Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Chemother Pharmacol ; 69(2): 431-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21805353

ABSTRACT

BACKGROUND: The triterpenoid 2-cyano-3,12-dioxoolean-1,9-dien-28-oic Acid (CDDO, previously RTA 401) is a multifunctional molecule that controls cellular growth and differentiation. While CDDO is capable of activating the transcription factor peroxisome proliferator activator receptor-γ (PPARγ), its apoptotic effects in malignant cells have been shown to occur independently of PPARγ. A phase I dose-escalation study was conducted to determine the toxicity, the maximum tolerated dose, and the pharmacokinetics and pharmacodynamics of CDDO, administered as a 5-day continuous infusion every 28 days in patients with advanced cancers. METHODS: An accelerated titration design was followed, with one patient per cohort entered, and doses ranging from 0.6 to 38.4 mg/m(2)/h. Pharmacokinetics of CDDO was assessed and cleaved poly (ADP-ribose) polymerase (c-PARP), as a marker of apoptosis, was measured in peripheral blood mononuclear cells to assess drug effect. RESULTS: Seven patients, one patient per dose level up to dose level 7 (38.4 mg/m(2)/h), were enrolled and received a total of 11 courses of treatment. Cmax increased proportionally with dose. Preclinically determined efficacious blood level (1 µM) of drug was attained at the highest dose level. One patient, at dose level 6, experienced grade 2 mucositis, nausea, vomiting, and anorexia. Four patients developed thromboembolic events subsequently considered as dose-limiting toxicity. No antitumor activity was noted. CONCLUSION: A causal relationship of observed thromboembolic events to CDDO was considered possible but could not be established.


Subject(s)
Neoplasms/drug therapy , Oleanolic Acid/analogs & derivatives , Anorexia/chemically induced , Apoptosis/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Humans , Immunoblotting , Infusions, Intravenous , Jurkat Cells , Male , Metabolic Clearance Rate , Middle Aged , Mucositis/chemically induced , Nausea/chemically induced , Neoplasms/metabolism , Neoplasms/pathology , Oleanolic Acid/adverse effects , Oleanolic Acid/pharmacokinetics , Oleanolic Acid/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Thromboembolism/chemically induced , Treatment Outcome , Vomiting/chemically induced
2.
J Clin Pharmacol ; 49(3): 351-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19246732

ABSTRACT

The US Food and Drug Administration draft drug interaction guidance recommends that 400 mg ketoconazole (KTZ) be administered once daily for several days (QD400) for maximal CYP3A inhibition. Some investigators suggest that a single dose of 400 mg (SD400) KTZ is sufficient given its short half-life (t(1/2) approximately 3-5 hr). To determine the impact of KTZ regimens on CYP3A inhibition, we simulated AUC fold-change (AUCR) in the presence of SD400, QD400, or 200 mg twice-daily (BID200) KTZ for theoretical CYP3A substrates. Ratios of AUCR (AUCR(QD400)/AUCR(SD400) and AUCR(BID200) AUCR(QD400)) increase with increasing bioavailability and increasing substrate t(1/2). The SD400 KTZ regimen may provide maximal inhibition only for a subset of substrates (ie, low bioavailability and short t(1/2)). For substrates with t(1/2) longer than that of KTZ, multiple KTZ dosing is critical and BID200 appears to provide greater inhibition than QD400. Also, timing of KTZ administration should be optimized to allow maximal presystemic enzyme inhibition prior to substrate administration.


Subject(s)
Antifungal Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors , Ketoconazole/pharmacokinetics , Models, Biological , Adult , Aged , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Area Under Curve , Biological Availability , Cross-Over Studies , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Female , Half-Life , Humans , Ketoconazole/metabolism , Ketoconazole/pharmacology , Male , Middle Aged , Time Factors , Young Adult
3.
J Clin Pharmacol ; 48(6): 662-70, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18378963

ABSTRACT

Predicting clinically significant drug interactions during drug development is a challenge for the pharmaceutical industry and regulatory agencies. Since the publication of the US Food and Drug Administration's (FDA's) first in vitro and in vivo drug interaction guidance documents in 1997 and 1999, researchers and clinicians have gained a better understanding of drug interactions. This knowledge has enabled the FDA and the industry to progress and begin to overcome these challenges. The FDA has continued its efforts to evaluate methodologies to study drug interactions and communicate recommendations regarding the conduct of drug interaction studies, particularly for CYP-based and transporter-based drug interactions, to the pharmaceutical industry. A drug interaction Web site was established to document the FDA's current understanding of drug interactions (http://www.fda.gov/cder/drug/drugInteractions/default.htm). This report provides an overview of the evolution of the drug interaction guidances, includes a synopsis of the steps taken by the FDA to revise the original drug interaction guidance documents, and summarizes and highlights updated sections in the current guidance document, Drug Interaction Studies-Study Design, Data Analysis, and Implications for Dosing and Labeling.


