Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Toxicol Appl Pharmacol ; 482: 116786, 2024 01.
Article in English | MEDLINE | ID: mdl-38086440

ABSTRACT

Histone deacetylase (HDAC) inhibitors diminish carcinogenesis, metastasis, and cancer cell proliferation by inducing death in cancer cells. Tissue regeneration and organ development are highly dependent on the Hippo signaling pathway. Targeting the dysregulated hippo pathway is an excellent approach for cancer treatment. According to the results of this study, the combination of panobinostat, a histone deacetylase inhibitor, and 5-fluorouracil (5-FU), a chemotherapy drug, can act synergistically to induce apoptosis in gastric cancer cells. The combination of panobinostat and 5-FU was more effective in inhibiting cell viability than either treatment alone by elevating the protein levels of cleaved PARP and cleaved caspase-9. By specifically targeting E-cadherin, vimentin, and MMP-9, the combination of panobinostat and 5-FU significantly inhibited cell migration. Additionally, panobinostat significantly increased the anticancer effects of 5-FU by activating Hippo signaling (Mst 1 and 2, Sav1, and Mob1) and inhibiting the Akt signaling pathway. As a consequence, there was a decrease in the amount of Yap protein. The combination therapy of panobinostat with 5-FU dramatically slowed the spread of gastric cancer in a xenograft animal model by deactivating the Akt pathway and supporting the Hippo pathway. Since combination treatment exhibits much higher anti-tumor potential than 5-FU alone, panobinostat effectively potentiates the anti-tumor efficacy of 5-FU. As a result, it is believed that panobinostat and 5-FU combination therapy will be useful as supplemental chemotherapy in the future.


Subject(s)
Histone Deacetylase Inhibitors , Stomach Neoplasms , Animals , Humans , Histone Deacetylase Inhibitors/therapeutic use , Panobinostat/pharmacology , Fluorouracil/pharmacology , Hippo Signaling Pathway , Stomach Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/pharmacology , Indoles/pharmacology , Cell Proliferation , Apoptosis , Cell Line, Tumor
2.
Clin Cancer Res ; 27(15): 4338-4352, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33994371

ABSTRACT

PURPOSE: We investigated whether targeting chromatin stability through a combination of the curaxin CBL0137 with the histone deacetylase (HDAC) inhibitor, panobinostat, constitutes an effective multimodal treatment for high-risk neuroblastoma. EXPERIMENTAL DESIGN: The effects of the drug combination on cancer growth were examined in vitro and in animal models of MYCN-amplified neuroblastoma. The molecular mechanisms of action were analyzed by multiple techniques including whole transcriptome profiling, immune deconvolution analysis, immunofluorescence, flow cytometry, pulsed-field gel electrophoresis, assays to assess cell growth and apoptosis, and a range of cell-based reporter systems to examine histone eviction, heterochromatin transcription, and chromatin compaction. RESULTS: The combination of CBL0137 and panobinostat enhanced nucleosome destabilization, induced an IFN response, inhibited DNA damage repair, and synergistically suppressed cancer cell growth. Similar synergistic effects were observed when combining CBL0137 with other HDAC inhibitors. The CBL0137/panobinostat combination significantly delayed cancer progression in xenograft models of poor outcome high-risk neuroblastoma. Complete tumor regression was achieved in the transgenic Th-MYCN neuroblastoma model which was accompanied by induction of a type I IFN and immune response. Tumor transplantation experiments further confirmed that the presence of a competent adaptive immune system component allowed the exploitation of the full potential of the drug combination. CONCLUSIONS: The combination of CBL0137 and panobinostat is effective and well-tolerated in preclinical models of aggressive high-risk neuroblastoma, warranting further preclinical and clinical investigation in other pediatric cancers. On the basis of its potential to boost IFN and immune responses in cancer models, the drug combination holds promising potential for addition to immunotherapies.


