Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(8): 6064-6080, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38595098

ABSTRACT

It has been shown that PRMT5 inhibition by small molecules can selectively kill cancer cells with homozygous deletion of the MTAP gene if the inhibitors can leverage the consequence of MTAP deletion, namely, accumulation of the MTAP substrate MTA. Herein, we describe the discovery of TNG908, a potent inhibitor that binds the PRMT5·MTA complex, leading to 15-fold-selective killing of MTAP-deleted (MTAP-null) cells compared to MTAPintact (MTAP WT) cells. TNG908 shows selective antitumor activity when dosed orally in mouse xenograft models, and its physicochemical properties are amenable for crossing the blood-brain barrier (BBB), supporting clinical study for the treatment of both CNS and non-CNS tumors with MTAP loss.


Subject(s)
Antineoplastic Agents , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Humans , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemical synthesis , Drug Discovery , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Cell Line, Tumor , Xenograft Model Antitumor Assays , Neoplasms/drug therapy , Brain/metabolism , Structure-Activity Relationship
2.
Clin Transl Sci ; 14(2): 536-543, 2021 03.
Article in English | MEDLINE | ID: mdl-33048459

ABSTRACT

Many targeted therapies are administered at or near the maximum tolerated dose (MTD). With the advent of precision medicine, a larger therapeutic window is expected. Therefore, dose optimization will require a new approach to early clinical trial design. We analyzed publicly available data for 21 therapies targeting six kinases, and four poly (ADP-ribose) polymerase inhibitors, focusing on potency and exposure to gain insight into dose selection. The free average steady-state concentration (Css ) at the approved dose was compared to the in vitro cell potency (half-maximal inhibitory concentration (IC50 )). Average steady-state area under the plasma concentration-time curve, the fraction unbound drug in plasma, and the cell potency were taken from the US drug labels, US and European regulatory reviews, and peer-reviewed journal articles. The Css was remarkably similar to the IC50 . The median Css /IC50 value was 1.2, and 76% of the values were within 3-fold of unity. However, three drugs (encorafenib, erlotinib, and ribociclib) had a Css /IC50 value > 25. Seven other therapies targeting the same 3 kinases had much lower Css /IC50 values ranging from 0.5 to 4. These data suggest that these kinase inhibitors have a large therapeutic window that is not fully exploited; lower doses may be similarly efficacious with improved tolerability. We propose a revised first-in-human trial design in which dose cohort expansion is initiated at doses less than the MTD when there is evidence of clinical activity and Css exceeds a potency threshold. This potency-guided approach is expected to maximize the therapeutic window thereby improving patient outcomes.


Subject(s)
Antineoplastic Agents/administration & dosage , Clinical Trials, Phase I as Topic/methods , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Maximum Tolerated Dose , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Research Design
3.
J Occup Environ Hyg ; 12(5): 342-9, 2015.
Article in English | MEDLINE | ID: mdl-25647534

ABSTRACT

Upper tolerance limits (UTLs) are often used in comparing exposure data sets with an occupational exposure limit (OEL) or other regulatory criterion (RC): if the 95%-95% UTL does not exceed the OEL, one is 95% confident that at most 5% of exposures exceed the OEL, and the comparison "passes." The largest of 59 observations is a nonparametric (distribution-free) 95%-95% UTL (NPUTL); the chance that this largest value equals or exceeds the actual 95th percentile is at least 95%, regardless of the underlying data distribution. That many observations may seem excessive in clean environments or small studies, though, and one would like to "pass" using UTLs based on fewer observations sufficiently far below the OEL or RC. "Quasi-nonparametric" UTLs (QNP UTLs) accomplish this. QNP UTLs assign a "pass" so long as one has "59 [values] less than the RC" (the NPUTL itself), "30 less than 1/2 [of the RC]," "21 less than 1/3," and on down to "8 less than 1/10," the last matching a rule-of-thumb given in 2006 American Industrial Hygiene Association (AIHA) guidance. They are derived using the conservative, experience-based assumption that the data distribution is lognormal with log-scale standard deviation σ at most 2.0 (geometric standard deviation at most 7.39). Although based on this assumption, their statistical performance is reasonably unaffected or conservative when data come from other distributions often assumed for contaminant concentrations; moreover, their performance is insensitive to analytical variation. This conservative robustness merits the description "quasi-nonparametric." QNP UTLs are very easy to use. Reporting Limit (RL) issues do not arise. QNP UTLs reduce the numbers of observations needed to support conservative risk management decisions when sampling from compliant working conditions.


