Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Mol Pharm ; 17(7): 2599-2611, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32379457

ABSTRACT

Parenteral chemotherapy is usually administered intravenously, although patient preference and health economics suggest the subcutaneous (sc) route could be an attractive alternative. However, due to the low aqueous solubility of hydrophobic drugs and injection volume limitations, the total amount of drug that can be administered in a single sc injection is frequently insufficient. We have developed hyaluronidase coated nanoparticles (NPs) that efficiently encapsulate such drugs, thus addressing both issues and allowing sufficient amounts of hydrophobic drug to be administered and absorbed effectively. CUDC-101, a poorly water-soluble multitargeted anticancer drug that simultaneously inhibits the receptor tyrosine kinases (RTKs) EGFR and HER2, as well as histone deacetylase (HDAC), was encapsulated in polymeric Molecular Envelope Technology (MET) NPs. The role of polymer chemistry, formulation parameters, and coating with hyaluronidase (HYD) on MET-CUDC-101 NP formulations was examined and optimized to yield high drug loading and colloidal stability, and, after freeze-drying, stable storage at room temperature for up to 90 days. The pharmacokinetic studies in healthy rats showed that plasma AUC0-24h after sc administration correlates tightly with formulation physical chemistry, specifically in vitro colloidal stability. Compared to uncoated NPs, the HYD-coating doubled the drug plasma exposure. In a murine A431 xenograft model, the coated HYD-MET-CUDC-101 NPs at a dose equivalent to 90 mg kg-1 CUDC-101 increased the survival time from 15 days (control animals treated with hyaluronidase alone) to 43 days. Polymer MET nanoparticles coated with hyaluronidase enabled the subcutaneous delivery of a hydrophobic drug with favorable therapeutic outcomes.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Drug Delivery Systems/methods , Histone Deacetylase Inhibitors/pharmacology , Hyaluronoglucosaminidase/chemistry , Hydroxamic Acids/administration & dosage , Nanoparticles/chemistry , Polymers/chemistry , Quinazolines/administration & dosage , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chitosan/analogs & derivatives , Chitosan/chemistry , Drug Carriers/chemistry , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/blood , Histone Deacetylases , Histones/metabolism , Hydrophobic and Hydrophilic Interactions , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Mice , Mice, Nude , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Particle Size , Quinazolines/blood , Quinazolines/pharmacokinetics , Rats , Solubility , Xenograft Model Antitumor Assays
2.
Invest New Drugs ; 38(4): 977-989, 2020 08.
Article in English | MEDLINE | ID: mdl-31493129

ABSTRACT

Cancer treatment is challenging, mainly due to high levels of drug toxicity and the resistance of tumours to chemotherapy. Hydroxamic acid derivatives have recently aroused attention due to their potential to treat malignancies. In the present study, we sought to investigate the anticancer effects of a new series of synthetic acetohydroxamates. The in vitro cytotoxic and antiproliferative effects of 11 synthetic acetohydroxamates were evaluated against the melanoma cell line A375. Apoptosis, cell cycle, and autophagy assays were employed to elucidate the cell death pathways induced by the compounds. The in vivo pharmacokinetic profiles of the most promising compounds were determined in CD-1 mice, while the in vivo antitumour efficacies were evaluated using the A375 melanoma xenograft model in nude mice. MTT assays revealed that all compounds presented concentration-dependent cytotoxicity against the A375 cell line. AKS 61 produced the most favourable antiproliferative activity according to the sulphorhodamine B and clonogenic assays. AKS 61 treatment resulted in decreased mitochondrial membrane potential and increased apoptosis and autophagy in the A375 cell line. However, AKS 61 failed to prevent in vivo tumour growth in a melanoma xenograft, whereas compound AKS 7 was able to inhibit tumour growth when administered orally. These in vivo findings may be explained by a more favourable pharmacokinetic profile presented by AKS 7 when compared to AKS 61. Taken together, these results suggest that acetohydroxamates have potential anticancer effects and will guide future optimisation of these molecules to allow for further non-clinical development.


Subject(s)
Antineoplastic Agents/therapeutic use , Hydroxamic Acids/therapeutic use , Melanoma/drug therapy , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Male , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Tumor Burden/drug effects
3.
Acta Neuropathol Commun ; 6(1): 103, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30270813

