Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Anticancer Drugs ; 24(9): 958-68, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23928571

ABSTRACT

Combination therapy is increasingly being utilized for the treatment of metastatic breast cancer. However, coadministration of drugs, particularly agents that are substrates for or inhibitors of p-glycoprotein, can result in increased tissue toxicity. Unfortunately, determination of levels of chemotherapeutics in human tissues is challenging, and plasma drug concentrations are not always indicative of tissue toxicokinetics or toxicodynamics, especially when tissue penetration is altered. The aim of the present work was to determine whether concomitant administration of compounds currently being combined in clinical trials for metastatic breast cancer treatment alters plasma and tissue pharmacokinetics in mice if both agents are p-glycoprotein substrates and/or inhibitors. Accordingly, we investigated the pharmacokinetic interactions of the classic cytotoxics and p-glycoprotein substrates docetaxel and doxorubicin when administered concurrently with the targeted agent and p-glycoprotein inhibitor lapatinib. Our time-course plasma and tissue distribution studies showed that coadministration of lapatinib with doxorubicin did not appreciably alter the pharmacokinetics of this anthracycline in the plasma or six tissues evaluated in mice, presumably because, at doses relevant to human exposure, lapatinib inhibition of p-glycoprotein did not significantly alter doxorubicin transport out of these tissue compartments. However, combining lapatinib with docetaxel significantly increased intestinal exposure to this chemotherapeutic, which has clinical implications for enhancing gastrointestinal toxicity. The significant lapatinib-docetaxel interaction is likely CYP3A4-mediated, suggesting that caution should be exercised when this combination is administered, particularly to patients with compromised CYP3A activity, and recipients should be monitored closely for enhanced toxicity, particularly for adverse effects on the intestine.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Doxorubicin/pharmacokinetics , Intestine, Small/drug effects , Quinazolines/pharmacokinetics , Taxoids/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Clinical Trials, Phase I as Topic , Cytochrome P-450 CYP3A/metabolism , Docetaxel , Doxorubicin/adverse effects , Doxorubicin/blood , Doxorubicin/metabolism , Drug Interactions , Female , Half-Life , Humans , Intestine, Small/metabolism , Lapatinib , Membrane Transport Modulators/adverse effects , Membrane Transport Modulators/blood , Membrane Transport Modulators/metabolism , Membrane Transport Modulators/pharmacokinetics , Mice , Mice, Inbred Strains , Neoplasm Metastasis/drug therapy , Quinazolines/adverse effects , Quinazolines/blood , Quinazolines/metabolism , Taxoids/adverse effects , Taxoids/blood , Taxoids/metabolism , Tissue Distribution/drug effects , Topoisomerase II Inhibitors/blood , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/pharmacokinetics , Tubulin Modulators/adverse effects , Tubulin Modulators/blood , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacokinetics
2.
J Pharmacokinet Pharmacodyn ; 40(4): 437-49, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23616082

ABSTRACT

Docetaxel is one of the most widely used anticancer agents. While this taxane has proven to be an effective chemotherapeutic drug, noteworthy challenges exist in relation to docetaxel administration due to the considerable interindividual variability in efficacy and toxicity associated with the use of this compound, largely attributable to differences between individuals in their ability to metabolize and eliminate docetaxel. Regarding the latter, the ATP-binding cassette transporter B1 (ABCB1, PGP, MDR1) is primarily responsible for docetaxel elimination. To further understand the role of ABCB1 in the biodistribution of docetaxel in mice, we utilized physiologically-based pharmacokinetic (PBPK) modeling that included ABCB1-mediated transport in relevant tissues. Transporter function was evaluated by studying docetaxel pharmacokinetics in wild-type FVB and Mdr1a/b constitutive knockout (KO) mice and incorporating this concentration-time data into a PBPK model comprised of eight tissue compartments (plasma, brain, heart, lung, kidney, intestine, liver and slowly perfused tissues) and, in addition to ABCB1-mediated transport, included intravenous drug administration, specific binding to intracellular tubulin, intestinal and hepatic metabolism, glomerular filtration and tubular reabsorption. For all tissues in both the FVB and KO cohorts, the PBPK model simulations closely mirrored the observed data. Furthermore, both models predicted AUC values that were with 15 % of the observed AUC values, indicating that our model-simulated drug exposures accurately reflected the observed tissue exposures. Overall, our PBPK model furthers the understanding of the role of ABCB1 in the biodistribution of docetaxel. Additionally, this exemplary model structure can be applied to investigate the pharmacokinetics of other ABCB1 transporter substrates.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/pharmacokinetics , Taxoids/pharmacokinetics , Administration, Intravenous , Animals , Area Under Curve , Biological Transport , Docetaxel , Female , Glomerular Filtration Rate , Mice , Mice, Knockout , Models, Biological , Tissue Distribution , Tubulin/metabolism
3.
J Pharmacokinet Pharmacodyn ; 40(2): 157-76, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23315145

