Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Drug Metab Dispos ; 47(10): 1111-1121, 2019 10.
Article in English | MEDLINE | ID: mdl-31387871

ABSTRACT

The identification of nonopioid alternatives to treat chronic pain has received a great deal of interest in recent years. Recently, the engineering of a series of Nav1.7 inhibitory peptide-antibody conjugates has been reported, and herein, the preclinical efforts to identify novel approaches to characterize the pharmacokinetic properties of the peptide conjugates are described. A cryopreserved plated mouse hepatocyte assay was designed to measure the depletion of the peptide-antibody conjugates from the media, with a correlation being observed between percentage remaining in the media and in vivo clearance (Pearson r = -0.5525). Physicochemical (charge and hydrophobicity), receptor-binding [neonatal Fc receptor (FcRn)], and in vivo pharmacokinetic data were generated and compared with the results from our in vitro hepatocyte assay, which was hypothesized to encompass all of the aforementioned properties. Correlations were observed among hydrophobicity; FcRn binding; depletion rates from the hepatocyte assay; and ultimately, in vivo clearance. Subsequent studies identified potential roles for the low-density lipoprotein and mannose/galactose receptors in the association of the Nav1.7 peptide conjugates with mouse hepatocytes, although in vivo studies suggested that FcRn was still the primary receptor involved in determining the pharmacokinetics of the peptide conjugates. Ultimately, the use of the cryopreserved hepatocyte assay along with FcRn binding and hydrophobic interaction chromatography provided an efficient and integrated approach to rapidly triage molecules for advancement while reducing the number of in vivo pharmacokinetic studies. SIGNIFICANCE STATEMENT: Although multiple in vitro and in silico tools are available in small-molecule drug discovery, pharmacokinetic characterization of protein therapeutics is still highly dependent upon the use of in vivo studies in preclinical species. The current work demonstrates the combined use of cryopreserved hepatocytes, hydrophobic interaction chromatography, and neonatal Fc receptor binding to characterize a series of Nav1.7 peptide-antibody conjugates prior to conducting in vivo studies, thus providing a means to rapidly evaluate novel protein therapeutic platforms while concomitantly reducing the number of in vivo studies conducted in preclinical species.


Subject(s)
Chronic Pain/drug therapy , Histocompatibility Antigens Class I/metabolism , Immunoconjugates/pharmacokinetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Receptors, Fc/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Administration, Intravenous , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Cryopreservation , Drug Evaluation, Preclinical/methods , Hepatocytes , Histocompatibility Antigens Class I/genetics , Immunoconjugates/administration & dosage , Macaca fascicularis , Male , Metabolic Clearance Rate , Mice , Mice, Knockout , Peptides/administration & dosage , Peptides/pharmacokinetics , Receptors, Fc/genetics , Tissue Distribution , Voltage-Gated Sodium Channel Blockers/administration & dosage
2.
Eur J Med Chem ; 137: 63-75, 2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28575722

ABSTRACT

Glycine receptors (GlyRs) are pentameric glycine-gated chloride ion channels that are enriched in the brainstem and spinal cord where they have been demonstrated to play a role in central nervous system (CNS) inhibition. Herein we describe two novel classes of glycine receptor potentiators that have been developed using similarity- and property-guided scaffold hopping enabled by parallel synthesis and pharmacophore-based virtual screening strategies. This effort resulted in the identification of novel, efficient and modular leads having favorable in vitro ADME profiles and high CNS multi-parameter optimization (MPO) scores, exemplified by azetidine sulfonamide 19 and aminothiazole sulfone (ent2)-20.


Subject(s)
Drug Discovery , Receptors, Glycine/antagonists & inhibitors , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
3.
J Med Chem ; 60(14): 5990-6017, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28324649