ABSTRACT

Chemotherapy-induced cognitive impairment (CICI) is a commonly reported neurotoxic side effect of chemotherapy, occurring in up to 75% cancer patients. CICI manifests as decrements in working memory, executive functioning, attention, and processing speed, and greatly interferes with patients' daily performance and quality of life. Currently no treatment for CICI has been approved by the US Food and Drug Administration. We show here that treatment with a brain-penetrating histone deacetylase 6 (HDAC6) inhibitor for two weeks was sufficient to fully reverse cisplatin-induced cognitive impairments in male mice, as demonstrated in the Y-maze test of spontaneous alternation, the novel object/place recognition test, and the puzzle box test. Normalization of cognitive impairment was associated with reversal of cisplatin-induced synaptosomal mitochondrial deficits and restoration of synaptic integrity. Mechanistically, cisplatin induced deacetylation of the microtubule protein α-tubulin and hyperphosphorylation of the microtubule-associated protein tau. These cisplatin-induced changes were reversed by HDAC6 inhibition. Our data suggest that inhibition of HDAC6 restores microtubule stability and reverses tau phosphorylation, leading to normalization of synaptosomal mitochondrial function and synaptic integrity and thereby to reversal of CICI. Remarkably, our results indicate that short-term daily treatment with the HDAC6 inhibitor was sufficient to achieve prolonged reversal of established behavioral, structural and functional deficits induced by cisplatin. Because the beneficial effects of HDAC6 inhibitors as add-ons to cancer treatment have been demonstrated in clinical trials, selective targeting of HDAC6 with brain-penetrating inhibitors appears a promising therapeutic approach for reversing chemotherapy-induced neurotoxicity while enhancing tumor control.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cognitive Dysfunction , Enzyme Inhibitors/therapeutic use , Histone Deacetylase 6/metabolism , Tauopathies/enzymology , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/enzymology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Green Fluorescent Proteins/metabolism , Histone Deacetylase 6/ultrastructure , Hydroxamic Acids/blood , Hydroxamic Acids/therapeutic use , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Pyrimidines/blood , Pyrimidines/therapeutic use , Recombinant Fusion Proteins/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/pathology , Synaptosomes/ultrastructure , Tauopathies/chemically induced , Tauopathies/drug therapy , Time Factors , Tubulin/metabolism , tau Proteins/metabolism
4.
Cancer Chemother Pharmacol ; 82(3): 565-570, 2018 09.
Article in English | MEDLINE | ID: mdl-29951694

ABSTRACT

PURPOSE: Belinostat is a second-generation histone deacetylase inhibitor (HDI) predominantly metabolized by UGT1A1-mediated glucuronidation. Two common polymorphisms (UGT1A1*28 and UGT1A1*60) were previously associated with impaired drug clearance and thrombocytopenia risk, likely from increased drug exposure. This latter phenomenon has been observed with other HDIs such as abexinostat, panobinostat, romidepsin, and vorinostat. It was the intention of this brief report to expand a population pharmacokinetic (PPK) model to include a pharmacodynamic (PD) model describing the change in platelet levels in patients with cancer administered belinostat as a 48-h continuous intravenous infusion, along with cisplatin and etoposide. METHODS: The PPK/PD model developed here introduced an additional rate constant to a commonly used mechanistic myelosuppression model to better describe the maturation of megakaryocytes into platelets before degradation and a feedback mechanism. The model employed a proportional error model to describe the observed circulating platelet data. RESULTS: Several covariates were explored, including sex, body weight, UGT1A1 genotype status, liver, and kidney function, but none significantly improved the model. Platelet levels rebounded to baseline within 21 days, before the next cycle of therapy. Simulations predicted that higher belinostat drug exposure does cause lower thrombocyte nadirs compared to lower belinostat levels. However, platelet levels rebound by the start of the next belinostat cycle. CONCLUSIONS: This model suggests a q3week schedule allows for sufficient platelet recovery before the next belinostat infusion is optimal.


Subject(s)
Blood Platelets/drug effects , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Models, Biological , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Blood Platelets/metabolism , Blood Platelets/pathology , Clinical Trials, Phase I as Topic , Computer Simulation , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/blood , Infusions, Intravenous , Male , Sulfonamides/administration & dosage , Sulfonamides/blood
5.
J Med Chem ; 61(8): 3697-3711, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29620892

ABSTRACT

Autotaxin (ATX) catalyzes the hydrolysis of lysophosphatidylcholine (LPC) generating the lipid mediator lysophosphatidic acid (LPA). Both ATX and LPA are involved in various pathological inflammatory conditions, including fibrosis and cancer, and have attracted great interest as medicinal targets over the past decade. Thus, the development of novel potent ATX inhibitors is of great importance. We have developed a novel class of ATX inhibitors containing the zinc binding functionality of hydroxamic acid. Such novel hydroxamic acids that incorporate a non-natural δ-amino acid residue exhibit high in vitro inhibitory potency over ATX (IC50 values 50-60 nM). Inhibitor 32, based on δ-norleucine, was tested for its efficacy in a mouse model of pulmonary inflammation and fibrosis induced by bleomycin and exhibited promising efficacy. The novel hydroxamic ATX inhibitors provide excellent tools for the study of the role of the enzyme and could contribute to the development of novel therapeutic agents for the treatment of fibrosis and other chronic inflammatory diseases.