Subject(s)
Occupational Exposure/standards , Occupational Health/standards , Environmental Monitoring/standards , Humans , Occupational Exposure/analysis , Probability , Risk Assessment , United States
4.
Bioconjug Chem ; 25(12): 2123-8, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25418333

ABSTRACT

The complementary nature of positron emission tomography (PET) and optical imaging (OI) has fueled increasing interest in the development of multimodal PET/OI probes that can be employed during the diagnosis, staging, and surgical treatment of cancer. Due to their high selectivity and affinity, antibodies have emerged as promising platforms for the development of hybrid PET/OI agents. However, the lack of specificity of many bioconjugation reactions can threaten immunoreactivity and lead to poorly defined constructs. To circumvent this issue, we have developed a chemoenzymatic strategy for the construction of multimodal PET/OI immunoconjugates that have been site-specifically labeled on the heavy chain glycans. The methodology consists of four steps: (1) the enzymatic removal of the terminal galactose residues on the heavy chain glycans; (2) the enzymatic incorporation of azide-bearing galactose (GalNAz) residues into the heavy chain glycans; (3) the strain-promoted click conjugation of chelator- and fluorophore-modified dibenzocyclooctynes to the azide-modified sugars; and (4) the radiolabeling of the immunoconjugate. For proof-of-concept, a model system was created using the colorectal cancer-targeting antibody huA33, the chelator desferrioxamine (DFO), the positron-emitting radiometal (89)Zr, and the near-infrared fluorescent dye Alexa Fluor 680. The bioconjugation strategy is robust and reproducible, reliably producing well-defined and immunoreactive conjugates labeled with (89)Zr, Alexa Fluor 680, or an easily and precisely tuned mixture of the two reporters. In in vivo PET and fluorescence imaging experiments, a hybrid (89)Zr- and Alexa Fluor 680-labeled huA33 conjugate displayed high levels of specific uptake (>45% ID/g) in athymic nude mice bearing A33 antigen-expressing SW1222 colorectal cancer xenografts.


Subject(s)
Immunoconjugates/chemistry , Optical Imaging/methods , Positron-Emission Tomography/methods , Animals , Colorectal Neoplasms/diagnosis , Electrophoresis, Polyacrylamide Gel , Fluorescent Dyes/chemistry , Galactose/chemistry , Humans , Immunoconjugates/pharmacokinetics , Mice , Multimodal Imaging , Neoplasms, Experimental/diagnosis , Polysaccharides/chemistry , Polysaccharides/immunology , Radioisotopes , Tissue Distribution , Xenograft Model Antitumor Assays , Zirconium
5.
Bioconjug Chem ; 24(6): 1057-67, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23688208