ABSTRACT

Because of its strong genetic validation, NaV1.7 has attracted significant interest as a target for the treatment of pain. We have previously reported on a number of structurally distinct bicyclic heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. Herein, we report the discovery and optimization of a series of atropisomeric quinolinone sulfonamide inhibitors [ Bicyclic sulfonamide compounds as sodium channel inhibitors and their preparation . WO 2014201206, 2014 ] of NaV1.7, which demonstrate nanomolar inhibition of NaV1.7 and exhibit high levels of selectivity over other sodium channel isoforms. After optimization of metabolic and pharmacokinetic properties, including PXR activation, CYP2C9 inhibition, and CYP3A4 TDI, several compounds were advanced into in vivo target engagement and efficacy models. When tested in mice, compound 39 (AM-0466) demonstrated robust pharmacodynamic activity in a NaV1.7-dependent model of histamine-induced pruritus (itch) and additionally in a capsaicin-induced nociception model of pain without any confounding effect in open-field activity.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Quinolones/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Capsaicin , Cell Line , Dogs , Histamine , Mice, Inbred C57BL , Molecular Docking Simulation , Pain/chemically induced , Pain/prevention & control , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Pruritus/chemically induced , Pruritus/prevention & control , Quinolones/administration & dosage , Quinolones/chemical synthesis , Quinolones/pharmacokinetics , Quinolones/pharmacology , Rats , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology
4.
J Med Chem ; 59(17): 7818-39, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27441383

ABSTRACT

The majority of potent and selective hNaV1.7 inhibitors possess common pharmacophoric features that include a heteroaryl sulfonamide headgroup and a lipophilic aromatic tail group. Recently, reports of similar aromatic tail groups in combination with an acyl sulfonamide headgroup have emerged, with the acyl sulfonamide bestowing levels of selectivity over hNaV1.5 comparable to the heteroaryl sulfonamide. Beginning with commercially available carboxylic acids that met selected pharmacophoric requirements in the lipophilic tail, a parallel synthetic approach was applied to rapidly generate the derived acyl sulfonamides. A biaryl acyl sulfonamide hit from this library was elaborated, optimizing for potency and selectivity with attention to physicochemical properties. The resulting novel leads are potent, ligand and lipophilic efficient, and selective over hNaV1.5. Representative lead 36 demonstrates selectivity over other human NaV isoforms and good pharmacokinetics in rodents. The biaryl acyl sulfonamides reported herein may also offer ADME advantages over known heteroaryl sulfonamide inhibitors.


Subject(s)
Benzamides/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Animals , Benzamides/chemical synthesis , Benzamides/pharmacokinetics , Benzamides/pharmacology , Cell Line , Female , Histamine , Humans , Male , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Docking Simulation , Pruritus/chemically induced , Pruritus/drug therapy , Radioligand Assay , Rats , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/chemical synthesis , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology
5.
Drug Metab Dispos ; 42(4): 707-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24423753

ABSTRACT

The mammalian target of rapamycin (mTOR) is a protein kinase that shows key involvement in age-related disease and promises to be a target for treatment of cancer. In the present study, the elimination of potent ATP-competitive mTOR inhibitor 3-(6-amino-2-methylpyrimidin-4-yl)-N-(1H-pyrazol-3-yl)imidazo[1,2-b]pyridazin-2-amine (compound 1) is studied in bile duct-cannulated rats, and the metabolism of compound 1 in liver microsomes is compared across species. Compound 1 was shown to undergo extensive N-glucuronidation in bile duct-catheterized rats. N-glucuronides were detected on positions N1 (M2) and N2 (M1) of the pyrazole moiety as well as on the primary amine (M3). All three N-glucuronide metabolites were detected in liver microsomes of the rat, dog, and human, while primary amine glucuronidation was not detected in cynomolgus monkey. In addition, N1- and N2-glucuronidation showed strong species selectivity in vitro, with rat, dog, and human favoring N2-glucuronidation and monkey favoring N1-glucuronide formation. Formation of M1 in monkey liver microsomes also followed sigmoidal kinetics, singling out monkey as unique among the species with regard to compound 1 N-glucuronidation. In this respect, monkeys might not always be the best animal model for N-glucuronidation of uridine diphosphate glucuronosyltransferase (UGT) 1A9 or UGT1A1 substrates in humans. The impact of N-glucuronidation of compound 1 could be more pronounced in higher species such as monkey and human, leading to high clearance in these species. While compound 1 shows promise as a candidate for investigating the impact of pan-mTOR inhibition in vivo, opportunities may exist through medicinal chemistry efforts to reduce metabolic liability with the goal of improving systemic exposure.