Subject(s)
Drug Design , Drug Interactions , Guidelines as Topic , Biological Transport/drug effects , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Humans , United States , United States Food and Drug Administration
4.
Mol Pharm ; 3(1): 62-9, 2006.
Article in English | MEDLINE | ID: mdl-16686370

ABSTRACT

Recently, increased interest in drug transporters and research in this area has revealed that drug transporters play an important role in modulating drug absorption, distribution, and elimination. Acting alone or in concert with drug metabolizing enzymes they can affect the pharmacokinetics and pharmacodynamics of a drug. This commentary will focus on the potential role that drug transporters may play in drug-drug interactions and what information may be needed during drug development and new drug application (NDA) submissions to address potential drug interactions mediated by transporters.


Subject(s)
Drug Interactions , Membrane Transport Proteins/physiology , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Structure-Activity Relationship
5.
Chem Res Toxicol ; 19(3): 376-81, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16544941

ABSTRACT

17-Dimethylaminoethylamino-17-demethoxygeldanamycin (DMAG) and 17-allylamino-17-demethoxygeldanamycin (17-AAG) are two derivatives of geldanamycin (GA) that are currently undergoing clinical evaluation as anticancer agents. These agents bind to heat shock protein 90 (hsp90), resulting in the destabilization of client proteins and inhibition of tumor growth. In a search for the mechanism of hepatotoxicity, which is a dose-limiting toxicity for these agents, we found that GA and its derivatives, 17-AAG and 17-DMAG, react chemically (i.e., nonenzymatically) with glutathione (GSH). A combination of liquid chromatography/electrospray ionization/mass spectrometry and nuclear magnetic resonance analyses were used to identify the product of this reaction as a GSH adduct in which the thiol group of GSH is substituted in the 19-position of the benzoquinone ring. The reaction proceeds rapidly with GA and 17-DMAG (half-lives of approximately 1.5 and 36 min, respectively) and less rapidly with 17-AAG and its major metabolite, 17-AG (half-lives of approximately 9.8 and 16.7 h). The reaction occurs at pH 7.0, 37 degrees C, and a physiological concentration of GSH, indicating that cellular GSH could play a role in modulating the cellular toxicity of these agents and therefore be a factor in their mechanism of differential toxicity. Moreover, reactions with thiol groups of critical cellular proteins could be important to the mechanism of toxicity with this class of anticancer agents.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Glutathione/chemistry , Quinones/chemistry , Benzoquinones , Buffers , Chromatography, High Pressure Liquid , Drug Stability , HSP90 Heat-Shock Proteins/chemistry , Lactams, Macrocyclic , Magnetic Resonance Spectroscopy , Mass Spectrometry , Phosphates/chemistry , Sulfhydryl Compounds/chemistry
6.
Chem Res Toxicol ; 18(12): 1842-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16359174

ABSTRACT

Evidence has been presented suggesting that a reactive metabolite, 2-phenylpropenal (ATPAL), may be responsible for the toxicities observed during therapy with the antiepileptic drug felbamate (FBM). Formation of ATPAL from its unstable immediate precursor, 3-carbamoyl-2-phenylpropionaldedhyde (CBMA) requires the loss of the hydrogen atom at position 2 in the propane chain, and it has been postulated that substitution of this atom with fluorine would prevent the formation of ATPAL. On the basis of this hypothesis, 2-fluoro-2-phenyl-1,3-propanediol dicarbamate (F-FBM) was synthesized and is presently undergoing drug development. To test this hypothesis, we compared the metabolism by human liver postmitochondrial suspensions (S9) in vitro of selected FBM and postulated F-FBM metabolites leading to formation of CBMA or 3-carbamoyl-2-fluoro-2-phenyl-propionaldehyde (F-CBMA). All S9 incubations included GSH as a trapping agent for any reactive metabolites formed. Our results indicated that, in phosphate buffer, pH 7.4, at 37 degrees C, the half-life for 4-hydroxy-5-phenyltetrahydro-1,3-oxazin-2-one (CCMF) was 2.8 and 3.6 h in the presence or absence of GSH, respectively; compared to 4-hydroxy-5-fluoro-5-phenyl-tetrahydro-1,3-oxazin-2-one (F-CCMF) which lost only 2.5% or 4.9% over 24 h under the same conditions. When incubated with S9 in the presence of the cofactor, NAD+, 2-phenyl-1,3-propanediol monocarbamate (MCF) was oxidized to CCMF which was further oxidized to 3-carbamoyl-2-phenylpropionic acid (CPPA). 2-Fluoro-2-phenyl-1,3-propanediol monocarbamate (F-MCF) under similar conditions was stable, and no metabolites were observed. When CCMF was incubated with S9 in the presence of NAD+ cofactor, oxidation to CPPA and reduction to MCF were observed. In addition, a new atropic acid GSH adduct (ATPA-GSH) was identified by mass spectrometry. When F-CCMF was incubated under the same conditions as CCMF, both reduced and oxidized metabolites, F-MCF and 3-carbamoyl-2-fluoro-2-phenylpropionic acid (F-CPPA), respectively, were formed but at significantly lower rates, and no GSH conjugates were identified. Our results support the hypothesis that F-FBM and F-CCMF are not metabolized by S9 in vitro to the known reactive FBM metabolite, ATPAL.


