Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Chemother Pharmacol ; 72(4): 747-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24013574

ABSTRACT

PURPOSE: Panobinostat is a potent oral pan-deacetylase inhibitor with promising clinical activity in hematologic malignancies. Panobinostat was shown to inhibit CYP2D6 activity in vitro; thus understanding the magnitude of the potential clinical inhibition of panobinostat on co-medications that are CYP2D6 substrates becomes important. METHODS: This study evaluated the effects of co-administration of panobinostat with a sensitive CYP2D6 substrate, dextromethorphan (DM), in patients with advanced cancer who have functional CYP2D6 genes. Patients received 60 mg DM alone on day 1, panobinostat at 20 mg alone on days 3 and 5, and both agents on day 8. Plasma concentrations of DM and its metabolite dextrorphan (DX) were determined by liquid chromatography-tandem mass spectrometry following serial blood collections on day 1 (DM alone) and day 8 (in combination with panobinostat). RESULTS: Panobinostat increased DM exposure by 64 % [geometric mean ratio (GMR), 1.64 (90 % confidence interval (CI), 1.17-2.31)] and DX exposure by 29 % (GMR, 1.29 [90 % CI, 1.10-1.51]). These results indicated that panobinostat weakly inhibited a sensitive CYP2D6 substrate in cancer patients by increasing DM exposure by less than twofold. CONCLUSION: Safety monitoring of sensitive CYP2D6 substrates with narrow therapeutic index is recommended when co-administering with panobinostat in future clinical practice.


Subject(s)
Antineoplastic Agents/pharmacology , Cytochrome P-450 CYP2D6 Inhibitors , Dextromethorphan/pharmacokinetics , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Neoplasms/pathology , Aged , Chromatography, Liquid , Cytochrome P-450 CYP2D6/metabolism , Dextrorphan/pharmacokinetics , Drug Interactions , Female , Humans , Male , Middle Aged , Panobinostat , Tandem Mass Spectrometry
2.
Cancer Chemother Pharmacol ; 72(3): 537-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23820963

ABSTRACT

PURPOSE: Panobinostat, a pan-deacetylase inhibitor, increases acetylation of proteins associated with growth and survival of malignant cells. This phase 2 study evaluated the efficacy of intravenous (IV) panobinostat in patients with castration-resistant prostate cancer (CRPC) who had previously received chemotherapy. The primary end point was 24-week progression-free survival. Secondary end points included safety, tolerability, and the proportion of patients with a prostate-specific antigen (PSA) decline. METHODS: IV panobinostat (20 mg/m(2)) was administered to patients on days 1 and 8 of a 21-day cycle. Tumor response was assessed by imaging every 12 weeks (4 cycles) according to modified response evaluation criteria in solid tumors (Scher et al. in Clin Cancer Res 11:5223-5232, 23), and PSA response was defined as a 50 % decrease from baseline maintained for ≥4 weeks. Safety monitoring was routinely performed and included electrocardiogram monitoring. RESULTS: Of 35 enrolled patients, four (11.4 %) were alive without progression of disease at 24 weeks. PSA was evaluated in 34 (97.1 %) patients: five (14.3 %) patients demonstrated a decrease in PSA but none ≥50 %; one patient (2.9 %) had carcinoembryonic antigen as a marker of his prostate cancer, which declined by 43 %. Toxicities regardless of relationship to panobinostat included fatigue (62.9 %), thrombocytopenia (45.7 %), nausea (51.4 %), and decreased appetite (37.1 %). CONCLUSIONS: Despite promising preclinical data and scientific rationale, treatment with IV panobinostat did not show a sufficient level of clinical activity to pursue further investigation as a single agent in CRPC.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Disease-Free Survival , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Indoles/administration & dosage , Indoles/adverse effects , Infusions, Intravenous , Male , Middle Aged , Orchiectomy , Panobinostat , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Treatment Outcome
3.
Cancer Chemother Pharmacol ; 69(2): 555-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22057852