ABSTRACT

An enzyme- and click chemistry-mediated methodology for the site-selective radiolabeling of antibodies on the heavy chain glycans has been developed and validated. To this end, a model system based on the prostate specific membrane antigen-targeting antibody J591, the positron-emitting radiometal (89)Zr, and the chelator desferrioxamine has been employed. The methodology consists of four steps: (1) the removal of sugars on the heavy chain region of the antibody to expose terminal N-acetylglucosamine residues; (2) the incorporation of azide-modified N-acetylgalactosamine monosaccharides into the glycans of the antibody; (3) the catalyst-free click conjugation of desferrioxamine-modified dibenzocyclooctynes to the azide-bearing sugars; and (4) the radiolabeling of the chelator-modified antibody with (89)Zr. The site-selective labeling methodology has proven facile, reproducible, and robust, producing (89)Zr-labeled radioimmunoconjguates that display high stability and immunoreactivity in vitro (>95%) in addition to highly selective tumor uptake (67.5 ± 5.0%ID/g) and tumor-to-background contrast in athymic nude mice bearing PSMA-expressing subcutaneous LNCaP xenografts. Ultimately, this strategy could play a critical role in the development of novel well-defined and highly immunoreactive radioimmunoconjugates for both the laboratory and clinic.


Subject(s)
Antibodies/metabolism , Deferoxamine/metabolism , Isotope Labeling , Organometallic Compounds/metabolism , Zirconium/metabolism , beta-Galactosidase/metabolism , Animals , Antibodies/chemistry , Binding Sites , Click Chemistry , Deferoxamine/chemistry , Humans , Male , Mice , Mice, Nude , Molecular Structure , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Polysaccharides/chemistry , Polysaccharides/metabolism , Tumor Cells, Cultured , Zirconium/chemistry , beta-Galactosidase/chemistry
6.
ACS Med Chem Lett ; 3(7): 524-9, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-24900504

ABSTRACT

A novel thiazolopyrimidinone series of PI3K-beta selective inhibitors has been identified. This chemotype has provided an excellent tool compound, 18, that showed potent growth inhibition in the PTEN-deficient breast cancer cell line MDA-MB-468 under anchorage-independent conditions, and it also demonstrated pharmacodynamic effects and efficacy in a PTEN-deficient prostate cancer PC-3 xenograft mouse model.

8.
Toxicol Pathol ; 38(5): 691-702, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20616376

ABSTRACT

Several multikinase angiogenesis inhibitors demonstrate mitochondrial and/or cardiovascular toxicity, suggesting an on-target pharmacologic effect. To evaluate whether cardiotoxicity is directly related to vascular endothelial growth factor receptor inhibition, we investigated the effects of sunitinib, sorafenib, and pazopanib on myocardial function and structure. We used a rat model to assess myocardial effects of the inhibitors concurrently exposed to the cardiac stressor dobutamine. Echocardiographic abnormalities including premature ventricular contractions, decreases in heart rate, circumferential strain, and radial and circumferential strain rates were noted with sorafenib, but not with sunitinib or pazopanib. Ultrastructural analysis of ventricular cardiomyocytes by transmission electron microscopy revealed mitochondrial swelling, dense deposits, and matrix cavitation in rats given sunitinib and disrupted mitochondrial cristae in rats given sorafenib, but there were no effects with pazopanib. Effects on neonatal rat cardiomyocyte cultures were assessed, which identified decreases in mitochondrial membrane potential with sunitinib treatment, but not with sorafenib or pazopanib. Intracellular adenosine triphosphate depletion was observed with sunitinib and sorafenib, but not pazopanib. Our results show that cardiotoxicity is not necessarily related to a pharmacologic classwide effect of vascular endothelial growth factor receptor inhibition, and the rat myocardial structural and functional changes identified in this study may be instead a result of inhibition of other kinase pathways, the mechanism of which may be associated with mitochondrial toxicity.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Heart/drug effects , Mitochondria/drug effects , Myocardium/ultrastructure , Animals , Benzenesulfonates/adverse effects , Echocardiography , Immunohistochemistry , In Situ Nick-End Labeling , Indazoles , Indoles/adverse effects , Male , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , Rats , Sorafenib , Sulfonamides/adverse effects , Sunitinib , Troponin I/biosynthesis
9.
Drug Metab Dispos ; 37(12): 2375-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741038