Subject(s)
Hydroxamic Acids/therapeutic use , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/metabolism , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin , Catalytic Domain , Drug Design , Hydroxamic Acids/blood , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Mice , Molecular Docking Simulation , Molecular Structure , Phosphodiesterase Inhibitors/blood , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Pulmonary Fibrosis/chemically induced , Structure-Activity Relationship
6.
J Clin Oncol ; 35(11): 1231-1239, 2017 Apr 10.
Article in English | MEDLINE | ID: mdl-28221861

ABSTRACT

Purpose This phase I trial evaluated epigenetic modulation of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor by using a histone deacetylase abexinostat in combination with pazopanib to enhance response and reverse resistance. Patients and Methods Pazopanib was administered once a day on days 1 to 28 and abexinostat was administered orally twice a day on days 1 to 5, 8 to 12, and 15 to 19 (schedule A) or on days 1 to 4, 8 to 11, and 15 to 18 (schedule B). Dose escalation (3 + 3 design) in all solid tumors was followed by dose expansion in renal cell carcinoma (RCC). Results Fifty-one patients with RCC (N = 22) were enrolled, including 30 (59%) with one or more lines of prior VEGF-targeting therapy. Five dose-limiting toxicities, including fatigue (n = 2), thrombocytopenia (n = 2), and elevated AST/ALT (n = 1), were observed with schedule A; one dose-limiting toxicity was observed (elevated AST/ALT) was observed with schedule B. Grade ≥ 3 related adverse events included fatigue (16%), thrombocytopenia (16%), and neutropenia (10%). The recommended phase II dose was established as abexinostat 45 mg/m2 twice a day administered per schedule B plus pazopanib 800 mg/d. Objective response rate was 21% overall and 27% in the RCC subset. Median duration of response was 9.1 months (1.2 to > 49 months). Eight patients (16%) had durable control of disease for > 12 months. Durable tumor regressions were observed in seven (70%) of 10 patients with pazopanib-refractory disease, including one patients with RCC with ongoing response > 3.5 years. Peripheral blood histone acetylation and HDAC2 gene expression were associated with durable response to treatment. Conclusion Abexinostat is well tolerated in combination with pazopanib, allowing prolonged exposure and promising durable responses in pazopanib- and other VEGF inhibitor-refractory tumors, which supports epigenetically mediated reversal of treatment resistance.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Renal Cell/drug therapy , Drug Resistance, Neoplasm/drug effects , Kidney Neoplasms/drug therapy , Acetylation , Adult , Aged , Alanine Transaminase/blood , Angiogenesis Inhibitors/administration & dosage , Aspartate Aminotransferases/blood , Benzofurans/administration & dosage , Benzofurans/blood , Benzofurans/pharmacokinetics , Carcinoma, Renal Cell/genetics , Disease Progression , Disease-Free Survival , Drug Resistance , Epigenesis, Genetic , Fatigue/chemically induced , Female , Gene Expression , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/pharmacokinetics , Histones/metabolism , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Indazoles , Kidney Neoplasms/genetics , Male , Maximum Tolerated Dose , Middle Aged , Neutropenia/chemically induced , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Thrombocytopenia/chemically induced , Treatment Outcome , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics , Young Adult
7.
Biomed Chromatogr ; 31(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27511598

ABSTRACT

Histone deacetylase inhibitors (HDAC inhibitors) are used to treat malignancies such as cutaneous T cell lymphoma and peripheral T cell lymphoma. Only four drugs are approved by the US Food and Drug Administration, namely vorinostat, romidepsin, panobinostat and belinostat, while chidamide has been approved in China. There are a number of bioanalytical methods reported for the measurement of HDAC inhibitors in clinical (human plasma and serum) and preclinical (mouse plasma, rat plasma, urine and tissue homogenates, etc.) studies. This review covers various HDAC inhibitors such as vorinostat, romidepsin, panobinostat, belinostat and chidamide. In addition to providing a comprehensive review of the available methods for the above mentioned HDAC inhibitors, it also provides case studies with perspectives for chosen drugs. Based on the review, it is concluded that the published methodologies using either HPLC or LC-MS/MS are well suited for the quantification of HDAC inhibitors in various biological fluids to delineate pharmacokinetic data.


Subject(s)
Chromatography, High Pressure Liquid/methods , Histone Deacetylase Inhibitors/pharmacokinetics , Tandem Mass Spectrometry/methods , Aminopyridines/blood , Aminopyridines/pharmacokinetics , Aminopyridines/urine , Animals , Benzamides/blood , Benzamides/pharmacokinetics , Benzamides/urine , Depsipeptides/blood , Depsipeptides/pharmacokinetics , Depsipeptides/urine , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/urine , Humans , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/urine , Indoles/blood , Indoles/pharmacokinetics , Indoles/urine , Neoplasms/drug therapy , Panobinostat , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Sulfonamides/urine , Vorinostat
8.
PLoS One ; 11(3): e0151260, 2016.
Article in English | MEDLINE | ID: mdl-26974544