Subject(s)
Glucuronides/metabolism , Heterocyclic Compounds, 2-Ring/metabolism , Microsomes, Liver/enzymology , Pyrimidines/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Biotransformation , Chromatography, High Pressure Liquid , Dogs , Female , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Kinetics , Macaca fascicularis , Magnetic Resonance Spectroscopy , Male , Microsomes, Liver/drug effects , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Species Specificity , Tandem Mass Spectrometry
6.
Biol Psychiatry ; 76(3): 213-22, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24090794

ABSTRACT

BACKGROUND: Dynorphin, an endogenous ligand at kappa opioid receptors (KORs), produces depressive-like effects and contributes to addictive behavior in male nonhuman primates and rodents. Although comorbidity of depression and addiction is greater in women than men, the role of KORs in female motivated behavior is unknown. METHODS: In adult Sprague-Dawley rats, we used intracranial self-stimulation to measure effects of the KOR agonist (±)-trans-U-50488 methanesulfonate salt (U-50488) (.0-10.0 mg/kg) on brain stimulation reward in gonadally intact and castrated males and in females at estrous cycle stages associated with low and high estrogen levels. Pharmacokinetic studies of U-50488 in plasma and brain were conducted. Immunohistochemistry was used to identify sex-dependent expression of U-50488-induced c-Fos in brain. RESULTS: U-50488 dose-dependently increased the frequency of stimulation (threshold) required to maintain intracranial self-stimulation responding in male and female rats, a depressive-like effect. However, females were significantly less sensitive than males to the threshold-increasing effects of U-50488, independent of estrous cycle stage in females or gonadectomy in males. Although initial plasma concentrations of U-50488 were higher in females, there were no sex differences in brain concentrations. Sex differences in U-50488-induced c-Fos activation were observed in corticotropin releasing factor-containing neurons of the paraventricular nucleus of the hypothalamus and primarily in non-corticotropin releasing factor-containing neurons of the bed nucleus of the stria terminalis. CONCLUSIONS: These data suggest that the role of KORs in motivated behavior of rats is sex-dependent, which has important ramifications for the study and treatment of mood-related disorders, including depression and drug addiction in people.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Depression/metabolism , Limbic System/metabolism , Neurons/metabolism , Receptors, Opioid, kappa/agonists , Reward , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/blood , Animals , Electric Stimulation , Female , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Self Stimulation/drug effects , Sex Factors
7.
Drug Metab Dispos ; 41(7): 1433-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23649703

ABSTRACT

The impact of inhibitor depletion on the determination of shifted IC50 (IC50 determined after 30 minutes of preincubation with inhibitor) is examined. In addition, IC50-shift data are analyzed using a mechanistic model that incorporates the processes of inhibitor depletion, as well as reversible and time-dependent inhibition. Anomalies such as a smaller-than-expected shift in IC50 and even increases in IC50 with preincubation were explained by the depletion of inhibitor during the preincubation. The IC50-shift assay remains a viable approach to characterizing a wide range of reversible and time-dependent inhibitors. However, as with more traditional time-dependent inactivation methods, it is recommended that IC50-shift experimental data be interpreted with some knowledge of the magnitude of inhibitor depletion. For the most realistic classification of time-dependent inhibitors using IC50-shift methods, shifted IC50 should be calculated using observed inhibitor concentrations at the end of the incubation rather than nominal inhibitor concentrations. Finally, a mechanistic model that includes key processes, such as competitive inhibition, enzyme inactivation, and inhibitor depletion, can be used to describe accurately the observed IC50 and shifted IC50 curves. For compounds showing an IC50 fold shift >1.5 based on the observed inhibitor concentrations, reanalyzing the IC50-shift data using the mechanistic model appeared to allow for reasonable estimation of Ki, KI, and kinact directly from the IC50 shift experiments.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Humans , Inhibitory Concentration 50 , Models, Biological , Nicardipine/pharmacology , Saquinavir/pharmacology , Time Factors
8.
Drug Metab Dispos ; 41(1): 238-47, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23118327