ABSTRACT

The macrolide antibiotics azithromycin and clarithromycin are large molecular weight compounds that exhibit moderate to excellent oral bioavailability in preclinical species and humans. Previous concomitant dosing studies in rats using rifamycin SV, a general organic anion-transporting polypeptide (OATP) inhibitor, suggested that the high oral absorption of azithromycin and clarithromycin may be caused by facilitative uptake by intestinal Oatps. In this study, we used OATP/Oatp-expressing cells to investigate the interaction of macrolides with rat Oatp1a5, human OATP1A2, and human/rat OATP2B1/Oatp2b1. These experiments showed that azithromycin and clarithromycin were potent inhibitors of rat Oatp1a5-mediated taurocholate uptake with apparent inhibitor constant (K(i)) values of 3.3 and 2.4 microM, respectively. The macrolides functioned as noncompetitive inhibitors but were not transport substrates for rat Oatp1a5, as assessed by direct uptake measurements of radiolabeled azithromycin and clarithromycin. cis-Inhibition and direct uptake studies further showed that azithromycin and clarithromycin were only very weak inhibitors and not substrates for human OATP1A2 and human/rat OATP2B1/Oatp2b1. In summary, these results indicate that the macrolides azithromycin and clarithromycin potently inhibit rat Oatp1a5 but do not significantly interact with OATP1A2 and OATP2B1/Oatp2b1. These intestinally expressed OATP/Oatp(s) are not responsible for the postulated facilitative uptake of azithromycin and clarithromycin, and alternative facilitative pathways must exist for their intestinal absorption.


Subject(s)
Anti-Bacterial Agents/pharmacology , Intestines/drug effects , Macrolides/pharmacology , Organic Anion Transporters/antagonists & inhibitors , Animals , Anti-Bacterial Agents/metabolism , Azithromycin/pharmacology , Biological Transport , COS Cells , Chlorocebus aethiops , Clarithromycin/pharmacology , Dogs , Dose-Response Relationship, Drug , Erythromycin/pharmacology , Estrone/analogs & derivatives , Estrone/metabolism , Humans , Intestinal Mucosa/metabolism , Kinetics , Macrolides/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Rats , Taurocholic Acid/metabolism , Transfection
10.
J Med Chem ; 52(7): 1828-44, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19284751

ABSTRACT

On the basis of a mechanistic understanding of the toxicity of the 4-aminoquinoline amodiaquine (1b), three series of amodiaquine analogues have been prepared where the 4-aminophenol "metabolic alert" has been modified by replacement of the 4'-hydroxy group with a hydrogen, fluorine, or chlorine atom. Following antimalarial assessment and studies on mechanism of action, two candidates were selected for detailed ADME studies and in vitro and in vivo toxicological assessment. 4'-Fluoro-N-tert-butylamodiaquine (2k) was subsequently identified as a candidate for further development studies based on potent activity versus chloroquine-sensitive and resistant parasites, moderate to excellent oral bioavailability, low toxicity in in vitro studies, and an acceptable safety profile.


Subject(s)
Aminoquinolines/chemical synthesis , Amodiaquine/analogs & derivatives , Amodiaquine/chemical synthesis , Antimalarials/chemical synthesis , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Amodiaquine/chemistry , Amodiaquine/pharmacokinetics , Amodiaquine/pharmacology , Animals , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Cell Survival , Chloroquine/pharmacology , Dogs , Drug Resistance , Female , Haplorhini , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , In Vitro Techniques , Malaria/drug therapy , Malaria/parasitology , Male , Mice , Parasitic Sensitivity Tests , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Rats , Rats, Wistar , Structure-Activity Relationship
11.
J Med Chem ; 52(5): 1408-15, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19222165