ABSTRACT

Lapatinib is an oral 4-anilinoquinazoline derivative that dually inhibits epidermal growth factor receptor and human epidermal growth factor receptor 2 (HER2). This drug is a mere decade old and has only been approved by the FDA for the treatment of breast cancer since 2007. Consequently, the intricacies of the pharmacokinetics are still being elucidated. In the work presented herein, we determined the biodistribution of orally administered lapatinib in mouse plasma, brain, heart, lung, kidney, intestine, liver, muscle and adipose tissue. Using this data, we subsequently developed a physiologically based pharmacokinetic (PBPK) model of lapatinib in mice that accurately predicted the tissue concentrations after doses of 30, 60 and 90 mg/kg. By taking into account interspecies differences in physiology and physiochemistry, we then extrapolated the mouse PBPK model to humans. Our model predictions closely reflected lapatinib plasma pharmacokinetics in healthy subjects. Additionally, we were also able to simulate the pharmacokinetics of this drug in the plasma of patients with solid malignancies by incorporating a decrease in liver metabolism into the model. Finally, our PBPK model also facilitated the estimation of various human tissue exposures to lapatinib, which harmonize with the organ-specific toxicities observed in clinical trials. This first-generation PBPK model of lapatinib can be further improved with a greater understanding of lapatinib absorption, distribution, metabolism and excretion garnered from subsequent in vitro and in vivo studies and expanded to include other pharmacokinetic determinants, including efflux transporters, metabolite generation, combination dosing, etc., to better predict lapatinib disposition in both mouse and man.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Quinazolines/pharmacokinetics , Tissue Distribution/physiology , Administration, Oral , Animals , Female , Humans , Lapatinib , Liver/metabolism , Mice , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Quinazolines/pharmacology
4.
Am J Vet Res ; 72(6): 764-71, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21627522

ABSTRACT

OBJECTIVE: To determine plasma concentrations and behavioral, antinociceptive, and physiologic effects of methadone administered via IV and oral transmucosal (OTM) routes in cats. ANIMALS: 8 healthy adult cats. PROCEDURES: Methadone was administered via IV (0.3 mg/kg) and OTM (0.6 mg/kg) routes to each cat in a balanced crossover design. On the days of drug administration, jugular catheters were placed in all cats under anesthesia; a cephalic catheter was also placed in cats that received methadone IV. Baseline measurements were obtained ≥ 90 minutes after extubation, and methadone was administered via the predetermined route. Heart and respiratory rates were measured; sedation, behavior, and antinociception were evaluated, and blood samples were collected for methadone concentration analysis at predetermined intervals for 24 hours after methadone administration. Data were summarized and evaluated statistically. RESULTS: Plasma concentrations of methadone were detected rapidly after administration via either route. Peak concentration was detected 2 hours after OTM administration and 10 minutes after IV administration. Mean ± SD peak concentration was lower after OTM administration (81.2 ± 14.5 ng/mL) than after IV administration (112.9 ± 28.5 ng/mL). Sedation was greater and lasted longer after OTM administration. Antinociceptive effects were detected 10 minutes after administration in both groups; these persisted ≥ 2 hours after IV administration and ≥ 4 hours after OTM administration. CONCLUSIONS AND CLINICAL RELEVANCE: Despite lower mean peak plasma concentrations, duration of antinociceptive effects of methadone was longer after OTM administration than after IV administration. Methadone administered via either route may be useful for perioperative pain management in cats.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Catheterization, Central Venous/veterinary , Cats , Methadone/administration & dosage , Methadone/pharmacokinetics , Pain/veterinary , Administration, Buccal , Analgesia/veterinary , Analgesics, Opioid/blood , Analgesics, Opioid/therapeutic use , Animals , Behavior, Animal , Catheterization, Central Venous/adverse effects , Cross-Over Studies , Female , Injections, Intravenous/veterinary , Jugular Veins/surgery , Male , Methadone/blood , Methadone/therapeutic use , Pain/drug therapy
5.
Vet Ophthalmol ; 14(3): 180-5, 2011 May.
Article in English | MEDLINE | ID: mdl-21521442