Subject(s)
Aza Compounds/chemistry , Fluorine/chemistry , Mitochondria, Liver/metabolism , Propylene Glycols/chemistry , Propylene Glycols/metabolism , Aldehydes/chemistry , Anticonvulsants/chemistry , Aza Compounds/metabolism , Cells, Cultured , Felbamate , Fluorine/metabolism , Humans , Mass Spectrometry , Mitochondria, Liver/chemistry , NAD/chemistry , Oxazines , Oxidation-Reduction , Phenylcarbamates , Signal Transduction
7.
Chem Biol Interact ; 142(1-2): 43-55, 2002 Nov 10.
Article in English | MEDLINE | ID: mdl-12399154

ABSTRACT

The quantification and identification of xenobiotic reactive intermediates is difficult in the absence of highly radiolabeled drug. We have developed a method for identifying these intermediates by measuring the formation of adducts to intracellularly generated radiolabeled glutathione (GSH). Freshly isolated adherent rat and human hepatocytes were incubated overnight in methionine and cystine-free ('thio-free') medium. They were then exposed to 100 microM methionine and 10 microCi 35S-labeled methionine in otherwise thio-free medium to replete cellular GSH pools with intracellularly generated 35S-labeled GSH. After 3 h, acetaminophen was added as a test compound and the cells were incubated for an additional 24 h. Intracellular GSH and its specific activity were quantified after reaction with monobromobimane followed by HPLC analysis with fluorescence and radiochemical detection. Radiolabeled GSH was detectable at 3 h and maintained high specific activity and physiological concentrations for up to 24 h. Incubation medium from acetaminophen treated and nontreated hepatocytes were analyzed for radiolabeled peaks by HPLC using radiochemical detection. Radiolabeled peaks not present in nontreated hepatocytes were identified as acetaminophen GSH adducts by LC-MS. Formation of acetaminophen 35S-GSH adducts by rat hepatocytes containing endogenously synthesized 35S-GSH was increased with acetaminophen concentrations ranging from 500 to 2 mM.


Subject(s)
Acetaminophen/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Glutathione/metabolism , Liver/metabolism , Methionine/metabolism , Acetaminophen/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Carbon Radioisotopes , Chromatography, Liquid , Cystine/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mass Spectrometry , Rats , Sulfur Radioisotopes
8.
Chem Biol Interact ; 142(1-2): 119-34, 2002 Nov 10.
Article in English | MEDLINE | ID: mdl-12399159

ABSTRACT

Antiepileptic therapy with a broad spectrum drug felbamate (FBM) has been limited due to reports of hepatotoxicity and aplastic anemia associated with its use. It was proposed that a bioactivation of FBM leading to formation of alpha,beta-unsaturated aldehyde, atropaldehyde (ATPAL) could be responsible for toxicities associated with the parent drug. Other members of this class of compounds, acrolein and 4-hydroxynonenal (HNE), are known for their reactivity and toxicity. It has been proposed that the bioactivation of FBM to ATPAL proceeds though a more stable cyclized product, 4-hydroxy-5-phenyltetrahydro-1,3-oxazin-2-one (CCMF) whose formation has been shown recently. Aldehyde dehydrogenase (ALDH) and glutathione transferase (GST) are detoxifying enzymes and targets for reactive aldehydes. This study examined effects of ATPAL and its precursor, CCMF on ALDH, GST and cell viability in liver, the target tissue for its metabolism and toxicity. A known toxin, HNE, which is also a substrate for ALDH and GST, was used for comparison. Interspecies difference in metabolism of FBM is well documented, therefore, human tissue was deemed most relevant and used for these studies. ATPAL inhibited ALDH and GST activities and led to a loss of hepatocyte viability. Several fold greater concentrations of CCMF were necessary to demonstrate a similar degree of ALDH inhibition or cytotoxicity as observed with ATPAL. This is consistent with CCMF requiring prior conversion to the more proximate toxin, ATPAL. GSH was shown to protect against ALDH inhibition by ATPAL. In this context, ALDH and GST are detoxifying pathways and their inhibition would lead to an accumulation of reactive species from FBM metabolism and/or metabolism of other endogenous or exogenous compounds and predisposing to or causing toxicity. Therefore, mechanisms of reactive aldehydes toxicity could include direct interaction with critical cellular macromolecules or indirect interference with cellular detoxification mechanisms.


Subject(s)
Anticonvulsants/toxicity , Liver/drug effects , Propylene Glycols/toxicity , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Aldehydes/pharmacology , Aldehydes/toxicity , Anticonvulsants/metabolism , Enzyme Inhibitors/pharmacology , Felbamate , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Humans , Liver/enzymology , Liver/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Phenylcarbamates , Propylene Glycols/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...