ABSTRACT

N-tert-Butyl isoquine (4) (GSK369796) is a 4-aminoquinoline drug candidate selected and developed as part of a public-private partnership between academics at Liverpool, MMV, and GSK pharmaceuticals. This molecule was rationally designed based on chemical, toxicological, pharmacokinetic, and pharmacodynamic considerations and was selected based on excellent activity against Plasmodium falciparum in vitro and rodent malaria parasites in vivo. The optimized chemistry delivered this novel synthetic quinoline in a two-step procedure from cheap and readily available starting materials. The molecule has a full industry standard preclinical development program allowing first into humans to proceed. Employing chloroquine (1) and amodiaquine (2) as comparator molecules in the preclinical plan, the first preclinical dossier of pharmacokinetic, toxicity, and safety pharmacology has also been established for the 4-aminoquinoline antimalarial class. These studies have revealed preclinical liabilities that have never translated into the human experience. This has resulted in the availability of critical information to other drug development teams interested in developing antimalarials within this class.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/pharmacology , Benzylamines/pharmacology , Aminoquinolines/chemical synthesis , Aminoquinolines/chemistry , Aminoquinolines/pharmacokinetics , Aminoquinolines/toxicity , Amodiaquine/analogs & derivatives , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacokinetics , Antimalarials/toxicity , Benzylamines/chemical synthesis , Benzylamines/chemistry , Benzylamines/toxicity , Cytochrome P-450 Enzyme Inhibitors , Dogs , Drug Evaluation, Preclinical , Drug Resistance , Female , Haplorhini , Heme/chemistry , Humans , Malaria/drug therapy , Mice , Models, Molecular , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii , Rats , Structure-Activity Relationship
12.
J Pharm Sci ; 98(1): 362-77, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18563832

ABSTRACT

The disposition of three 4-aminoquinoline leads, namely isoquine (ISO), des-ethyl isoquine (DEI) and N-tert-butyl isoquine (NTBI), were studied in a range of in vivo and in vitro assays to assist in selecting an appropriate candidate for further development. Analogous to amodiaquine (ADQ), ISO undergoes oxidative N-dealkylation to form DEI in vivo. Blood clearance of DEI was as much as 10-fold lower than that of ISO in animals and after oral administration, metabolite exposure exceeded that of parent by as much as 14-fold. Replacement of the N-ethyl with an N-tert-butyl substituent substantially reduced N-dealkylation as blood clearance of NTBI was approximately 2 to 3-fold lower than DEI in mouse, rat, dog and monkey. Mean NTBI oral bioavailability was generally higher than the other leads (>/=68%). Blood cell association was substantial for NTBI, particularly in dog and monkey, where blood to plasma concentration ratios >4 were observed. Human plasma protein binding was similar for NTBI, DEI, and des-ethyl amodiaquine (DEA). Allometric scaling predicted human blood clearance (CL) for NTBI to be low ( approximately 12% liver blood flow). All the 4-aminoquinolines inhibited recombinant human cytochrome P450 2D6 with similar potency; DEI also inhibited 1A2. On balance, NTBI appeared the most promising lead to progress towards full development.


Subject(s)
Aminoquinolines/pharmacokinetics , Antimalarials/pharmacokinetics , Aminoquinolines/blood , Aminoquinolines/metabolism , Animals , Antimalarials/blood , Antimalarials/metabolism , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Male , Mice , Rats , Rats, Sprague-Dawley , Species Specificity
13.
Drug Metab Dispos ; 36(12): 2492-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18755851

ABSTRACT

Macrolide antibiotics azithromycin (AZI) and clarithromycin (CLARI) are large molecular weight compounds and are substrates for apically polarized efflux transporters such as P-glycoprotein, which can potentially restrict intestinal absorption. However, despite these undesired physicochemical and biopharmaceutical properties, AZI and CLARI exhibit moderate to excellent p.o. bioavailability in preclinical species and humans. Intestinal uptake transporters, such as organic anion transporting polypeptides (OATPs), can facilitate the uptake of drugs that are substrates and hence increase p.o. absorption. The present study was designed to determine whether the intestinal Oatps are involved in absorption of these macrolides. AZI or CLARI was dosed p.o. to Sprague-Dawley rats after p.o. administration with vehicle or rifamycin SV (RIF), an OATP inhibitor. The p.o. exposures of AZI and CLARI were reduced 65 and 45%, respectively, when coadministered with an optimized RIF regimen. The p.o. RIF had no affect on the total blood clearance of these macrolides and most likely did not cause induction of metabolizing enzymes and/or transporters. Therefore, the results suggest that inhibition of an RIF-sensitive uptake transporter such as Oatp along the rat gastrointestinal tract was responsible for reduced p.o. exposure of AZI and CLARI. In addition, AZI and CLARI caused inhibition of taurocholate uptake in rat Oatp1a5-transfected Madin-Darby canine kidney cell monolayers. The in vitro and in vivo results suggest that the intestinal Oatps are involved in the p.o. absorption of AZI and CLARI in the rat.