ABSTRACT

Compound 1 [(E)-4-fluoro-N-(6-((4-(2-hydroxypropan-2-yl)piperidin-1-yl)methyl)-1-((1S,4S)-4-(isopropylcarbamoyl)cyclohexyl)-1H-benzo[d]imidazol-2(3H)-ylidene)benzamide], a new, potent, selective anaplastic lymphoma kinase (ALK) inhibitor with potential application for the treatment of cancer, was selected as candidate to advance into efficacy studies in mice. However, the compound underwent mouse-specific enzymatic hydrolysis in plasma to a primary amine product (M1). Subsequent i.v. pharmacokinetics studies in mice showed that compound 1 had high clearance (CL) and a short half-life. Oral dose escalation studies in mice indicated that elimination of compound 1 was saturable, with higher doses achieving sufficient exposures above in vitro IC(50). Chemistry efforts to minimize hydrolysis resulted in the discovery of several analogs that were stable in mouse plasma. Three were taken in vivo into mice and showed decreased CL corresponding to increased in vitro stability in plasma. However, the more stable compounds also showed reduced potency against ALK. Kinetic studies in NADPH-fortified and unfortified microsomes and plasma produced submicromolar K(m) values and could help explain the saturation of elimination observed in vivo. Predictions of CL based on kinetics from hydrolysis and NADPH-dependent pathways produced predicted hepatic CL values of 3.8, 3.0, 1.6, and 1.2 l/h⋅kg for compound 1, compound 2 [(E)-3,5-difluoro-N-(6-((4-(2-hydroxypropan-2-yl)piperidin-1-yl)methyl)-1-((1s,4s)-4-(isopropylcarbamoyl)cyclohexyl)-1H-benzo[d]imidazol-2(3H)-ylidene)benzamide], compound 3 [(E)-3-chloro-5-fluoro-N-(6-((4-(2-hydroxypropan-2-yl)piperidin-1-yl)methyl)-1-((1s,4s)-4-(isopropylcarbamoyl)cyclohexyl)-1H-benzo[d]imidazol-2(3H)-ylidene)benzamide], and compound 4 [(E)-N-(6-((4-(2-hydroxypropan-2-yl)piperidin-1-yl)methyl)-1-((1s,4s)-4-(isopropylcarbamoyl)cyclohexyl)-1H-benzo[d]imidazol-2(3H)-ylidene)-3-(trifluoromethyl)benzamide], respectively. The in vivo observed CLs for compounds 1, 2, 3, and 4 were 5.52, 3.51, 2.14, and 2.66 l/h⋅kg, respectively. These results indicate that in vitro metabolism kinetic data, incorporating contributions from both hydrolysis and NADPH-dependent metabolism, could be used to predict the systemic CL of compounds cleared via hydrolytic pathways provided that the in vitro assays thoroughly investigate the processes, including the contribution of other metabolic pathways and the possibility of saturation kinetics.


Subject(s)
Protein Kinase Inhibitors/pharmacokinetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Area Under Curve , Chromatography, Liquid , Hydrolysis , Inhibitory Concentration 50 , Male , Mice , Protein Kinase Inhibitors/blood , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
9.
BMC Pharmacol ; 12: 5, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22642416

ABSTRACT

BACKGROUND: Nor-BNI, GNTI and JDTic induce κ opioid antagonism that is delayed by hours and can persist for months. Other effects are transient. It has been proposed that these drugs may be slowly absorbed or distributed, and may dissolve in cell membranes, thus slowing elimination and prolonging their effects. Recent evidence suggests, instead, that they induce prolonged desensitization of the κ opioid receptor. METHODS: To evaluate these hypotheses, we measured relevant physicochemical properties of nor-BNI, GNTI and JDTic, and the timecourse of brain and plasma concentrations in mice after intraperitoneal administration (using LC-MS-MS). RESULTS: In each case, plasma levels were maximal within 30 min and declined by >80% within four hours, correlating well with previously reported transient effects. A strong negative correlation was observed between plasma levels and the delayed, prolonged timecourse of κ antagonism. Brain levels of nor-BNI and JDTic peaked within 30 min, but while nor-BNI was largely eliminated within hours, JDTic declined gradually over a week. Brain uptake of GNTI was too low to measure accurately, and higher doses proved lethal. None of the drugs were highly lipophilic, showing high water solubility (> 45 mM) and low distribution into octanol (log D7.4 < 2). Brain homogenate binding was within the range of many shorter-acting drugs (>7% unbound). JDTic showed P-gp-mediated efflux; nor- BNI and GNTI did not, but their low unbound brain uptake suggests efflux by another mechanism. CONCLUSIONS: The negative plasma concentration-effect relationship we observed is difficult to reconcile with simple competitive antagonism, but is consistent with desensitization. The very slow elimination of JDTic from brain is surprising given that it undergoes active efflux, has modest affinity for homogenate, and has a shorter duration of action than nor-BNI under these conditions. We propose that this persistence may result from entrapment in cellular compartments such as lysosomes.