ABSTRACT

Invasive aspergillosis (IA) is a life-threatening systemic mycosis caused primarily by Aspergillus fumigatus. Early diagnosis of IA is based, in part, on an immunoassay for circulating fungal cell wall carbohydrate, galactomannan (GM). However, a wide range of sensitivity and specificity rates have been reported for the GM test across various patient populations. To obtain iron in vivo, A. fumigatus secretes the siderophore, N,N',N"-triacetylfusarinine C (TAFC) and we hypothesize that TAFC may represent a possible biomarker for early detection of IA. We developed an ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for TAFC analysis from serum, and measured TAFC in serum samples collected from patients at risk for IA. The method showed lower and upper limits of quantitation (LOQ) of 5 ng/ml and 750 ng/ml, respectively, and complete TAFC recovery from spiked serum. As proof of concept, we evaluated 76 serum samples from 58 patients with suspected IA that were investigated for the presence of GM. Fourteen serum samples obtained from 11 patients diagnosed with probable or proven IA were also analyzed for the presence of TAFC. Control sera (n = 16) were analyzed to establish a TAFC cut-off value (≥6 ng/ml). Of the 36 GM-positive samples (≥0.5 GM index) from suspected IA patients, TAFC was considered positive in 25 (69%). TAFC was also found in 28 additional GM-negative samples. TAFC was detected in 4 of the 14 samples (28%) from patients with proven/probable aspergillosis. Log-transformed TAFC and GM values from patients with proven/probable IA, healthy individuals and SLE patients showed a significant correlation with a Pearson r value of 0.77. In summary, we have developed a method for the detection of TAFC in serum that revealed this fungal product in the sera of patients at risk for invasive aspergillosis. A prospective study is warranted to determine whether this method provides improved early detection of IA.


Subject(s)
Aspergillosis/blood , Aspergillus fumigatus , Ferric Compounds/blood , Hydroxamic Acids/blood , Siderophores/blood , Adult , Biomarkers/blood , Female , Humans , Male , Sensitivity and Specificity
9.
Sci Transl Med ; 8(326): 326ra23, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888431

ABSTRACT

Histone deacetylase inhibitors (HDACi) are approved for treating rare cancers and are of interest as potential therapies for neurodegenerative disorders. We evaluated a triple combination formulation (TCF) comprising the pan-HDACi vorinostat, the caging agent 2-hydroxypropyl-ß-cyclodextrin (HPBCD), and polyethylene glycol (PEG) for treating a mouse model (the Npc1(nmf164) mouse) of Niemann-Pick type C (NPC) disease, a difficult-to-treat cerebellar disorder. Vorinostat alone showed activity in cultured primary cells derived from Npc1(nmf164) mice but did not improve animal survival. However, low-dose, once-weekly intraperitoneal injections of the TCF containing vorinostat increased histone acetylation in the mouse brain, preserved neurites and Purkinje cells, delayed symptoms of neurodegeneration, and extended mouse life span from 4 to almost 9 months. We demonstrate that the TCF boosted the ability of HDACi to cross the blood-brain barrier and was not toxic even when used long term. Further, the TCF enabled dose reduction, which has been a major challenge in HDACi therapy. TCF simultaneously treats neurodegenerative and systemic symptoms of Niemann-Pick type C disease in a mouse model.


Subject(s)
Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/therapeutic use , Niemann-Pick Disease, Type C/drug therapy , 2-Hydroxypropyl-beta-cyclodextrin , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Disease Models, Animal , Disease Progression , Fibroblasts/drug effects , Fibroblasts/metabolism , Histone Deacetylase Inhibitors/blood , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/blood , Hydroxamic Acids/therapeutic use , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Liver/drug effects , Liver/pathology , Mice , Mutation/genetics , Nerve Degeneration/blood , Nerve Degeneration/complications , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Niemann-Pick Disease, Type C/blood , Niemann-Pick Disease, Type C/complications , Niemann-Pick Disease, Type C/pathology , Polyethylene Glycols/chemistry , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Purkinje Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/drug effects , Spleen/pathology , Survival Analysis , Vorinostat , beta-Cyclodextrins/chemistry
10.
Invest New Drugs ; 34(2): 193-201, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26769244

ABSTRACT

BACKGROUND: Belinostat, a potent pan-inhibitor of histone deacetylase (HDAC) enzymes, is approved in the United States (US) for relapsed/refractory peripheral T-cell lymphoma. In nonclinical studies, bile and feces were identified as the predominant elimination routes (50-70%), with renal excretion accounting for ~30-50%. A Phase 1 human mass balance study was conducted to identify species-dependent variations in belinostat metabolism and elimination. METHODS: Patients received a single 30-min intravenous (i.v.) infusion of (14)C-labeled belinostat (1500 mg). Venous blood samples and pooled urine and fecal samples were evaluated using liquid chromatography-tandem mass spectroscopy for belinostat and metabolite concentrations pre-infusion through 7 days post-infusion. Total radioactivity was determined using liquid scintillation counting. Continued treatment with nonradiolabled belinostat (1000 mg/m(2) on Days 1-5 every 21 days) was permitted. RESULTS: Belinostat was extensively metabolized and mostly cleared from plasma within 8 h (N = 6), indicating that metabolism is the primary route of elimination. Systemic exposure for the 5 major metabolites was >20% of parent, with belinostat glucuronide the predominant metabolite. Mean recovery of radioactive belinostat was 94.5% ± 4.0%, with the majority excreted within 48 and 96 h in urine and feces, respectively. Renal elimination was the principal excretion route (mean 84.8% ± 9.8% of total dose); fecal excretion accounted for 9.7% ± 6.5%. Belinostat was well tolerated, with mostly mild to moderate adverse events and no treatment-related severe/serious events. CONCLUSION: Mass balance was achieved (~95% mean recovery), with metabolism identified as the primary route of elimination. Radioactivity was predominantly excreted renally as belinostat metabolites.