Subject(s)
Azithromycin/metabolism , Clarithromycin/metabolism , Intestinal Absorption/physiology , Organic Anion Transporters/metabolism , Administration, Oral , Animals , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/pharmacology , Area Under Curve , Azithromycin/pharmacokinetics , Azithromycin/pharmacology , Biological Transport, Active/drug effects , Cell Line , Clarithromycin/pharmacokinetics , Clarithromycin/pharmacology , Dogs , Histamine H1 Antagonists, Non-Sedating/pharmacology , Infusions, Intravenous , Intestinal Absorption/drug effects , Male , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Rats , Rats, Sprague-Dawley , Rifamycins/administration & dosage , Rifamycins/pharmacology , Taurocholic Acid/metabolism , Terfenadine/analogs & derivatives , Terfenadine/pharmacology , Transfection
14.
Drug Metab Dispos ; 36(9): 1846-52, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18556439

ABSTRACT

The pharmacokinetics and brain penetration of the novel neurokinin (NK)-1 receptor antagonist casopitant [1-piperidinecarboxamide, 4-(4-acetyl-1-piperazinyl)-N-((1R)-1-(3,5-bis(trifluoromethyl)phenyl)ethyl)-2-(4-fluoro-2-methylphenyl)-N-methyl-, (2R,4S)-; GW679769] were examined in ferrets. The ferret is known to respond to the full spectrum of agents recognized to induce emesis in humans, and the cisplatin-induced emesis models in the ferret have been used to establish the antiemetic potential of casopitant. Following single i.p. dosing to the ferret, casopitant was rapidly absorbed, with plasma and brain concentrations being approximately equal at 2 h postdose. The predominant radioactive component present in the ferret brain after a single dose of [(14)C]casopitant was parent compound, accounting for approximately 76% of the radioactivity. The major metabolites present in brain tissue following administration of [(14)C]casopitant were hydroxylated casopitant (M1) and the corresponding ketone product of the M1 metabolite (M2), which accounted for approximately 19 and 3% of the radioactivity in the brain extracts, respectively. All three molecules had relatively similar potency against ferret brain cortical NK-1, suggesting that the pharmacologic activity of casopitant in the ferret is largely attributable to parent compound and, to a lesser extent, to its oxidative metabolites. Because casopitant is intended to be administered in combination with ondansetron and because therapeutic synergy has been observed with this combination in the ferret, a drug interaction study was conducted. The additional pharmacodynamic benefit of the combination dose was not because of an alteration in the pharmacokinetics of either agent but is likely the result of the complementary mechanisms of pharmacologic action of the two drugs.


Subject(s)
Brain/metabolism , Neurokinin-1 Receptor Antagonists , Piperazines/pharmacokinetics , Piperidines/pharmacokinetics , Animals , Area Under Curve , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Ferrets , Male , Piperazines/metabolism , Piperazines/pharmacology , Piperidines/metabolism , Piperidines/pharmacology , Tandem Mass Spectrometry
15.
J Pharm Pharm Sci ; 10(4): 486-95, 2007.
Article in English | MEDLINE | ID: mdl-18396564