ABSTRACT

PURPOSE: Oxidative stress plays a major role in the pathogenesis of many neurodegenerative diseases. It has also been implicated as part of the pathogenic mechanisms in the development of glaucoma. Alpha-luminol has shown profound anti-inflammatory and antioxidant effects in both experimental animal and human clinical studies. The purpose of this pilot study was to investigate for the first time the ocular penetration of topical alpha-luminol. METHODS: Nine animals were divided into three treated groups (three animals each; one drop OU/n = 18), each group receiving a different concentration of the eyedrop (0.5%, 1.5%, 2.5%). Aqueous humor and peripheral blood samples were obtained from each rabbit at three different timepoints (20 min, 4 h and 12 h). Samples were analyzed by means of high performance liquid chromatography and mass spectrometry; median values were compared. RESULTS: Alpha-luminol was found in the aqueous humor in all treated groups at all timepoints. At the 2nd and 3rd timepoints (4 h and 12 h), aqueous humor levels decreased significantly (P < 0.05) for two of the three dosages tested and it was not detectable in some eyes. The highest aqueous humor concentration of the drug was 272 ng/mL after 20 min (0.0217% of one drop, 2.5% group). Alpha-luminol was found in the vitreous in two animals, one in the 1.5% and another in the 2.5% group (16.4 and 21.5 ng/mL, respectively), at 12 h. CONCLUSIONS: Topically administered alpha-luminol readily penetrates into the anterior chamber and can penetrate into the vitreous chamber. Further investigation is warranted to better understand the intraocular pharmacokinetics of alpha-luminol.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Eye/metabolism , Phthalazines/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dose-Response Relationship, Drug , Female , Luminol/analogs & derivatives , Male , Ophthalmic Solutions , Phthalazines/administration & dosage , Phthalazines/pharmacology , Pilot Projects , Rabbits
6.
J Pharm Sci ; 100(3): 1156-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20803616

ABSTRACT

Population pharmacokinetic (PK) analyses have been successfully incorporated into drug dosing optimization; however, these analyses necessitate relatively large patient cohorts that many clinical trials do not have the luxury of affording. To address this problem, we developed an approach that utilizes physiologically based pharmacokinetic (PBPK) modeling coupled with Monte Carlo simulation to generate a virtual population, complete with associated patient characteristics and PK data, for population PK analysis. For this work, we used a previously published PBPK model for docetaxel and found that the systemic clearance of this drug was significantly affected by blood volume, slowly perfused tissue volume, and two liver metabolic parameters--the maximum rate of liver metabolism and the Michaelis constant for liver metabolism. These findings, as well as the PK variability predictions, are consistent with those previously associated with docetaxel clearance in population PK analyses performed with actual patient populations, namely plasma protein levels, body size, and hepatic function. Thus, this in silico exercise demonstrates the utility of simulation modeling coupled to population PK analysis for the estimation of PK variability and the identification of patient characteristics that affect a drug's PK in the absence of data assembled from large clinical trials.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Taxoids/pharmacokinetics , Biochemical Phenomena , Computer Simulation , Demography , Docetaxel , Humans , Models, Biological , Monte Carlo Method , Physiological Phenomena , Tissue Distribution
7.
Cancer Chemother Pharmacol ; 65(4): 743-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19633850

