HDACi conjugate was therefore investigated. The binding of cis-[Pt(II)(NH3)2(malSAHA-2H)] to DNA was found to be enhanced by the presence of thiol-containing molecules such as glutathione and thiourea, and activation occurred in cytosolic but not nuclear extract of human cancer cells. The activity of cis-[Pt(NH3)2(malSAHA-2H)] as a HDAC inhibitor was also examined; the conjugate exhibited no inhibition of HDAC activity in CH1 cells. In light of these results, novel Pt-HDACi conjugates are currently being developed, with particular emphasis, through subtle structural modifications, on enhancing the rate of DNA binding and enhancing HDAC inhibitory activity.
Collapse
Affiliation(s)
- Viktor Brabec
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 61265 Brno, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
46
|
Zagouri F, Sergentanis TN, Chrysikos D, Filipits M, Bartsch R. Molecularly targeted therapies in cervical cancer. A systematic review. Gynecol Oncol 2012; 126:291-303. [PMID: 22504292 DOI: 10.1016/j.ygyno.2012.04.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 04/03/2012] [Accepted: 04/04/2012] [Indexed: 12/22/2022]
Abstract
Cervical cancer represents the third most common cause of female cancer mortality. Even with the best currently available treatment, a significant proportion of patients will experience recurrence and eventually die. Evidently, there is a clear need for the development of new agents with novel mechanisms of action in this disease. A number of biological agents modulating different signal transduction pathways are currently in clinical development, inhibiting angiogenesis, targeting epidermal growth factor receptor, cell cycle, histone deacetylases, cyclooxygenase-2 (COX-2), or mammalian target of rapamycin (mTOR). This is the first systematic review of the literature to synthesize all available data emerging from trials and evaluate the efficacy and safety of molecularly targeted drugs in cervical cancer. However, it should be stressed that relatively fewer molecularly targeted agents have been tested in cervical cancer in comparison with other cancer types; of note, no related phase 3 trials have been published and consequently no agent has been approved for use in clinical practice. Nevertheless, the promising results of bevacizumab in therapeutic trials for cervical cancer have shown that targeting the VEGF pathway is an attractive therapeutic strategy. As knowledge accumulates on the molecular mechanisms underlying carcinogenesis in the cervix, the anticipated development and assessment of molecularly targeted agents may offer a promising perspective for cervical cancer.
Collapse
Affiliation(s)
- Flora Zagouri
- Comprehensive Cancer Center Vienna, Department of Medicine I/Division of Oncology, Medical University of Vienna, Austria.
| | | | | | | | | |
Collapse
|
47
|
Roy S, Morse D. A full suite of histone and histone modifying genes are transcribed in the dinoflagellate Lingulodinium. PLoS One 2012; 7:e34340. [PMID: 22496791 PMCID: PMC3319573 DOI: 10.1371/journal.pone.0034340] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 03/01/2012] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Dinoflagellates typically lack histones and nucleosomes are not observed in DNA spreads. However, recent studies have shown the presence of core histone mRNA sequences scattered among different dinoflagellate species. To date, the presence of all components required for manufacturing and modifying nucleosomes in a single dinoflagellate species has not been confirmed. METHODOLOGY AND RESULTS Analysis of a Lingulodinium transcriptome obtained by Illumina sequencing of mRNA shows several different copies of each of the four core histones as well as a suite of histone modifying enzymes and histone chaperone proteins. Phylogenetic analysis shows one of each Lingulodinium histone copies belongs to the dinoflagellate clade while the second is more divergent and does not share a common ancestor. All histone mRNAs are in low abundance (roughly 25 times lower than higher plants) and transcript levels do not vary over the cell cycle. We also tested Lingulodinium extracts for histone proteins using immunoblotting and LC-MS/MS, but were unable to confirm histone expression at the protein level. CONCLUSION We show that all core histone sequences are present in the Lingulodinium transcriptome. The conservation of these sequences, even though histone protein accumulation remains below currently detectable levels, strongly suggests dinoflagellates possess histones.
Collapse
Affiliation(s)
- Sougata Roy
- Institut de Recherche en Biologie Végétale, Département de Sciences Biologiques, Université de Montréal, Montréal, Québec, Canada
| | | |
Collapse
|
48
|
Victoriano AFB, Okamoto T. Transcriptional control of HIV replication by multiple modulators and their implication for a novel antiviral therapy. AIDS Res Hum Retroviruses 2012; 28:125-38. [PMID: 22077140 DOI: 10.1089/aid.2011.0263] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Transcriptional regulation is critical for the human immunodeficiency virus 1 (HIV-1) life cycle and is the only step at which the virus amplifies the content of its genetic information. Numerous known and still unknown transcriptional factors, both host and viral, regulate HIV-1 gene expression and latency. This article is a comprehensive review of transcription factors involved in HIV-1 gene expression and presents the significant implications of nuclear factor kappa B (NF-κB) and the HIV-1 transactivator of transcription (Tat) protein. We include recent findings on chromatin remodeling toward HIV transcription and its therapeutic implication is also discussed. The current status of small-molecular-weight compounds that affect HIV transcription is also described.
Collapse
Affiliation(s)
- Ann Florence B. Victoriano
- Department of Molecular and Cellular Biology, Nagoya City University Graduate School for Medical Sciences, Nagoya, Japan
- Japanese Foundation for AIDS Prevention, Tokyo, Japan
| | - Takashi Okamoto
- Department of Molecular and Cellular Biology, Nagoya City University Graduate School for Medical Sciences, Nagoya, Japan
| |
Collapse
|
49
|
Terracciano S, Chini MG, Riccio R, Bruno I, Bifulco G. Design, Synthesis, and Biological Activity of Hydroxamic Tertiary Amines as Histone Deacetylase Inhibitors. ChemMedChem 2012; 7:694-702. [DOI: 10.1002/cmdc.201100531] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 12/22/2011] [Indexed: 12/29/2022]
|
50
|
Abstract
Histone deacetylase (HDAC) inhibitors are a new class of anticancer agents. HDAC inhibitors induce acetylation of histones and nonhistone proteins which are involved in regulation of gene expression and in various cellular pathways including cell growth arrest, differentiation, DNA damage and repair, redox signaling, and apoptosis (Marks, 2010). The U.S. Food and Drug Administration has approved two HDAC inhibitors, vorinostat and romidepsin, for the treatment of cutaneous T-cell lymphoma (Duvic & Vu, 2007; Grant et al., 2010; Marks & Breslow, 2007). Over 20 chemically different HDAC inhibitors are in clinical trials for hematological malignancies and solid tumors. This review considers the mechanisms of resistance to HDAC inhibitors that have been identified which account for the selective effects of these agents in inducing cancer but not normal cell death. These mechanisms, such as functioning Chk1, high levels of thioredoxin, or the prosurvival BCL-2, may also contribute to resistance of cancer cells to HDAC inhibitors.
Collapse
|