Subject(s)
Carbazoles/administration & dosage , Carbazoles/pharmacology , Chromatin/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Neuroblastoma/drug therapy , Panobinostat/administration & dosage , Panobinostat/pharmacology , Animals , Drug Combinations , Drug Evaluation, Preclinical , Mice , Tumor Cells, Cultured
3.
Hum Cell ; 34(1): 271-278, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32997328

ABSTRACT

Atypical teratoid/rhabdoid tumor (AT/RT) is a rare intracranial tumor occurring predominantly in young children. The prognosis is poor, and no effective treatment is currently available. To develop novel effective therapies, there is a need for experimental models for AT/RT. In this research, we established a cell line from a patient's AT/RT tissue (designated ATRT_OCGH) and performed drug screening using 164 FDA-approved anti-cancer agents, to identify candidates for therapeutic options. We found that bortezomib, a proteasome inhibitor, was among the agents for which the cell line showed high sensitivity, along with tyrosine kinase inhibitors, topoisomerase inhibitors, and histone deacetylase inhibitors, which are known to exert anti-AT/RT effects. Concomitant use of panobinostat potentiated the inhibitory effect of bortezomib on AT/RT cell proliferation. Our findings may provide a rationale for considering combination therapy of panobinostat and bortezomib for treatment of AT/RT.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Proteasome Inhibitors/pharmacology , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/pathology , Teratoma/drug therapy , Teratoma/pathology , Antineoplastic Agents/administration & dosage , Bortezomib/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Drug Therapy, Combination , Humans , Panobinostat/administration & dosage , Panobinostat/pharmacology , Prognosis , Proteasome Inhibitors/administration & dosage
4.
Article in English | MEDLINE | ID: mdl-31707263

ABSTRACT

The free-living amebae Naegleria, Acanthamoeba, and Balamuthia cause rare but life-threatening infections. All three parasites can cause meningoencephalitis. Acanthamoeba can also cause chronic keratitis and both Balamuthia and Acanthamoeba can cause skin and systemic infections. There are minimal drug development pipelines for these pathogens despite a lack of available treatment regimens and high fatality rates. To identify anti-amebic drugs, we screened 159 compounds from a high-value repurposed library against trophozoites of the three amebae. Our efforts identified 38 compounds with activity against at least one ameba. Multiple drugs that bind the ATP-binding pocket of mTOR and PI3K are active, highlighting these compounds as important inhibitors of these parasites. Importantly, 24 active compounds have progressed at least to phase II clinical studies and overall 15 compounds were active against all three amebae. Based on central nervous system (CNS) penetration or exceptional potency against one amebic species, we identified sixteen priority compounds for the treatment of meningoencephalitis caused by these pathogens. The top five compounds are (i) plicamycin, active against all three free-living amebae and previously U.S. Food and Drug Administration (FDA) approved, (ii) TG02, active against all three amebae, (iii and iv) FDA-approved panobinostat and FDA orphan drug lestaurtinib, both highly potent against Naegleria, and (v) GDC-0084, a CNS penetrant mTOR inhibitor, active against at least two of the three amebae. These results set the stage for further investigation of these clinically advanced compounds for treatment of infections caused by the free-living amebae, including treatment of the highly fatal meningoencephalitis.


Subject(s)
Acanthamoeba/drug effects , Amebiasis/drug therapy , Amoebozoa/drug effects , Antiprotozoal Agents/pharmacology , Central Nervous System Protozoal Infections/drug therapy , Naegleria/drug effects , Amebiasis/parasitology , Carbazoles/pharmacology , Carbazoles/therapeutic use , Cell Culture Techniques , Central Nervous System Protozoal Infections/parasitology , Culture Media , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Inhibitory Concentration 50 , Oxazines/pharmacology , Oxazines/therapeutic use , Panobinostat/pharmacology , Panobinostat/therapeutic use , Plicamycin/pharmacology , Plicamycin/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
5.
Sci Transl Med ; 11(519)2019 11 20.
Article in English | MEDLINE | ID: mdl-31748226

ABSTRACT

Diffuse midline gliomas (DMGs) are universally lethal malignancies occurring chiefly during childhood and involving midline structures of the central nervous system, including thalamus, pons, and spinal cord. These molecularly related cancers are characterized by high prevalence of the histone H3K27M mutation. In search of effective therapeutic options, we examined multiple DMG cultures in sequential quantitative high-throughput screens (HTS) of 2706 approved and investigational drugs. This effort generated 19,936 single-agent dose responses that inspired a series of HTS-enabled drug combination assessments encompassing 9195 drug-drug examinations. Top combinations were validated across patient-derived cell cultures representing the major DMG genotypes. In vivo testing in patient-derived xenograft models validated the combination of the multi-histone deacetylase (HDAC) inhibitor panobinostat and the proteasome inhibitor marizomib as a promising therapeutic approach. Transcriptional and metabolomic surveys revealed substantial alterations to key metabolic processes and the cellular unfolded protein response after treatment with panobinostat and marizomib. Mitigation of drug-induced cytotoxicity and basal mitochondrial respiration with exogenous application of nicotinamide mononucleotide (NMN) or exacerbation of these phenotypes when blocking nicotinamide adenine dinucleotide (NAD+) production via nicotinamide phosphoribosyltransferase (NAMPT) inhibition demonstrated that metabolic catastrophe drives the combination-induced cytotoxicity. This study provides a comprehensive single-agent and combinatorial drug screen for DMG and identifies concomitant HDAC and proteasome inhibition as a promising therapeutic strategy that underscores underrecognized metabolic vulnerabilities in DMG.