ABSTRACT

PURPOSE: Grobeta-T, a human CXC chemokine, has been studied for its potential to mobilize stem cells. Chemokines bind specifically to receptors on target immune cells but also to a homologous erythrocyte blood group antigen, the Duffy Antigen/Receptor for Chemokines (DARC)that is subject to genetic polymorphism in humans.A mutation in the DARC gene is common among African Americans and results in lack of expression of the erythrocyte antigen. We used a combination of in vitro studies of Grobeta-DARC interaction and pharmacokinetic simulation to anticipate the potential impact of this polymorphism on the pharmacokinetics of Grobeta-T. METHODS: [125I]Grobeta-T was incubated in Caucasian blood to characterize the concentration dependence of the blood to plasma concentration ratio (B/P). Affinity and capacity of binding was estimated by Scatchard analysis; specificity was investigated by competitive displacement with a CC chemokine. The B/P value (7 nM) was then determined in blood from 8 African American subjects. Duffy antigen expression was determined by antibody agglutination. A pharmacokinetic model was developed which accounted for blood-cell binding. Simulations were performed to explore effects of dose regimen and DARC expression on the Grobeta-T plasma concentration-time profile. RESULTS: Grobeta-T affinity and capacity for DARC (Caucasian blood)were 23.0 +/- 1.2 and 37.7 +/- 0.6 nM, respectively;excess CC chemokine fully displaced [125I]Grobeta-T. Chemokine binding was highly correlated with the presence or absence of the Duffy antigen (p<0.01) in African American blood; the proportion of subjects for which binding was observed (3/8), was consistent with the reported frequency of DARC expression in this population. Counter to intuition,in the terminal disposition phase at low doses,concentrations of free Grobeta-T in the presence of DARC may be substantially higher than in the absence of DARC. CONCLUSION: Dissociation from the erythrocyte antigen may lead to greater persistence, at low doses, of free Grobetabeta-T in the blood of individuals expressing the chemokine sink.


Subject(s)
Chemokine CXCL2/blood , Chemokine CXCL2/genetics , Duffy Blood-Group System/blood , Duffy Blood-Group System/genetics , Polymorphism, Genetic/genetics , Erythrocytes/immunology , Humans
16.
J Pharm Sci ; 95(12): 2657-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16892205

ABSTRACT

GW844520 is a potent and selective inhibitor of the cytochrome bc1 complex of mitochondrial electron transport in P. falciparum, the parasite primarily responsible for the mortality associated with malaria worldwide. GW844520 is fully active against the parasite including resistance isolates, showing no cross resistance with agents in use. To evaluate full potential of this development candidate, we conducted drug metabolism and pharmacokinetic studies of this novel anti-malarial. GW844520 had low blood clearance of about 0.5-4% of hepatic blood flow and a steady-state volume of distribution of 2-4 times total body water in mouse, rat, dog, and monkey. Oral bioavailability was high (51-100%). Consistent with the in vivo data, GW844520 had low intrinsic clearance in liver microsomes and hepatocytes of animal and human origin, high passive cellular permeability and was not a P-glycoprotein substrate. GW844520 did not associate appreciably with blood cells but was highly bound to plasma proteins (>99%) in all species. GW844520 was a substrate and inhibitor of human CYP2D6 but not of CYP1A2, 2C9, 2C19, and 3A4. This conjunctive analysis supports continued evaluation of this compound in definitive pre-IND studies and exemplifies our strategy supporting the discovery of novel agents to treat diseases of the developing world.