Subject(s)
Guanidines/pharmacokinetics , Morphinans/pharmacokinetics , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacokinetics , Piperidines/pharmacokinetics , Receptors, Opioid, kappa/metabolism , Tetrahydroisoquinolines/pharmacokinetics , Animals , Biological Transport , Brain/metabolism , Guanidines/blood , Hydrophobic and Hydrophilic Interactions , Injections, Intraperitoneal , LLC-PK1 Cells , Male , Mice , Morphinans/blood , Naltrexone/blood , Naltrexone/pharmacokinetics , Narcotic Antagonists/blood , Permeability , Piperidines/blood , Swine , Tetrahydroisoquinolines/blood
10.
Bioorg Med Chem Lett ; 22(12): 4089-93, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22595176

ABSTRACT

Deregulation of the receptor tyrosine kinase c-Met has been implicated in several human cancers and is an attractive target for small molecule drug discovery. Herein, we report the discovery of a structurally diverse series of carbon-linked quinoline triazolopyridinones, which demonstrates nanomolar inhibition of c-Met kinase activity. This novel series of inhibitors exhibits favorable pharmacokinetics as well as potent inhibition of HGF-mediated c-Met phosphorylation in a mouse liver pharmacodynamic model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridones/chemical synthesis , Quinolines/chemical synthesis , Triazoles/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Hepatocyte Growth Factor/metabolism , Humans , Male , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Pyridones/pharmacology , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Triazoles/pharmacology
11.
Xenobiotica ; 42(9): 830-40, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22439758

ABSTRACT

We previously reported that the accuracy of clearance (CL) prediction could be differentiated by permeability. CL was drastically under-predicted by in vitro metabolic intrinsic clearance (CL(int)) for compounds with low permeability (<5 × 10(-6) cm/s). We determined apparent uptake CL(int) by measuring initial disappearance from medium using attached rat hepatocytes and metabolic CL(int) by measuring parent depletion in suspended rat hepatocytes (cells and medium). Uptake and metabolic CL(int) were comparable for highly permeable metabolic marker compounds. In contrast, uptake CL(int) was 3- to 40-fold higher than metabolic CL(int) for rosuvastatin, bosentan, and 15 proprietary compounds, which had low permeability, suggesting that uptake could be a rate-determining step in hepatic elimination for these poorly permeable compounds. The prediction of hepatic CL was improved significantly when using uptake CL(int) for the compounds with low permeability. The average fold error was 2.2 and 6, as opposed to >11 and >47 by metabolic CL(int), with and without applying a scaling factor of 4, respectively. Uptake CL(int) from attached hepatocytes can be used as an alternative approach to predict hepatic clearance and to understand the significance of hepatic uptake in elimination in an early drug discovery setting.


Subject(s)
Capillary Permeability/physiology , Hepatocytes/physiology , Metabolic Clearance Rate/physiology , Xenobiotics/pharmacokinetics , Animals , Bosentan , Chromatography, Liquid , Cyclosporine/pharmacology , Dogs , Fluorobenzenes/pharmacokinetics , Madin Darby Canine Kidney Cells , Male , Pyrimidines/pharmacokinetics , Quinidine/pharmacology , Rats , Rats, Sprague-Dawley , Rifampin/pharmacology , Rosuvastatin Calcium , Sulfonamides/pharmacokinetics , Tandem Mass Spectrometry , Verapamil/pharmacokinetics , Xenobiotics/metabolism
12.
Drug Metab Lett ; 5(4): 290-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22022868