ABSTRACT

PURPOSE: The aim of this study was to investigate typical population pharmacokinetic (PK) parameters, potential covariates, and interindividual and residual variabilities of PI-88, a heparanase endoglycosidase enzyme inhibitor being developed for the treatment of cancer. METHODS: A population PK model of PI-88 was developed and evaluated using nonlinear mixed effects modeling (NONMEM). Plasma concentration versus time data was obtained from a total of 76 subjects that participated in phase I trials of PI-88 delivered subcutaneously (SC) at doses ranging from 80 to 315 mg. Overall, the PK effects of 12 clinical covariates were evaluated, including weight, age, creatinine clearance, body surface area, body mass index, sex, cancer (vs. healthy subject), docetaxel coadministration, prior chemotherapy, prior investigational therapy, prior radiotherapy and prior surgery. RESULTS: Population PK analysis of the data-set showed that apparent clearance (CL/F) and apparent volume of distribution (V/F) of PI-88 were positively correlated with body surface area and the absorption rate constant (KA) was positively correlated with body mass index. In addition, CL/F was found to be significantly lower in patients with malignancies versus healthy subjects. By incorporating these covariates into the PK parameter equations, the interindividual variability of CL/F was reduced from 30.6 to 20.2% (decrease of 34%), V/F was reduced from 31.4 to 20.7% (decrease of 34.1%) and KA was reduced from 52.6 to 46.2% (decrease of 12.2%). CONCLUSIONS: This population PK model indicates that the PK variability of PI-88 can be significantly reduced by taking BSA into account when dosing this drug SC.


Subject(s)
Glucuronidase/antagonists & inhibitors , Models, Biological , Neoplasms/drug therapy , Oligosaccharides/pharmacokinetics , Adult , Aged , Algorithms , Area Under Curve , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Glucuronidase/metabolism , Humans , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/metabolism , Oligosaccharides/administration & dosage , Time Factors , Young Adult
8.
J Comp Physiol B ; 174(8): 633-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15517283

ABSTRACT

Leptin and adiponectin are proteins produced and secreted from white adipose tissue and are important regulators of energy balance and insulin sensitivity. Seasonal changes in leptin and adiponectin have not been investigated in mammalian hibernators in relationship to changes in fat cell and fat mass. We sought to determine the relationship between serum leptin and adiponectin levels with seasonal changes in lipid mass. We collected serum and tissue samples from marmots (Marmota flaviventris) in different seasons while measuring changes in fat mass, including fat-cell size. We found that leptin is positively associated with increasing fat mass and fat-cell size, while adiponectin is negatively associated with increasing lipid mass. These findings are consistent with the putative roles of these adipokines: leptin increases with fat mass and is involved in enhancing lipid oxidation while adiponectin appears to be higher in summer when hepatic insulin sensitivity should be maintained since the animals are eating. Our data suggest that during autumn/winter animals have switched from a lipogenic condition to a lipolytic state, which may include leptin resistance.


Subject(s)
Adipocytes/metabolism , Hibernation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Leptin/blood , Marmota/physiology , Weight Gain/physiology , Weight Loss/physiology , Adiponectin , Analysis of Variance , Animals , Blotting, Northern , Blotting, Western , Body Composition , Colorado , Electric Conductivity , Female , Male , Marmota/metabolism , Seasons
9.
Am J Physiol Regul Integr Comp Physiol ; 286(3): R498-504, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14656767

ABSTRACT

Yellow-bellied marmots (Marmota flaviventris) exhibit a circannual cycle of hyperphagia and nutrient storage in the summer followed by hibernation in the winter. This annual cycle of body mass gain and loss is primarily due to large-scale accumulation of lipid in the summer, which is then mobilized and oxidized for energy during winter. The rapid and predictable change in body mass makes these animals ideal for studies investigating the molecular basis for body weight regulation. In the study described herein, we monitored seasonal changes in the protein levels and activity of a central regulator of anabolic metabolism, the serine-threonine kinase Akt-protein kinase B (Akt/PKB), during the months accompanying maximal weight gain and entry into hibernation (June-November). Interestingly, under fasting conditions, Akt/PKB demonstrated a tissue-specific seasonal activation. Specifically, although Akt/PKB levels did not change, the activity of Akt/PKB (isoforms 1/alpha and 2/beta) in white adipose tissue (WAT) increased significantly in July. Moreover, glycogen synthase, which lies downstream of Akt/PKB on a linear pathway linking the enzyme to the stimulation of glycogen synthesis, demonstrated a similar pattern of seasonal activation. By contrast, Akt/PKB activity in skeletal muscle peaked much later (i.e., September). These data suggest the existence of a novel, tissue-specific mechanism regulating Akt/PKB activation during periods of marked anabolism.


Subject(s)
Glycogen Synthase/physiology , Hibernation/physiology , Marmota/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Seasons , Adipose Tissue/physiology , Animals , Blotting, Western , Body Weight/physiology , Female , Insulin/blood , Insulin/physiology , Lipid Metabolism , Male , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiology , Organ Specificity , Phosphorylation , Proto-Oncogene Proteins c-akt , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...