Subject(s)
Brain Neoplasms/drug therapy , Drug Evaluation, Preclinical , Glioma/drug therapy , High-Throughput Screening Assays/methods , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Stem Neoplasms/drug therapy , Cell Death , Cell Line, Tumor , Drug Synergism , Female , Glioma/genetics , Glioma/metabolism , Humans , Lactones/pharmacology , Lactones/therapeutic use , Male , Metabolomics , Mice , Panobinostat/pharmacology , Panobinostat/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Reproducibility of Results , Sequence Analysis, RNA , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
6.
Sci Rep ; 8(1): 14702, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279437

ABSTRACT

Persistence of latent HIV-1 in macrophages (MACs) and T-helper lymphocytes (THLs) remain a major therapeutic challenge. Currently available latency reversing agents (LRAs) are not very effective in vivo. Therefore, understanding of physiologic mechanisms that dictate HIV-1 latency/reactivation in reservoirs is clearly needed. Mesenchymal stromal/stem cells (MSCs) regulate the function of immune cells; however, their role in regulating virus production from latently-infected MACs & THLs is not known. We documented that exposure to MSCs or their conditioned media (MSC-CM) rapidly increased HIV-1 p24 production from the latently-infected U1 (MAC) & ACH2 (THL) cell lines. Exposure to MSCs also increased HIV-1 long terminal repeat (LTR) directed gene expression in the MAC and THL reporter lines, U937-VRX and J-Lat (9.2), respectively. MSCs exposed to CM from U1 cells (U1-CM) showed enhanced migratory ability towards latently-infected cells and retained their latency-reactivation potential. Molecular studies showed that MSC-mediated latency-reactivation was dependent upon both the phosphatidyl inositol-3-kinase (PI3K) and nuclear factor-κB (NFκB) signaling pathways. The pre-clinically tested inhibitors of PI3K (PX-866) and NFκB (CDDO-Me) suppressed MSC-mediated HIV-1 reactivation. Furthermore, coexposure to MSC-CM enhanced the latency-reactivation efficacy of the approved LRAs, vorinostat and panobinostat. Our findings on MSC-mediated latency-reactivation may provide novel strategies against persistent HIV-1 reservoirs.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/physiology , Mesenchymal Stem Cells/metabolism , Virus Activation/drug effects , Anti-HIV Agents/therapeutic use , Cell Line , Culture Media, Conditioned/pharmacology , Drug Evaluation, Preclinical , Gene Expression Regulation, Viral/drug effects , Gonanes/pharmacology , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , Humans , Mesenchymal Stem Cells/drug effects , NF-kappa B/metabolism , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Panobinostat/pharmacology , Panobinostat/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Virus Latency/drug effects , Vorinostat/pharmacology , Vorinostat/therapeutic use
7.
Br J Pharmacol ; 174(21): 3766-3779, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28094846

ABSTRACT

BACKGROUND AND PURPOSE: In the pharmaceutical industry risk assessments of chronic cardiac safety liabilities are mostly performed during late stages of preclinical drug development using in vivo animal models. Here, we explored the potential of human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) to detect chronic cardiac risks such as drug-induced cardiomyocyte toxicity. EXPERIMENTAL APPROACH: Video microscopy-based motion field imaging was applied to evaluate the chronic effect (over 72 h) of cardiotoxic drugs on the contractile motion of hiPS-CMs. In parallel, the release of cardiac troponin I (cTnI), heart fatty acid binding protein (FABP3) and N-terminal pro-brain natriuretic peptide (NT-proBNP) was analysed from cell medium, and transcriptional profiling of hiPS-CMs was done at the end of the experiment. KEY RESULTS: Different cardiotoxic drugs altered the contractile motion properties of hiPS-CMs together with increasing the release of cardiac biomarkers. FABP3 and cTnI were shown to be potential surrogates to predict cardiotoxicity in hiPS-CMs, whereas NT-proBNP seemed to be a less valuable biomarker. Furthermore, drug-induced cardiotoxicity produced by chronic exposure of hiPS-CMs to arsenic trioxide, doxorubicin or panobinostat was associated with different profiles of changes in contractile parameters, biomarker release and transcriptional expression. CONCLUSION AND IMPLICATIONS: We have shown that a parallel assessment of motion field imaging-derived contractile properties, release of biomarkers and transcriptional changes can detect diverse mechanisms of chronic drug-induced cardiac liabilities in hiPS-CMs. Hence, hiPS-CMs could potentially improve and accelerate cardiovascular de-risking of compounds at earlier stages of drug discovery. LINKED ARTICLES: This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.