Subject(s)
Carbon Radioisotopes/metabolism , Carbon Radioisotopes/pharmacokinetics , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacokinetics , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics , Aged , Carbon Radioisotopes/blood , Carbon Radioisotopes/therapeutic use , Female , Humans , Hydroxamic Acids/blood , Hydroxamic Acids/therapeutic use , Male , Metabolic Networks and Pathways , Metabolomics , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/metabolism , Neoplasms/blood , Neoplasms/metabolism , Radioactivity , Sulfonamides/blood , Sulfonamides/therapeutic use , Treatment Outcome
11.
Biomed Chromatogr ; 30(7): 1138-1144, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26633099

ABSTRACT

A highly sensitive, specific and rapid LC-ESI-MS/MS method has been developed and validated for the quantification of rocilinostat in small volume mouse plasma (20 µL) using vorinostat as an internal standard (IS) as per regulatory guidelines. Sample preparation was accomplished through a protein precipitation procedure with acetonitrile. Chromatography was achieved on Prodigy ODS-2 column using a binary gradient using mobile phase A (0.2% formic acid in water) and B (acetonitrile) at a flow rate of 0.38 mL/min. The total chromatographic run time was 4.1 min and the elution of rocilinostat and IS occurred at ~3.2 and 2.9 min, respectively. A linear response function was established in the concentration range of 0.28-1193 ng/mL in mouse plasma. The intra- and inter-day accuracy and precisions were in the ranges of 3.12-8.93 and 6.41-11.6%, respectively. This novel method has been applied to a pharmacokinetic study in mice. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Chromatography, Liquid/methods , Hydroxamic Acids/blood , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Animals , Limit of Detection , Male , Mice , Mice, Inbred BALB C
12.
Eur J Clin Pharmacol ; 72(2): 153-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26494130

ABSTRACT

PURPOSE: Panobinostat, a potent pan-deacetylase inhibitor, improved progression-free survival (PFS) in patients with relapsed and refractory multiple myeloma when combined with bortezomib and dexamethasone in a phase 3 trial, PANORAMA-1. This study aims to explore exposure-response relationship for panobinostat in this combination in a phase 1 trial, B2207 and contrast with data from historical single-agent studies. METHODS: Panobinostat plasma concentration-time profiles were obtained in patients from PANORAMA-1 (n = 12) and B2207 (n = 12) trials. Overall response rates (ORR) and major adverse events (AE) by panobinostat exposure were investigated in the B2207 trial. Panobinostat PK data from combination trials were contrasted with data from single-agent studies. RESULTS: At maximum tolerated dose (MTD), the geometric mean of panobinostat area under curve from 0 to 24 h (AUC0-24) was 47.5 ng h/mL (77 % CV), and maximum plasma concentration (Cmax) was 8.1 ng/mL (90 % CV). These values were comparable with exposure data obtained in PANORAMA-1, but were 20 % lower than those without dexamethasone, and ∼ 50 % lower from single-agent trials, likely due to enzyme induction by dexamethasone. Higher levels of panobinostat exposure were associated with higher response rates and higher incidences of diarrhea and thrombocytopenia. CONCLUSIONS: Apparent panobinostat exposure-AE and exposure-ORR relationships were observed when combined with bortezomib and dexamethasone in the treatment of patients with relapsed and refractory multiple myeloma. The addition of dexamethasone facilitated best response even though plasma exposure of panobinostat was reduced. Combination with a strong enzyme inducer should be avoided in future trials to prevent further reduction of panobinostat exposure.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bortezomib/pharmacokinetics , Dexamethasone/pharmacology , Hydroxamic Acids/pharmacokinetics , Indoles/pharmacokinetics , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bortezomib/adverse effects , Bortezomib/blood , Bortezomib/therapeutic use , Dexamethasone/adverse effects , Dexamethasone/therapeutic use , Double-Blind Method , Drug Resistance, Neoplasm , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/blood , Hydroxamic Acids/therapeutic use , Indoles/adverse effects , Indoles/blood , Indoles/therapeutic use , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neoplasm Recurrence, Local , Panobinostat
13.
Antimicrob Agents Chemother ; 60(1): 180-9, 2016 01.
Article in English | MEDLINE | ID: mdl-26482300