ABSTRACT

AMG 900 is an orally available small molecule that is highly potent and selective as a pan-aurora kinase inhibitor. AMG 900 is currently undergoing phase 1 clinical evaluation in patients with advanced solid tumors. The metabolism of AMG 900 was investigated in both male and female rats. We conducted studies in bile-duct catheterized (BDC) rats where bile, urine and plasma were analyzed to obtain metabolism profiles for each gender. These studies identified gender differences in the metabolism profiles in bile. Bile contained the majority of the drug related material and contained little unchanged AMG 900 which indicated that metabolism was the prominent process in drug elimination. Although bile contained the same metabolites for both genders, the amount of specific metabolites differed. Male rats metabolized AMG 900 primarily through hydroxylation with subsequent sulfate conjugation on the pyrimidinyl-pyridine side-chain whereas female rats favored a different oxidation site on the thiophene ring's methyl group, which is then metabolized to a carboxylic acid with subsequent conjugation to an acyl glucuronide. CYP phenotyping identified the prominent isoforms as being gender specific or biased in the oxidative metabolism of AMG 900. The metabolism in male rats favored both CYP2C11 and CYP2A2 whereas females favored the CYP2C12. The prominent sulfate conjugate identified in the male rat bile could also be due to male biased metabolism since it has been reported that sulfate conjugation is more prevalent in male rats. All the prominent rat metabolism routes for AMG 900 either have male or female bias. These differences in the rat AMG 900 metabolism profiles in bile can be explained by gender specific P450CYP isoforms.


Subject(s)
Phthalazines/administration & dosage , Phthalazines/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Aurora Kinases , Bile/metabolism , Biotransformation , Cytochrome P450 Family 2 , Female , Glucuronides/metabolism , Hydroxylation , Male , Molecular Structure , Phthalazines/blood , Phthalazines/chemistry , Phthalazines/urine , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/urine , Rats , Rats, Sprague-Dawley , Sex Factors , Steroid 16-alpha-Hydroxylase/metabolism , Steroid Hydroxylases/metabolism , Sulfates/metabolism
13.
Drug Metab Dispos ; 39(11): 2103-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21852367

ABSTRACT

Prediction of human volume of distribution at steady state (V(ss)) before first administration of a new drug candidate to humans has become an important part of the drug development process. This study examines the assumptions behind interspecies scaling techniques used to predict human V(ss) from preclinical data, namely the equivalency of V(ss,u) and/or f(ut) across species. In addition, several interspecies scaling techniques are evaluated side by side using a set of 67 reference compounds where observed V(ss) from rats, dogs, monkeys, and humans were compiled from the literature and where plasma protein binding was determined across species using an ultracentrifugation technique. Species similarity in V(ss,u) or f(ut) does not appear to be the norm among rats, dogs, monkeys, or humans. Despite this, interspecies scaling from rats, dogs, and monkeys is useful and can provide reasonably accurate predictions of human V(ss), although some interspecies scaling approaches were better than others. For example, the performance of the common V(ss,u) or f(ut) equivalency approaches using average V(ss,u) or f(ut) across three preclinical species was superior to allometric scaling techniques. In addition, considering data from several preclinical species, using the equivalency approach, was superior to scaling from any single species. Although the mechanistic tissue composition equations available in the Simcyp population-based pharmacokinetic simulator did not necessarily provide the most accurate predictions, and the equations used likely need refinement, they still provide the best opportunity for a mechanistic understanding and prediction of human V(ss).


Subject(s)
Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Dogs , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Haplorhini , Humans , Protein Binding , Rats , Species Specificity , Tissue Distribution
14.
J Med Chem ; 54(13): 4427-45, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21634377

ABSTRACT

Clinical human genetic studies have recently identified the tetrodotoxin (TTX) sensitive neuronal voltage gated sodium channel Nav1.7 (SCN9A) as a critical mediator of pain sensitization. Herein, we report structure-activity relationships for a novel series of 2,4-diaminotriazines that inhibit hNav1.7. Optimization efforts culminated in compound 52, which demonstrated pharmacokinetic properties appropriate for in vivo testing in rats. The binding site of compound 52 on Nav1.7 was determined to be distinct from that of local anesthetics. Compound 52 inhibited tetrodotoxin-sensitive sodium channels recorded from rat sensory neurons and exhibited modest selectivity against the hERG potassium channel and against cloned and native tetrodotoxin-resistant sodium channels. Upon oral administration to rats, compound 52 produced dose- and exposure-dependent efficacy in the formalin model of pain.