Subject(s)
Antineoplastic Agents/toxicity , Cardiotoxicity/etiology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Arsenic Trioxide , Arsenicals , Biomarkers/metabolism , Cardiotoxicity/physiopathology , Cells, Cultured , Doxorubicin/toxicity , Drug Evaluation, Preclinical/methods , Humans , Hydroxamic Acids/toxicity , Indoles/toxicity , Microscopy, Video , Muscle Contraction/drug effects , Myocytes, Cardiac/pathology , Oxides/toxicity , Panobinostat
8.
Future Oncol ; 13(6): 477-488, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27776419

ABSTRACT

Multiple myeloma (MM) is the second most common blood cancer following non-Hodgkin's lymphoma. While the treatments for MM have improved over the past decade, for the most part, it remains an incurable disease. For this reason newer therapeutic agents are needed to combat this malignancy. Panobinostat is a pan-deacetylase inhibitor that impedes protein destruction by disturbing the enzymatic activity of deacetylases. It was US FDA approved in February 2015 for the management of relapsed/refractory MM in combination with bortezomib and dexamethasone. Several trials are ongoing, exploring the utility of panobinostat in various other settings for the management of MM. This review will detail the biology, clinical efficacy and potential future applications of panobinostat in the treatment of MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Multiple Myeloma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , Disease Management , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Multiple Myeloma/metabolism , Panobinostat , Treatment Outcome
9.
Methods Mol Biol ; 1510: 127-148, 2017.
Article in English | MEDLINE | ID: mdl-27761818

ABSTRACT

Histone deacetylase (HDAC) inhibitors are promising drugs. These agents lead to growth inhibition, cell cycle arrest, premature senescence, and apoptosis of malignant cells. Aim of our studies was to determine the efficacy of HDAC inhibitors on the clinically most relevant population of human leukemic progenitor cells in vitro. We here present stroma-free long-term cultures (LTC) of primary acute myeloid leukemia (AML) cells as a useful system for drug sensitivity testing in functional assays. AML-LTC are established by isolating mononuclear cells from peripheral blood samples of AML patients followed by selection of CD34+ progenitor cells. AML-LTC cells can be maintained in liquid culture supplemented with cytokines and utilized for in vitro analyses to assess proliferation, apoptosis, expression of surface proteins or intracellular proteins and signal transduction, respectively.


Subject(s)
Biomarkers, Tumor/genetics , Cell Culture Techniques , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Neoplastic Stem Cells/drug effects , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cell Separation/methods , Cytokines/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Panobinostat , Primary Cell Culture , Signal Transduction , Survivin , Tumor Cells, Cultured , Valproic Acid/pharmacology , Vorinostat
10.
J Ovarian Res ; 9(1): 58, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27633667