ABSTRACT

GSK1322322 is a novel inhibitor of peptide deformylase (PDF) with good in vitro activity against bacteria associated with community-acquired pneumonia and skin infections. We have characterized the in vivo pharmacodynamics (PD) of GSK1322322 in immunocompetent animal models of infection with Streptococcus pneumoniae and Haemophilus influenzae (mouse lung model) and with Staphylococcus aureus (rat abscess model) and determined the pharmacokinetic (PK)/PD index that best correlates with efficacy and its magnitude. Oral PK studies with both models showed slightly higher-than-dose-proportional exposure, with 3-fold increases in area under the concentration-time curve (AUC) with doubling doses. GSK1322322 exhibited dose-dependent in vivo efficacy against multiple isolates of S. pneumoniae, H. influenzae, and S. aureus. Dose fractionation studies with two S. pneumoniae and S. aureus isolates showed that therapeutic outcome correlated best with the free AUC/MIC (fAUC/MIC) index in S. pneumoniae (R(2), 0.83), whereas fAUC/MIC and free maximum drug concentration (fCmax)/MIC were the best efficacy predictors for S. aureus (R(2), 0.9 and 0.91, respectively). Median daily fAUC/MIC values required for stasis and for a 1-log10 reduction in bacterial burden were 8.1 and 14.4 for 11 S. pneumoniae isolates (R(2), 0.62) and 7.2 and 13.0 for five H. influenzae isolates (R(2), 0.93). The data showed that for eight S. aureus isolates, fAUC correlated better with efficacy than fAUC/MIC (R(2), 0.91 and 0.76, respectively), as efficacious AUCs were similar for all isolates, independent of their GSK1322322 MIC (range, 0.5 to 4 µg/ml). Median fAUCs of 2.1 and 6.3 µg · h/ml were associated with stasis and 1-log10 reductions, respectively, for S. aureus.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Haemophilus Infections/drug therapy , Hydroxamic Acids/pharmacokinetics , Pneumonia, Pneumococcal/drug therapy , Staphylococcal Infections/drug therapy , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacology , Area Under Curve , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/blood , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacology , Haemophilus Infections/blood , Haemophilus Infections/microbiology , Haemophilus influenzae/drug effects , Haemophilus influenzae/enzymology , Haemophilus influenzae/growth & development , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacology , Lung/drug effects , Lung/microbiology , Male , Mice , Microbial Sensitivity Tests , Pneumonia, Pneumococcal/blood , Pneumonia, Pneumococcal/microbiology , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Staphylococcus aureus/growth & development , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/growth & development
14.
Pharmacology ; 95(3-4): 160-5, 2015.
Article in English | MEDLINE | ID: mdl-25896603

ABSTRACT

BACKGROUND: Because lapatinib and vorinostat [suberoylanilide hydroxamic acid (SAHA)] are both new anticancer drugs, interactions between SAHA and lapatinib remain unclear. This study examines pharmacokinetic interactions in simultaneous oral administration of SAHA and lapatinib to rats. METHODS: Twenty-four rats were divided randomly into 3 groups: a lapatinib group (lapatinib 25 mg/kg, n = 8), a SAHA group (SAHA 25 mg/kg, n = 8), and a coadministration group (SAHA 25 mg/kg and lapatinib 25 mg/kg, n = 8). Using ultrahigh-performance liquid chromatography-tandem mass spectrometry, the concentrations of lapatinib and SAHA were determined in the plasma of the test rats. RESULTS: Statistically significant pharmacokinetic differences appeared for lapatinib levels between the lapatinib and the coadministration group. When lapatinib was coadministered with SAHA, the AUC0-t decreased from 39,816.6 to 23,712.8 ng/ml ∙ h (p < 0.05), while the mean residence time (MRT)0-t increased from 7.0 to 10.3 h (p < 0.05) and t1/2 increased from 3.5 to 6.4 h (p < 0.05). Between the SAHA levels for the SAHA group and those for the coadministration group, there appeared to be no statistically significant differences. CONCLUSION: The resulting data indicate that, when administered together, lapatinib does not influence the pharmacokinetic profile of SAHA in rats, while, in contrast, SAHA influences the pharmacokinetic profile of lapatinib.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Quinazolines/pharmacokinetics , Animals , Antineoplastic Agents/blood , Drug Interactions , Hydroxamic Acids/blood , Lapatinib , Male , Quinazolines/blood , Rats, Sprague-Dawley , Vorinostat
15.
Biomed Chromatogr ; 29(8): 1249-58, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25616154

ABSTRACT

IKH12 is a novel histone deacetylase 6 selective inhibitor. A rapid and sensitive liquid chromatography tandem mass spectrometry method was developed and validated for the quantification of IKH12 in rat plasma and tissue with kendine 91 as internal standard (IS). The samples were prepared by liquid-liquid extraction with tert-butyl methyl ether. The chromatographic separation was accomplished by using a Zorbax Extend C18 4.6 × 150 mm, 5 µm column, with a mobile phase consisting of methanol and 0.1% formic acid (75:25 v/v). Multiple reaction monitoring, using electrospray ionization in positive ion mode, was employed to quantitatively detect IKH12 and IS. The monitored transitions were set at m/z 418 → 252 and 444 → 169 for IKH12 and kendine 91, respectively. The calibration curve was linear over the concentration range 2-1000 ng mL(-1) . The intra- and inter-assay precision and accuracy of the quality controls and the limit of quantification were satisfactory in all cases (according to European Medicines Agency guidelines). Stability studies showed that plasma samples were stable in the chromatography rack for 24 h and at -80°C for 2 months and also after three freeze-thaw cycles. This method was successfully applied to a pharmacokinetic study of IKH12 in rat.