Subject(s)
Acetamides/chemical synthesis , Analgesics/chemical synthesis , Nerve Tissue Proteins/antagonists & inhibitors , Pain/drug therapy , Sodium Channel Blockers/chemical synthesis , Triazines/chemical synthesis , Acetamides/pharmacokinetics , Acetamides/pharmacology , Administration, Oral , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Binding Sites , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Formaldehyde , Ganglia, Spinal/cytology , Humans , In Vitro Techniques , Microsomes, Liver/metabolism , NAV1.1 Voltage-Gated Sodium Channel , Neurons/drug effects , Neurons/physiology , Pain Measurement , Patch-Clamp Techniques , Rats , Sodium Channel Blockers/pharmacokinetics , Sodium Channel Blockers/pharmacology , Sodium Channels , Solubility , Structure-Activity Relationship , Tetrodotoxin/pharmacology , Triazines/pharmacokinetics , Triazines/pharmacology
15.
Xenobiotica ; 41(5): 400-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21294625

ABSTRACT

AMG 900 is a small molecule being developed as an orally administered, highly potent, and selective pan-aurora kinase inhibitor. The aim of the investigations was to characterize in vitro and in vivo pharmacokinetic (PK) properties of AMG 900 in preclinical species. AMG 900 was rapidly metabolized in liver microsomes and highly bound to plasma proteins in the species tested. It was a weak Pgp substrate with good passive permeability. AMG 900 exhibited a low-to-moderate clearance and a small volume of distribution. Its terminal elimination half-life ranged from 0.6 to 2.4 h. AMG 900 was well-absorbed in fasted animals with an oral bioavailability of 31% to 107%. Food intake had an effect on rate (rats) or extent (dogs) of AMG 900 oral absorption. The clearance and volume of distribution at steady state in humans were predicted to be 27.3 mL/h/kg and 93.9 mL/kg, respectively. AMG 900 exhibited acceptable PK properties in preclinical species and was predicted to have low clearance in humans. AMG 900 is currently in Phase I clinical testing as a treatment for solid tumours. Preliminary human PK results appear to be consistent with the predictions.


Subject(s)
Phthalazines/pharmacology , Phthalazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Small Molecule Libraries/pharmacokinetics , Administration, Oral , Animals , Aurora Kinases , Biological Availability , Blood Proteins/metabolism , Cell Line , Fasting , Humans , Injections, Intravenous , Male , Phthalazines/blood , Phthalazines/chemistry , Protein Binding/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Species Specificity
16.
Drug Metab Dispos ; 38(1): 115-21, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19797608

ABSTRACT

To predict volume of distribution at steady-state (V(ss)), empirical (e.g., allometry) and mechanistic (using physicochemical property data and plasma protein binding) methods have been used. None of these approaches has been able to predict V(ss) accurately for the total compliment of a wide range of drugs. Therefore, alternative approaches would be of value. This study evaluates the utility of in vitro nonspecific tissue-binding measurements in predicting V(ss) for a wide range of drugs in rats. Literature as well as proprietary compounds were studied. It was found that in vitro tissue-binding measurements combined with calculated effects of the pH partition hypothesis often predict V(ss) more accurately than other available mechanistic methods and that this approach can compliment existing methods. The V(ss) values for some compounds were not accurately predicted using either nonspecific tissue-binding experiments or other available mechanistic methods. The V(ss) for these drugs may not be describable by nonspecific tissue binding alone; there may be significant specific components to the mechanism of distribution for these drugs, such as pH-dependent uptake into lysosomes (primarily strongly basic drugs), active transport, and/or enterohepatic recirculation. A lack of prediction for certain drugs warrants further investigation into these mechanisms and their application to more accurate prediction of V(ss) by mechanistic means.


Subject(s)
Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Tissue Extracts/metabolism , Algorithms , Animals , Chemical Phenomena , Hydrogen-Ion Concentration , Male , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Plasma/metabolism , Rats , Rats, Sprague-Dawley
17.
Drug Metab Dispos ; 38(2): 223-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19875499