ABSTRACT

BACKGROUND: Pre-clinical studies have demonstrated that natural and synthetic histone deacetylase (HDAC) inhibitors can impede the in vitro and in vivo growth of cell lines from a variety of gynecologic and other malignancies. We investigated the anti-tumor activity of panobinostat (LBH589) both in vitro and in vivo as either a single agent or in combination with conventional cytotoxic chemotherapy using patient-derived xenograft (PDX) models of primary serous ovarian tumors. METHODS: The ovarian cancer cell lines OVCAR8, SKOV3 and their paclitaxel-resistant derivatives OVCAR8-TR and SKOV3-TR were treated with increasing doses of LBH589. The effect of LBH589 on cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Serially transplanted primary human high-grade serous ovarian adenocarcinoma tissue was utilized to generate xenografts in 6-week old female NOD/SCID mice. The mice were then randomized into one of 4 treatment groups: (1) vehicle control; (2) paclitaxel and carboplatin (P/C); (3) LBH589; or (4) P/C + LBH589. Mice were treated for 21 days and tumor volumes and mouse weights were obtained every 3 days. These experiments were performed in triplicate with three different patient derived tumors. Wilcoxan rank-sum testing was utilized to assess tumor volume differences. RESULTS: In vitro treatment with LBH589 significantly reduced the viability of both taxol-sensitive and taxol-resistant ovarian cancer cell lines (p < 0.01). In vivo treatment with LBH589 alone appeared tumorstatic and reduced tumor growth when compared to vehicle treatment (p < 0.007) after 21 days. This single agent activity was confirmed in two additional experiments with other PDX tumors (p < 0.03, p < 0.05). A potential additive effect of LBH589 and P/C, manifested as enhanced tumor regression with the addition of LBH589 compared to vehicle (p < 0.02), in one of the three analyzed serous PDX models. CONCLUSIONS: Our findings suggest that pan-HDAC inhibition with panobinostat precludes the growth of ovarian cancer cell lines in vitro and PDXs in vivo. Added benefit of LBH589 to standard P/C therapy was observed in one of three PDX models suggesting improved response in a subset of serous ovarian cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Grading , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Panobinostat , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Expert Opin Investig Drugs ; 25(9): 1117-31, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27485472

ABSTRACT

INTRODUCTION: Therapeutic strategies in patients with acute myeloid leukemia (AML) have not changed significantly over the last decades. Appropriate strategies are ultimately driven by the assessment of patients' fitness to define suitability for intensive induction chemotherapy, which produces high initial remission rates but, increased likelihood of relapse. Old/unfit AML patients still represent an urgent and unmet therapeutic need. Epigenetic deregulation represents a strategic characteristic of AML pathophysiology whereby aberrant gene transcription provides an advantage to leukemic cell survival. Efforts to re-establish impaired epigenetic regulation include hypomethylating agents and histone deacetylase inhibitors (HDACi). AREAS COVERED: The review discusses the underlying mechanisms leading to disruption of lysine acetyltransferases (KAT or HAT)/deacetylase (KDAC or HDAC) balance and the rationale for using the HDACi panobinostat (LBH-589) in AML. EXPERT OPINION: Although panobinostat has produced significant results in myeloma, its efficacy remains limited in AML. Panobinostat exerts pleiotropic activity and lack of specificity, which likely contributes to its inadequate safety in elderly AML patients. Phase I-II trials, utilizing panobinostat associated with well-known chemotherapeutic agents are ongoing and combinations with other druggable targets may likely be evaluated in future trials. The clinical use of this HDACi in AML the near future does not appearing promising.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Clinical Trials as Topic , DNA Methylation/drug effects , Drug Evaluation, Preclinical , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacokinetics , Leukemia, Myeloid, Acute/genetics , Panobinostat , Treatment Outcome
12.
Proc Natl Acad Sci U S A ; 113(11): 3018-23, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929321

ABSTRACT

Mutations in chromatin-modifying proteins and transcription factors are commonly associated with a wide variety of cancers. Through gain- or loss-of-function, these mutations may result in characteristic alterations of accessible chromatin, indicative of shifts in the landscape of regulatory elements genome-wide. The identification of compounds that reverse a specific chromatin signature could lead to chemical probes or potential therapies. To explore whether chromatin accessibility could serve as a platform for small molecule screening, we adapted formaldehyde-assisted isolation of regulatory elements (FAIRE), a chemical method to enrich for nucleosome-depleted genomic regions, as a high-throughput, automated assay. After demonstrating the validity and robustness of this approach, we applied this method to screen an epigenetically targeted small molecule library by evaluating regions of aberrant nucleosome depletion mediated by EWSR1-FLI1, the chimeric transcription factor critical for the bone and soft tissue tumor Ewing sarcoma. As a class, histone deacetylase inhibitors were greatly overrepresented among active compounds. These compounds resulted in diminished accessibility at targeted sites by disrupting transcription of EWSR1-FLI1. Capitalizing on precise differences in chromatin accessibility for drug discovery efforts offers significant advantages because it does not depend on the a priori selection of a single molecular target and may detect novel biologically relevant pathways.