Subject(s)
Antineoplastic Agents/blood , Histone Deacetylase Inhibitors/blood , Hydroxamic Acids/blood , Proline/analogs & derivatives , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Chromatography, High Pressure Liquid/methods , Limit of Detection , Liquid-Liquid Extraction/methods , Male , Methyl Ethers/chemistry , Proline/blood , Rats, Wistar , Tandem Mass Spectrometry/methods
16.
Cancer Chemother Pharmacol ; 75(1): 87-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25377157

ABSTRACT

PURPOSE: This study assessed the pharmacokinetics and safety of oral panobinostat and its metabolite BJB432 in patients with advanced solid tumors and normal to severely impaired renal function. METHODS: Patients with varying degrees of renal impairment, defined by their 24-h baseline urine creatinine clearance (as normal, mild, moderate or severe), received a single oral dose of 30 mg panobinostat. Serial plasma samples were collected pre-dose and up to 96-h post-dose. Serial urine samples were collected for 24-h post-dose. Following the serial PK sampling, patients received 30 mg oral panobinostat thrice weekly for as long as the patient had benefit. Pharmacokinetic parameters were derived using non-compartmental analysis. RESULTS: Thirty-seven patients were enrolled, and median age was 64 (range 40-81) years. Eleven patients had normal renal function; 10, 10, and 6 patients had mild, moderate, and severe renal impairment, respectively. Geometric means of AUC(0-∞) in the normal, mild, moderate, and severe groups were 224.5, 144.3, 223.1, and 131.7 ng h/mL, respectively. Geometric mean ratio of BJB432 to parent drug plasma AUC(0-∞) was 0.64 in the normal group and increased to 0.81, 1.13, and 1.20 in the mild, moderate, and severe groups, respectively. The fraction excreted as unchanged panobinostat was small (<2 %), with a large variability. The renal clearance of panobinostat and tolerability was similar across all four groups. CONCLUSION: Systemic exposure to panobinostat did not increase with severity of renal impairment, and the drug was tolerated equally; thus, patients with renal impairment do not require starting dose adjustments.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Drugs, Investigational/pharmacokinetics , Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Indoles/pharmacokinetics , Neoplasms/drug therapy , Renal Insufficiency/complications , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Biotransformation , Drug Monitoring , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Drugs, Investigational/therapeutic use , Half-Life , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/blood , Hydroxamic Acids/metabolism , Hydroxamic Acids/therapeutic use , Indoles/adverse effects , Indoles/blood , Indoles/metabolism , Indoles/therapeutic use , Kidney/drug effects , Kidney/physiopathology , Metabolic Clearance Rate , Middle Aged , Neoplasm Grading , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/physiopathology , Panobinostat , Patient Dropouts , Renal Insufficiency/physiopathology , Severity of Illness Index
17.
Article in English | MEDLINE | ID: mdl-24636840

ABSTRACT

Vorinostat (suberoylanilide hydroxamic acid) is the first approved histone deacetylase (HDAC) inhibitor for the treatment of cutaneous T-cell lymphoma after progressive disease following two systemic therapies. Intracellular access of vorinostat is essential to exert its epigenetic effects. Therefore, we studied the relationship between vorinostat extracellular (plasma) and intracellular (peripheral blood mononuclear cells, PBMCs) concentration and assessed its concentration-effect relationship by HDAC activity testing. Assays were developed and validated for the low nanomolar quantification of vorinostat and two inactive metabolites in human plasma and PBMCs. For the vorinostat extraction from plasma and PBMCs solid-phase extraction and liquid-liquid extraction methods were applied. Extraction recoveries ranged from 88.6% to 114.4% for all analytes and extraction methods. Extracts were chromatographed on a Phenomenex Luna column isocratically (plasma) or by gradient (PBMCs) consisting of acidic ammonium acetate, acetonitrile, and methanol. The analytes were quantified using deuterated internal standards and positive electrospray tandem mass spectrometry (multiple reaction monitoring) with lower limits of quantification of 11.0 ng/mL (plasma) and 0.1 ng/3 × 10(6) cells (PBMCs). The calibrated ranges were linear for vorinostat in plasma 11.0-1100 (11,000) ng/mL (metabolites) and PBMCs 0.1-10.0 ng/3 × 10(6) cells with correlation coefficients >0.99, an overall accuracy varying between -6.7% and +3.8% in plasma, -8.1% and -1.5% in PBMCs, and an overall precision ranging from 3.2% to 6.1% in plasma and 0.8% to 4.0% in PBMCs (SD batch-to-batch). The application to blood samples from healthy volunteers incubated with vorinostat revealed accumulation of vorinostat in PBMCs, effective intracellular HDAC inhibition at therapeutic vorinostat concentrations and a direct vorinostat concentration dependency to HDAC inhibition.