Subject(s)
Antimalarials/pharmacokinetics , Pyridones/pharmacokinetics , Animals , Antimalarials/blood , Biological Transport , Biotransformation , Cell Line , Cell Membrane Permeability , Cytochrome P-450 Enzyme Inhibitors , Dogs , Drug Evaluation, Preclinical , Electron Transport Complex III/antagonists & inhibitors , Female , Hepatocytes/metabolism , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Microsomes, Liver/metabolism , Protein Binding , Pyridones/blood , Rats , Rats, Sprague-Dawley
17.
Clin Pharmacol Ther ; 71(4): 235-45, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11956506

ABSTRACT

BACKGROUND: SB 249417 is a humanized anti-factor IX/IXa antibody that, on administration to rats and monkeys, produces an immediate suppression of factor IX activity and prolongation of activated partial thromboplastin times (aPTT). OBJECTIVE: Our objective was to establish the pharmacokinetics of SB 249417 and to explore its effects on factor IX activity levels and aPTT in humans. METHODS: In this phase I, single-blind, randomized, placebo-controlled, parallel-group, single intravenous infusion study, individual and mean data from a total of 26 healthy volunteers at 5 dosing levels were analyzed. A 2-compartment pharmacokinetic model was used in the analysis of total SB 249417 concentration-time profiles. A modified indirect-response model was used, with the total concentration indirectly serving as the driving force for the suppression of free factor IX concentration (as assessed by factor IX activity). The aPTT was related to factor IX activity with a biexponential equation, and a population approach was used to generate posterior parameter estimates for the individual fittings. RESULTS: Mean parameter estimates from individual fittings are 0.092 L/kg for volume of distribution, 0.15 L/kg for steady-state volume of distribution, and 0.0021 L/kg per hour for systemic clearance. The model described well the factor IX activity and aPTT time course in response to SB 249417 at 5 dose levels. The estimated half-life of factor IX in blood was 21 hours. CONCLUSIONS: This model was stable and robust in fitting both mean and individual data. Endogenous factor IX baseline levels and dose were the major determinants of the decline in factor IX activity during the infusion period. Thereafter the recovery of factor IX activity was governed solely by the endogenous factor IX turnover rate.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Factor IX/immunology , Factor IX/metabolism , Models, Biological , Adult , Antibodies, Monoclonal/blood , Binding Sites, Antibody , Dose-Response Relationship, Immunologic , Factor IX/antagonists & inhibitors , Humans , Infusions, Intravenous , Middle Aged , Models, Chemical , Partial Thromboplastin Time , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Single-Blind Method
18.
J Rheumatol ; 29(2): 220-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11838838

ABSTRACT

OBJECTIVE: Double blind studies were conducted with the anti-CD4 monoclonal antibody (Mab) keliximab in patients with active, stable rheumatoid arthritis (RA), to confirm preliminary evidence of efficacy and safety from open. uncontrolled studies. METHODS: We enrolled 136 and 186 patients into 2 consecutive, randomized, double blind trials, with similar populations [apart from inclusion of disease modifying antirheumatic drug (DMARD)-naïve patients in Study 2]. Patients received 4 weeks intravenous placebo or keliximab [40, 80, 120, or 140 mg twice weekly (bw), or 240 mg once weekly (ow)]. The primary endpoint was the American College of Rheumatology (ACR) 20 response criteria, one week after the end of treatment. RESULTS: ACR 20 response rates in Study I were 19%, 42%, 51%*, and 69%* (*p < 0.05 compared to placebo), with placebo, 40, 80, or 140 mg keliximab bw, respectively. The response rates in Study 2 were 30%, 39%, 46% and 47% with placebo, 80 or 120 mg bw, or 240 mg keliximab ow, respectively. In the 2 studies, there was a dose dependent increase in peripheral blood CD4+ T cell coating with keliximab, but a different pattern of CD4 depletion was seen. While only 12% of keliximab treated patients in Study I had CD4 counts below 250 cells/mm3 at the end of the treatment period, 47% fell below this level in Study 2. Clinical response was not correlated with CD4 depletion, but was correlated with CD4+ T cell coating with keliximab. CONCLUSION: Coating of peripheral blood CD4+ T cells with keliximab, but not CD4 depletion, is a determinant of clinical response.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/therapy , CD4 Antigens/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Double-Blind Method , Female , Health Status , Humans , Joints/drug effects , Joints/physiopathology , Male , Middle Aged , Severity of Illness Index , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...