Subject(s)
Chromatin/drug effects , High-Throughput Screening Assays/methods , Oncogene Proteins, Fusion/antagonists & inhibitors , Transcription, Genetic/drug effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Chromatin/ultrastructure , Drug Design , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Molecular Targeted Therapy , Nucleosomes/ultrastructure , Oncogene Proteins, Fusion/genetics , Panobinostat , Phenylbutyrates/pharmacology , Sarcoma, Ewing/pathology , Small Molecule Libraries , Vorinostat
13.
Am J Health Syst Pharm ; 73(7): 441-50, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27001985

ABSTRACT

PURPOSE: The mechanism of action, pharmacodynamics, pharmacokinetics, clinical efficacy, interaction potential, adverse effects, and place in therapy of panobinostat are reviewed. SUMMARY: Panobinostat (Farydak, Novartis) is a novel pan-deacetylase inhibitor approved for use in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (RRMM) who have received at least two regimens containing an immunomodulatory drug and bortezomib. National Comprehensive Cancer Network (NCCN) guidelines recommend the use of panobinostat plus bortezomib and dexamethasone as a preferred regimen for previously treated multiple myeloma (MM). A Phase III trial comparing panobinostat or placebo use in combination with bortezomib and dexamethasone demonstrated improved median progression-free survival in the panobinostat group (11.99 months [95% CI, 10.33-12.94 months] versus 8.08 months [95% CI, 7.56-9.23 months]; hazard ratio, 0.63 [95% CI, 0.52-0.76]; p < 0.0001), as well as a significantly higher rate of complete or near complete response (27.6% [95% CI, 23.2-32.4%] versus 15.7% [95% CI, 12.2-19.8%]; p = 0.00006). Common grade 3 or 4 laboratory abnormalities and adverse events associated with panobinostat include thrombocytopenia, lymphopenia, diarrhea, asthenia, fatigue, and peripheral neuropathy. CONCLUSION: Panobinostat is a promising alternative to well-studied, NCCN-recommended regimens for the treatment of RRMM. It has demonstrated efficacy when used in combination with bortezomib and dexamethasone for the treatment of patients with MM who have received at least two prior regimens including bortezomib and an immunomodulatory agent. Despite the observed benefits, concern regarding toxicity may limit panobinostat use in practice.


Subject(s)
Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Indoles/administration & dosage , Multiple Myeloma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bortezomib/administration & dosage , Clinical Trials, Phase II as Topic/methods , Clinical Trials, Phase III as Topic/methods , Dexamethasone/administration & dosage , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/epidemiology , Panobinostat , Recurrence , Treatment Outcome
14.
Best Pract Res Clin Haematol ; 28(2-3): 124-32, 2015.
Article in English | MEDLINE | ID: mdl-26590769

ABSTRACT

Despite the steady increase in the number of stem cell transplants performed since 1980 and improvements in survival rates, disease relapse remains the major cause of death after HLA matched sibling and unrelated donor transplants for acute myeloid leukemia (AML). Given this situation, maintenance therapy after transplant may be an appropriate strategy to reduce the relapse rate and prolong survival. A number of agents are being investigated as maintenance therapy after stem cell transplant in AML patients, including azacitidine, decitabine, and other agents. This paper focuses on the role of maintenance treatment to reduce the risk of relapse after transplant.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Maintenance Chemotherapy/methods , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Benzothiazoles/therapeutic use , Decitabine , Humans , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Panobinostat , Phenylurea Compounds/therapeutic use , Recurrence , Siblings , Sorafenib , Survival Analysis , Transplantation, Homologous , Unrelated Donors
15.
Nat Commun ; 6: 6838, 2015 Apr 27.
Article in English | MEDLINE | ID: mdl-25913720

ABSTRACT

Histone deacetylase inhibitors (HDACi) are approved for treating certain haematological malignancies, however, recent evidence also illustrates they are modulators of the immune system. In experimental models, HDACi are particularly potent against malignancies originating from the B-lymphocyte lineage. Here we examine the ability of this class of compounds to modify both protective and autoimmune antibody responses. In vitro, HDACi affect B-cell proliferation, survival and differentiation in an HDAC-class-dependent manner. Strikingly, treatment of lupus-prone Mrl/lpr mice with the HDACi panobinostat significantly reduces autoreactive plasma-cell numbers, autoantibodies and nephritis, while other immune parameters remain largely unaffected. Immunized control mice treated with panobinostat or the clinically approved HDACi vorinostat have significantly impaired primary antibody responses, but these treatments surprisingly spare circulating memory B cells. These studies indicate that panobinostat is a potential therapy for B-cell-driven autoimmune conditions and HDACi do not induce major long-term detrimental effects on B-cell memory.