Subject(s)
Histone Deacetylase Inhibitors/analysis , Histone Deacetylase Inhibitors/blood , Hydroxamic Acids/analysis , Hydroxamic Acids/blood , Leukocytes, Mononuclear/chemistry , Chromatography, Liquid/methods , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/metabolism , Limit of Detection , Liquid-Liquid Extraction/methods , Solid Phase Extraction/methods , Tandem Mass Spectrometry/methods , Vorinostat
18.
J Vet Pharmacol Ther ; 37(2): 196-200, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24236915

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA), or vorinostat, is a histone deacetylase inhibitor approved for use as chemotherapy for lymphoma in humans. The goal of this study was to establish pharmacological parameters of SAHA in cats. Our interest in treating cats with SAHA is twofold: as an anticancer chemotherapeutic and as antilatency therapy for feline retroviral infections. Relying solely on data from studies in other animals would be inappropriate as SAHA is partially metabolized by glucuronidation, which is absent in feline metabolism. SAHA was administered to cats intravenously (2 mg/kg) or orally (250 mg/m², ~17 mg/kg) in a cross-over study design. Clinically, SAHA was well tolerated at these dosages as no abnormalities were noted following administration. The pharmacokinetics of SAHA in cats was found to be similar to that of dogs, but the overall serum drug exposure was much less than that of humans at an equivalent dose. The pharmacodynamic effect of an increase in acetylated histone proteins in blood was detected after both routes of administration. An increased oral dose of 60 mg SAHA/kg administered to one animal resulted in a surprisingly modest increase in peak drug concentration, suggesting possible saturation of absorption kinetics. This study provides a foundation for future studies of the clinical efficacy of SAHA in treating feline disease.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Animals , Antineoplastic Agents/blood , Area Under Curve , Cats , Cross-Over Studies , Female , Half-Life , Hydroxamic Acids/blood , Male , Vorinostat
19.
Invest New Drugs ; 31(6): 1539-46, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24114121

ABSTRACT

BACKGROUND: A phase I study to assess the maximum-tolerated dose (MTD), dose-limiting toxicity (DLT), pharmacokinetics (PK) and antitumor activity of vorinostat in combination with bortezomib in patients with advanced solid tumors. METHODS: Patients received vorinostat orally once daily on days 1-14 and bortezomib intravenously on days 1, 4, 8 and 11 of a 21-day cycle. Starting dose (level 1) was vorinostat (400 mg) and bortezomib (0.7 mg/m(2)). Bortezomib dosing was increased using a standard phase I dose-escalation schema. PKs were evaluated during cycle 1. RESULTS: Twenty-three patients received 57 cycles of treatment on four dose levels ranging from bortezomib 0.7 mg/m(2) to 1.5 mg/m(2). The MTD was established at vorinostat 400 mg daily and bortezomib 1.3 mg/m(2). DLTs consisted of grade 3 fatigue in three patients (1 mg/m(2),1.3 mg/m(2) and 1.5 mg/m(2)) and grade 3 hyponatremia in one patient (1.5 mg/m(2)). The most common grade 1/2 toxicities included nausea (60.9%), fatigue (34.8%), diaphoresis (34.8%), anorexia (30.4%) and constipation (26.1%). Objective partial responses were observed in one patient with NSCLC and in one patient with treatment-refractory soft tissue sarcoma. Bortezomib did not affect the PKs of vorinostat; however, the Cmax and AUC of the acid metabolite were significantly increased on day 2 compared with day 1. CONCLUSIONS: This combination was generally well-tolerated at doses that achieved clinical benefit. The MTD was established at vorinostat 400 mg daily × 14 days and bortezomib 1.3 mg/m(2) on days 1, 4, 8 and 11 of a 21-day cycle.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Boronic Acids/administration & dosage , Bortezomib , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/blood , Pyrazines/administration & dosage , Vorinostat , Young Adult
20.
Antimicrob Agents Chemother ; 57(5): 2005-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23403431

ABSTRACT

GSK1322322 is a potent inhibitor of peptide deformylase, an essential bacterial enzyme required for protein maturation. GSK1322322 is active against community-acquired skin and respiratory tract pathogens, including methicillin-resistant Staphylococcus aureus, multidrug-resistant Streptococcus pneumoniae, and atypical pathogens. This phase I, randomized, double-blind, placebo-controlled, 2-part, single-dose, dose escalation study (first time in humans) evaluated the safety, tolerability, and pharmacokinetics of GSK1322322 (powder-in-bottle formulation) in healthy volunteers. In part A, dose escalation included GSK1322322 doses of 100, 200, 400, 800, and 1,500 mg under fasting conditions and 800 mg administered with a high-fat meal. In part B, higher doses of GSK1322322 (2,000, 3,000, and 4,000 mg) were evaluated under fasting conditions. Of the 39 volunteers enrolled in the study, 29 and 10 volunteers were treated with GSK1322322 and placebo, respectively. Upon single-dose administration, GSK1322322 was absorbed rapidly, with median times to maximum plasma concentration (T(max)) ranging from 0.5 to 1.0 h. The maximum observed plasma concentration (C(max)) and exposure (area under the concentration-time curve [AUC]) of GSK1322322 were greater than dose proportional between 100 and 1,500 mg and less than dose proportional between 1,500 and 4,000 mg. Administration of the drug with a high-fat meal reduced the rate of absorption (reduced C(max) and delayed T(max)) without affecting the extent of absorption (no effect on AUC). GSK1322322 was generally well tolerated, with all adverse events being mild to moderate in intensity during both parts of the study. The most frequently reported adverse event was headache. Data from this study support further evaluation of GSK1322322.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Adolescent , Adult , Aged , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/blood , Area Under Curve , Biological Availability , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/blood , Dietary Fats/administration & dosage , Double-Blind Method , Drug Administration Schedule , Fasting/blood , Headache/etiology , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/blood , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...