ABSTRACT

PURPOSE: Panobinostat is a novel oral pan-deacetylase inhibitor with promising anti-cancer activity. The study aimed to determine the influence of food on the oral bioavailability of panobinostat. METHODS: This multicenter study consisted of a randomized, three-way crossover, food-effect study period (cycle 1) followed by single-agent panobinostat continual treatment phase in patients with advanced cancer. Patients received panobinostat 20 mg twice weekly, and panobinostat pharmacokinetics was investigated on days 1, 8, and 15 with a randomly assigned sequence of three prandial states (fasting, high-fat, and normal breakfast). RESULTS: Thirty-six patients were assessed for the food effect on pharmacokinetics and safety in cycle 1, after which 29 patients continued treatment, receiving single-agent panobinostat. Safety and antitumor activity were assessed during the extension period. Panobinostat systemic exposure was marginally reduced (14-16%) following food [geometric mean ratio (GMR) of the AUC(0-∞)/high-fat breakfast/fasting, 0.84 (90% confidence interval {CI}, 0.74-0.96); normal breakfast/fasting, 0.86 (90% CI, 0.75-1.00)], and interpatient variability (coefficient of variation, 59%) remained essentially unchanged with or without food. Panobinostat C (max) was reduced by 44% (high-fat) and 36% (normal) with median T (max) prolonged by 1-1.5 h following food. Panobinostat was well tolerated, with thrombocytopenia, fatigue, nausea, and vomiting as common adverse events, and demonstrated antitumor activity with one patient with a partial response and six patients with stable disease as best response. CONCLUSIONS: Food produced minor changes in oral panobinostat exposure; thus, panobinostat can be given without regard to food intake in future clinical studies.


Subject(s)
Food , Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Neoplasms/metabolism , Administration, Oral , Adult , Aged , Aged, 80 and over , Area Under Curve , Biological Availability , Cross-Over Studies , Diet, High-Fat , Drug Administration Schedule , Eating , Fasting , Fatigue/chemically induced , Female , Food-Drug Interactions , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Indoles , Male , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Neoplasms/drug therapy , Neoplasms/pathology , Panobinostat , Treatment Outcome , Vomiting/chemically induced
4.
Cancer Chemother Pharmacol ; 68(3): 805-13, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21706316

ABSTRACT

PURPOSE: Panobinostat is partly metabolized by CYP3A4 in vitro. This study evaluated the effect of a potent CYP3A inhibitor, ketoconazole, on the pharmacokinetics and safety of panobinostat. METHODS: Patients received a single panobinostat oral dose on day 1, followed by 4 days wash-out period. On days 5-9, ketoconazole was administered. On day 8, a single panobinostat dose was co-administered with ketoconazole. Panobinostat was administered as single agent three times a week on day 15 and onward. RESULTS: In the presence of ketoconazole, there was 1.6- and 1.8-fold increase in C (max) and AUC of panobinostat, respectively. No substantial change in T (max) or half-life was observed. No difference in panobinostat-pharmacokinetics between patients carrying CYP3A5*1/*3 and CYP3A5*3/*3 alleles was observed. Most frequently reported adverse events were gastrointestinal related. Patients had asymptomatic hypophosphatemia (64%), and urine analysis suggested renal phosphate wasting. CONCLUSIONS: Co-administration of panobinostat with CYP3A inhibitors is feasible as the observed increase in panobinostat PK parameters was not considered clinically relevant. Considering the variability in exposure following enzyme inhibition and the fact that chronic dosing of panobinostat was not studied with CYP3A inhibitors, close monitoring of panobinostat-related adverse events is necessary.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 CYP3A Inhibitors , Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Ketoconazole/pharmacology , Adult , Aged , Antifungal Agents/blood , Area Under Curve , Cytochrome P-450 CYP3A/genetics , Drug Interactions , Electrocardiography/drug effects , Female , Half-Life , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/therapeutic use , Hypophosphatemia/blood , Hypophosphatemia/metabolism , Indoles , Ketoconazole/blood , Male , Mass Spectrometry , Middle Aged , Neoplasms/drug therapy , Panobinostat , Pharmacogenetics , Sample Size
SELECTION OF CITATIONS
SEARCH DETAIL
...