ABSTRACT

In vitro intrinsic metabolic clearance (CL(int)) is used routinely for compound selection in drug discovery; however, in vitro CL(int) often underpredicts in vivo clearance (CL). Forty-one proprietary compounds and 16 marketed drugs were selected to determine whether permeability and efflux status could influence the predictability of CL from in vitro CL(int) obtained from liver microsomal and hepatocyte incubations. For many of the proprietary compounds examined, rat CL was significantly underpredicted using the well stirred model incorporating both fraction of unbound drug in blood and fraction of unbound drug in the microsomal or hepatocyte incubation. Further analysis revealed that the accuracy of the prediction was differentiated by permeability and P-glycoprotein- (P-gp) and mouse breast cancer resistance protein (mBcrp)-mediated efflux. For proprietary compounds with passive permeability greater than 5 x 10(-6) cm/s and efflux ratios less than 5 in both P-gp- and mBcrp-expressing cells, CL(int) provided reasonable prediction. The average -fold error (AFE) was 1.8 for rat liver microsomes (RLMs) and 2.3 for rat hepatocytes. In contrast, CL was dramatically underpredicted for compounds with passive permeability less than 5 x 10(-6) cm/s; AFEs of 54.4 and 29.2 were observed for RLM and rat hepatocytes, respectively. In vivo CL was also underpredicted for compounds that were good efflux substrates (permeability >5 x 10(-6) cm/s). The AFEs were 7.4 and 8.1 for RLM and rat hepatocytes, respectively. A similar relationship between permeability, efflux status, and human CL prediction reported in the literature was observed for 16 marketed drugs. These data show that permeability and efflux status are determinants for the predictability of CL from in vitro metabolic CL(int).


Subject(s)
Cell Membrane Permeability , Metabolic Clearance Rate , Pharmaceutical Preparations/metabolism , Pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Algorithms , Animals , Biological Transport , Cell Line , Chemical Phenomena , Dogs , Genes, MDR , Hepatocytes/metabolism , Humans , Male , Mice , Microsomes, Liver/metabolism , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/classification , Rats , Rats, Sprague-Dawley , Sus scrofa
18.
Bioorg Med Chem Lett ; 19(22): 6307-12, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19819693

ABSTRACT

Deregulation of the receptor tyrosine kinase c-Met has been implicated in several human cancers and is an attractive target for small molecule drug discovery. We previously showed that O-linked triazolopyridazines can be potent inhibitors of c-Met. Herein, we report the discovery of a related series of N-linked triazolopyridazines which demonstrate nanomolar inhibition of c-Met kinase activity and display improved pharmacodynamic profiles. Specifically, the potent time-dependent inhibition of cytochrome P450 associated with the O-linked triazolopyridazines has been eliminated within this novel series of inhibitors. N-linked triazolopyridazine 24 exhibited favorable pharmacokinetics and displayed potent inhibition of HGF-mediated c-Met phosphorylation in a mouse liver PD model. Once-daily oral administration of 24 for 22days showed significant tumor growth inhibition in an NIH-3T3/TPR-Met xenograft mouse efficacy model.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/physiology , Neovascularization, Physiologic/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Cell Survival , Humans , Mice , Mice, Nude , Phosphorylation , Xenograft Model Antitumor Assays
19.
Drug Metab Lett ; 3(2): 70-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19601867

ABSTRACT

Species and tissue differences in the activity of three major classes of esterases, carboxylesterase (CE), butyrylcholinesterase (BChE) and paraoxonase (PON), were studied. Substantial species differences in activity of these esterases were observed between the mouse, rat, dog monkey and human. Such species differences must be considered when using these preclinical species to optimize the pharmacokinetic properties of ester compounds intended for human use.


Subject(s)
Aryldialkylphosphatase/metabolism , Butyrylcholinesterase/metabolism , Carboxylesterase/metabolism , Animals , Dogs , Drug Design , Drug Evaluation, Preclinical/methods , Humans , Macaca fascicularis , Mice , Models, Animal , Rats , Species Specificity
20.
J Med Chem ; 51(13): 3688-91, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18553959

ABSTRACT

Deregulation of the receptor tyrosine kinase c-Met has been implicated in human cancers. Pyrazolones with N-1 bearing a pendent hydroxyalkyl side chain showed selective inhibition of c-Met over VEGFR2. However, studies revealed the generation of active, nonselective metabolites. Blocking this metabolic hot spot led to the discovery of 17 (AMG 458). When dosed orally, 17 significantly inhibited tumor growth in the NIH3T3/TPR-Met and U-87 MG xenograft models with no adverse effect on body weight.


Subject(s)
Aminopyridines/administration & dosage , Aminopyridines/chemistry , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Administration, Oral , Aminopyridines/chemical synthesis , Aminopyridines/pharmacokinetics , Animals , Cell Survival/drug effects , Cells, Cultured , Drug Design , Humans , Mice , Mice, Inbred BALB C , Molecular Structure , Mutation/genetics , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...