Subject(s)
B-Lymphocytes/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Female , Germinal Center/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Immunologic Memory/drug effects , Indoles/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Male , Mice, Inbred C57BL , Panobinostat
16.
PLoS One ; 9(12): e115068, 2014.
Article in English | MEDLINE | ID: mdl-25500581

ABSTRACT

Mutations in the WNT/beta-catenin pathway are present in the majority of all sporadic colorectal cancers (CRCs), and histone deacetylase inhibitors induce apoptosis in CRC cells with such mutations. This apoptosis is counteracted by (1) the signaling heterogeneity of CRC cell populations, and (2) the survival pathways induced by mitogens secreted from apoptotic cells. The phenomena of signaling heterogeneity and apoptosis-induced survival constitute the immediate mechanisms of resistance to histone deacetylase inhibitors, and probably other chemotherapeutic agents. We explored the strategy of augmenting CRC cell death by inhibiting all survival pathways induced by the pro-apoptotic agent LBH589, a histone deacetylase inhibitor: AKT, JAK/STAT, and ERK signaling. The apoptosis-enhancing ability of a cocktail of synthetic inhibitors of proliferation was compared to the effects of the natural product propolis. We utilized colorectal adenoma, drug-sensitive and drug-resistant colorectal carcinoma cells to evaluate the apoptotic potential of the combination treatments. The results suggest that an effective approach to CRC combination therapy is to combine apoptosis-inducing drugs (e.g., histone deacetylase inhibitors, such as LBH589) with agents that suppress all compensatory survival pathways induced during apoptosis (such as the cocktail of inhibitors of apoptosis-associated proliferation). The same paradigm can be applied to a CRC prevention approach, as the apoptotic effect of butyrate, a diet-derived histone deacetylase inhibitor, is augmented by other dietary agents that modulate survival pathways (e.g., propolis and coffee extract). Thus, dietary supplements composed by fermentable fiber, propolis, and coffee extract may effectively counteract neoplastic growth in the colon.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dietary Supplements , Histone Deacetylase Inhibitors/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Indoles/administration & dosage , Panobinostat , Propolis/administration & dosage
17.
BMJ Case Rep ; 20122012 Dec 17.
Article in English | MEDLINE | ID: mdl-23257272

ABSTRACT

Here, we report the case of a patient, who showed an antitumour response to a new combination therapy of sorafenib and the histon deacetylase inhibitor panobinostat (LBH-589). D-CEUS (Dynamic contrast-enhanced ultrasonography) was able to predict response to the new therapy regime and may be an interesting tool in the early evaluation of response to therapy. It might be especially useful to differentiate between responders and non-responders of new-targeted pharmaceuticals like multikinase inhibitors in hepatocellular carcinomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/drug therapy , Aged , Carcinoma, Hepatocellular/pathology , Contrast Media , Fatal Outcome , Humans , Hydroxamic Acids/administration & dosage , Indoles/administration & dosage , Liver Neoplasms/pathology , Male , Neoplasm Staging , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Panobinostat , Phenylurea Compounds/administration & dosage , Sorafenib , Ultrasonography/methods
18.
J Hepatol ; 56(6): 1343-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22322234

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is a heterogeneous cancer in which sorafenib is the only approved systemic therapy. Histone deacetylases (HDAC) are commonly dysregulated in cancer and therefore represent promising targets for therapies, however their role in HCC pathogenesis is still unknown. We analyzed the expression of 11 HDACs in human HCCs and assessed the efficacy of the pan-HDAC inhibitor panobinostat alone and in combination with sorafenib in preclinical models of liver cancer. METHODS: Gene expression and copy number changes were analyzed in a cohort of 334 human HCCs, while the effects of panobinostat and sorafenib were evaluated in three liver cancer cell lines and a murine xenograft model. RESULTS: Aberrant HDAC expression was identified and validated in 91 and 243 HCCs, respectively. Upregulation of HDAC3 and HDAC5 mRNAs was significantly correlated with DNA copy number gains. Inhibiting HDACs with panobinostat led to strong anti-tumoral effects in vitro and vivo, enhanced by the addition of sorafenib. Cell viability and proliferation declined, while apoptosis and autophagy increased. Panobinostat increased histone H3 and HSP90 acetylation, downregulated BIRC5 (survivin) and upregulated CDH1. Combination therapy with panobinostat and sorafenib significantly decreased vessel density, and most significantly decreased tumor volume and increased survival in HCC xenografts. CONCLUSIONS: Aberrant expression of several HDACs and copy number gains of HDAC3 and HDAC5 occur in HCC. Treatment with panobinostat combined with sorafenib demonstrated the highest preclinical efficacy in HCC models, providing the rationale for clinical studies with this novel combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzenesulfonates/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Liver Neoplasms/drug therapy , Pyridines/administration & dosage , Animals , Antigens, CD , Apoptosis/drug effects , Autophagy , Cadherins/genetics , Carcinoma, Hepatocellular/pathology , Drug Synergism , Humans , Indoles , Inhibitor of Apoptosis Proteins/genetics , Liver Neoplasms/pathology , Niacinamide/analogs & derivatives , Panobinostat , Phenylurea Compounds , Polymorphism, Single Nucleotide , RNA, Messenger/analysis , Sorafenib , Survivin , Xenograft Model Antitumor Assays
19.
Clin Cancer Res ; 18(2): 408-16, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22038994

ABSTRACT

PURPOSE: Metastasis is responsible for the death of most cancer patients, yet few therapeutic agents are available which specifically target the molecular events that lead to metastasis. We recently showed that inactivating mutations in the tumor suppressor gene BAP1 are closely associated with loss of melanocytic differentiation in uveal melanoma (UM) and metastasis. The purpose of this study was to identify therapeutic agents that reverse the phenotypic effects of BAP1 loss in UM. EXPERIMENTAL DESIGN: In silico screens were done to identify therapeutic compounds predicted to differentiate UM cells using Gene Set Enrichment Analysis and Connectivity Map databases. Valproic acid (VPA), trichostatin A, LBH-589, and suberoylanilide hydroxamic acid were evaluated for their effects on UM cells using morphologic evaluation, MTS viability assays, bromodeoxyuridine incorporation, flow cytometry, clonogenic assays, gene expression profiling, histone acetylation and ubiquitination assays, and a murine xenograft tumorigenicity model. RESULTS: Histone deacetylase (HDAC) inhibitors induced morphologic differentiation, cell-cycle exit, and a shift to a differentiated, melanocytic gene expression profile in cultured UM cells. VPA inhibited the growth of UM tumors in vivo. CONCLUSIONS: These findings suggest that HDAC inhibitors may have therapeutic potential for inducing differentiation and prolonged dormancy of micrometastatic disease in UM.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Melanoma/drug therapy , Uveal Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Chemoradiotherapy, Adjuvant , Computer Simulation , Gene Knockdown Techniques , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Models, Biological , Neoplasm Micrometastasis/prevention & control , Panobinostat , Tumor Burden/drug effects , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Uveal Neoplasms/pathology , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Vorinostat , Xenograft Model Antitumor Assays
20.
Br J Cancer ; 106(1): 107-15, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22116303

ABSTRACT

BACKGROUND: We examine the potential value of a series of clinically relevant PI3K-mTOR inhibitors alone, or in combination with histone deacetylase inhibitors, in a model of head and neck squamous cell carcinoma (HNSCC). METHODS: Head and neck squamous cell carcinoma cell lines, human keratinocyte and HNSCC xenograft models were treated with histone deacetylase inhibitors (HDACIs) and new generation PI3K and dual PI3K-mTOR inhibitors either alone or in combination. Cell and tumour tissue viability and proliferation were then determined in vitro and in vivo. RESULTS: Phosphatidylinositol-3-phosphate kinase, AKT and dual PI3K-mTOR inhibitors caused marked in vitro enhancement of cytotoxicity induced by HDACIs in HNSCC cancer cells. This effect correlates with AKT inhibition and is attenuated by expression of constitutively active AKT. Histone deacetylase inhibitor and phosphatidylinositol-3-phosphate kinase inhibitors (PI3KIs) inhibited tumour growth in xenograft models of HNSCC. Importantly, we observed intratumoural HDAC inhibition and PI3K inhibition as assessed by histone H3 acetylation status and phospho-AKT staining, respectively. However, we saw no evidence of improved efficacy with an HDACI/PI3KI combination. INTERPRETATION: That PI3K and dual PI3K-mTOR inhibitors possess antitumour effect against HNSCC in vivo.


Subject(s)
Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Histone Deacetylase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , Indoles , Mice , Mice, Inbred NOD , Mice, SCID , Panobinostat , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL