Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 240
Filter
1.
Appl Biochem Biotechnol ; 195(9): 5338-5354, 2023 Sep.
Article in English | MEDLINE | ID: mdl-35195835

ABSTRACT

In the present paper, several computational binding analyses were performed on ethyl 3,3,5,5-tetracyano-2-hydroxy-2-methyl-4,6-diphenylcyclohexane-1-carboxylate which was newly synthesized by three-component condensation of benzaldehyde with ethyl acetoacetate and malononitrile in the presence of trichloroacetic acid, and the structure was finally proved by X-ray analysis. The visualization of molecular interaction was carried out through Hirshfeld surface analysis and ESP. The atomic charges, HOMO, LUMO, and electrostatic potential were also studied to explore the insight of the molecule deeper, and then, natural bonding orbitals (NBO) and non-linear optical properties (NLO) were calculated to reveal the interactions that happen to be between the filled and vacant orbitals. Afterwards, molecular docking studies predicted the compound binding mode fits in the minor groove of DNA and remained interacts via stable bonding as validated by molecular dynamics simulations. The binding energy estimation also affirmed domination van der Waals and electrostatic energies. Lastly, the compound was found as good drug-like molecule and had good pharmacokinetic profile with exception of toxic moieties.


Subject(s)
Cyclohexanes , DNA , Cyclohexanes/chemical synthesis , Cyclohexanes/chemistry , Cyclohexanes/pharmacokinetics , DNA/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Reproducibility of Results , Static Electricity , Thermodynamics , Trichloroacetic Acid/chemistry
2.
J Med Chem ; 62(13): 6190-6213, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31259550

ABSTRACT

Recruitment of suppressive CD4+ FOXP3+ regulatory T cells (Treg) to the tumor microenvironment (TME) has the potential to weaken the antitumor response in patients receiving treatment with immuno-oncology (IO) agents. Human Treg express CCR4 and can be recruited to the TME through the CC chemokine ligands CCL17 and CCL22. In some cancers, Treg accumulation correlates with poor patient prognosis. Preclinical data suggests that preventing the recruitment of Treg and increasing the population of activated effector T cells (Teff) in the TME can potentiate antitumor immune responses. We developed a novel series of potent, orally bioavailable small molecule antagonists of CCR4. From this series, several compounds exhibited high potency in distinct functional assays in addition to good in vitro and in vivo ADME properties. The design, synthesis, and SAR of this series and confirmation of its in vivo activity are reported.


Subject(s)
Cell Movement/drug effects , Pyrazines/pharmacology , Pyrazoles/pharmacology , Receptors, CCR4/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/drug effects , Animals , Cyclohexanes/chemical synthesis , Cyclohexanes/pharmacokinetics , Cyclohexanes/pharmacology , Drug Discovery , Humans , Mice, Transgenic , Molecular Structure , Piperazines/chemical synthesis , Piperazines/pharmacokinetics , Piperazines/pharmacology , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Rats , Structure-Activity Relationship
3.
Bioorg Med Chem Lett ; 29(16): 2100-2106, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31288965

ABSTRACT

We describe here a novel GPR119 agonist 24, which showed a potent and long-acting hypoglycemic effect in rats via oral dosing. For the discovery of 24, we chose compound 5, which possessed an oxadiazole linker, as a lead compound among our spirocyclic cyclohexane GPR119 agonist series, taking into account its lower plasma protein binding nature. 3,5-Difluoro and 4-methylsulfonylmethy groups on the left side phenyl group, and a gem-difluoro group on the right side of 24 are important for its agonist potency and metabolic stability, respectively.


Subject(s)
Cyclohexanes/pharmacology , Hypoglycemic Agents/pharmacology , Oxadiazoles/pharmacology , Receptors, G-Protein-Coupled/agonists , Spiro Compounds/pharmacology , Animals , Cyclohexanes/chemical synthesis , Cyclohexanes/pharmacokinetics , Drug Stability , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Microsomes, Liver/metabolism , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Rats, Sprague-Dawley , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacokinetics , Structure-Activity Relationship
4.
J Pharm Sci ; 108(6): 1934-1943, 2019 06.
Article in English | MEDLINE | ID: mdl-30639736

ABSTRACT

Solubilization of new chemical entities for toxicity assessment must use excipients that do not negatively impact drug pharmacokinetics and toxicology. In this study, we investigated the tolerability of a model freebase compound, GDC-0152, solubilized by pH adjustment with succinic acid and complexation with hydroxypropyl-ß-cyclodextrin (HP-ß-CD) to enable intravenous use. Solubility, critical micelle concentration, and association constant with HP-ß-CD were determined. Blood compatibility and potential for hemolysis were assessed in vitro. Local tolerability was assessed after intravenous and subcutaneous injections in rats. A pharmacokinetic study was conducted in rats after intravenous bolus administration. GDC-0152 exhibited pH-dependent solubility that was influenced by self-association. The presence of succinic acid increased solubility in a concentration-dependent manner. HP-ß-CD alone also increased solubility, but the extent of solubility enhancement was significantly lower than succinic acid alone. Inclusion of HP-ß-CD in the solution of GDC-0152 improved blood compatibility, reduced hemolytic potential by ∼20-fold in vitro, and increased the maximum tolerated dose to 80 mg/kg.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacokinetics , Cyclohexanes/toxicity , Drug Evaluation, Preclinical/methods , Excipients/pharmacokinetics , Pyrroles/toxicity , Toxicity Tests, Acute/methods , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Animals , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Excipients/administration & dosage , Hemolysis/drug effects , Injections, Intravenous , Injections, Subcutaneous , Male , Maximum Tolerated Dose , Models, Animal , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Rats , Solubility
5.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29925666

ABSTRACT

Current approaches do not eliminate all human immunodeficiency virus type 1 (HIV-1) maternal-to-infant transmissions (MTIT); new prevention paradigms might help avert new infections. We administered maraviroc (MVC) to rhesus macaques (RMs) to block CCR5-mediated entry, followed by repeated oral exposure of a CCR5-dependent clone of simian immunodeficiency virus (SIV) mac251 (SIVmac766). MVC significantly blocked the CCR5 coreceptor in peripheral blood mononuclear cells and tissue cells. All control animals and 60% of MVC-treated infant RMs became infected by the 6th challenge, with no significant difference between the number of exposures (P = 0.15). At the time of viral exposures, MVC plasma and tissue (including tonsil) concentrations were within the range seen in humans receiving MVC as a therapeutic. Both treated and control RMs were infected with only a single transmitted/founder variant, consistent with the dose of virus typical of HIV-1 infection. The uninfected RMs expressed the lowest levels of CCR5 on the CD4+ T cells. Ramp-up viremia was significantly delayed (P = 0.05) in the MVC-treated RMs, yet peak and postpeak viral loads were similar in treated and control RMs. In conclusion, in spite of apparent effective CCR5 blockade in infant RMs, MVC had a marginal impact on acquisition and only a minimal impact on the postinfection delay of viremia following oral SIV infection. Newly developed, more effective CCR5 blockers may have a more dramatic impact on oral SIV transmission than MVC.IMPORTANCE We have previously suggested that the very low levels of simian immunodeficiency virus (SIV) maternal-to-infant transmissions (MTIT) in African nonhuman primates that are natural hosts of SIVs are due to a low availability of target cells (CCR5+ CD4+ T cells) in the oral mucosa of the infants, rather than maternal and milk factors. To confirm this new MTIT paradigm, we performed a proof-of-concept study in which we therapeutically blocked CCR5 with maraviroc (MVC) and orally exposed MVC-treated and naive infant rhesus macaques to SIV. MVC had only a marginal effect on oral SIV transmission. However, the observation that the infant RMs that remained uninfected at the completion of the study, after 6 repeated viral challenges, had the lowest CCR5 expression on the CD4+ T cells prior to the MVC treatment appears to confirm our hypothesis, also suggesting that the partial effect of MVC is due to a limited efficacy of the drug. New, more effective CCR5 inhibitors may have a better effect in preventing SIV and HIV transmission.


Subject(s)
CCR5 Receptor Antagonists/administration & dosage , Cyclohexanes/administration & dosage , Infectious Disease Transmission, Vertical/prevention & control , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/transmission , Triazoles/administration & dosage , Animals , CCR5 Receptor Antagonists/pharmacokinetics , Cyclohexanes/pharmacokinetics , Humans , Infant , Maraviroc , Palatine Tonsil/chemistry , Serum/chemistry , Treatment Outcome , Triazoles/pharmacokinetics , Viral Load
6.
Int J Radiat Biol ; 94(6): 590-596, 2018 06.
Article in English | MEDLINE | ID: mdl-29659318

ABSTRACT

PURPOSE: Dioxime derivative is reported to exhibit high affinity towards tumor cells. The objective of the present study is to synthesize a new dioxime derivative to be labeled with technetium-99m for using as a solid tumor marker. MATERIALS AND METHODS: ((2E,2',3E,3')-3,3'-(cyclohexane-1,2-diylbis (azanylylidene)) bis-(butan-2-one)dioxime) was synthesized by condensation of Butan-2,3-dione monooxime and diaminocyclohexane and labeled with 99mTc. The in-vivo distribution of the agent was studied by carrying out biodistribution in tumor bearing Albino mice. RESULTS: A new cyclohexane dioxime derivative was synthesized with a good yield of 93 ± 2% and its complexation with 99mTc was prepared with 85 ± 4% radiochemical yield under the optimized conditions and the preparation exhibited in-vitro stability up to 6 h. Biodistribution studies showed high uptake in tumor cells with T/NT (target to non-target ratio) = 3.4 ± 0.2 after 0.5 h post injection. CONCLUSION: As a result of biodistribution studies, the newly synthesized cyclohexane dioxime derivative showed its good uptake in tumor cells, which affords a potential radiopharmaceutical that could be used as a good tumor imaging agent.


Subject(s)
Cyclohexanes/chemical synthesis , Cyclohexanes/pharmacokinetics , Molecular Imaging/methods , Oximes/chemistry , Animals , Cell Line, Tumor , Chemistry Techniques, Synthetic , Cyclohexanes/chemistry , Humans , Isotope Labeling , Mice , Tissue Distribution
7.
J Pharmacol Exp Ther ; 365(2): 301-313, 2018 05.
Article in English | MEDLINE | ID: mdl-29491038

ABSTRACT

Methionine aminopeptidase 2 (MetAP2) inhibition is a promising approach to treating diabetes, obesity, and associated metabolic disorders. Beloranib, a MetAP2 inhibitor previously investigated for treatment of Prader-Willi syndrome, was associated with venous thrombotic adverse events likely resulting from drug effects on vascular endothelial cells (ECs). Here, we report the pharmacological characterization of ZGN-1061, a novel MetAP2 inhibitor being investigated for treatment of diabetes and obesity. Four weeks of subcutaneous administration of ZGN-1061 to diet-induced obese (DIO) insulin-resistant mice produced a 25% reduction in body weight, primarily due to reduced fat mass, that was comparable to beloranib. ZGN-1061 also produced improvements in metabolic parameters, including plasma glucose and insulin, and, in HepG2 cells, initiated gene changes similar to beloranib that support observed in vivo pharmacodynamics. In vitro studies in ECs demonstrated that ZGN-1061 effects on EC proliferation and coagulation proteins were greatly attenuated, or absent, relative to beloranib, due to lower intracellular drug concentrations, shorter half-life of inhibitor-bound MetAP2 complex, and reduced cellular enzyme inhibition. In dogs, ZGN-1061 was more rapidly absorbed and cleared, with a shorter half-life than beloranib. Unlike beloranib, ZGN-1061 did not increase coagulation markers in dogs, and ZGN-1061 had a greatly improved safety profile in rats relative to beloranib. In conclusion, ZGN-1061 and beloranib demonstrated similar efficacy in a mouse model of obesity, while ZGN-1061 had a markedly improved safety profile in multiple in vitro and in vivo models. The lower duration of exposure characteristic of ZGN-1061 is expected to provide a meaningfully enhanced clinical safety profile.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Azetidines/adverse effects , Azetidines/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Morpholines/adverse effects , Morpholines/pharmacology , Obesity/drug therapy , Safety , Animals , Azetidines/pharmacokinetics , Azetidines/therapeutic use , Blood Coagulation/drug effects , Cinnamates/pharmacokinetics , Cinnamates/pharmacology , Cyclohexanes/pharmacokinetics , Cyclohexanes/pharmacology , Dogs , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Epoxy Compounds/pharmacokinetics , Epoxy Compounds/pharmacology , Female , Hep G2 Cells , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Morpholines/pharmacokinetics , Morpholines/therapeutic use , Obesity/enzymology , Rats , Sesquiterpenes/pharmacokinetics , Sesquiterpenes/pharmacology , Tissue Distribution
8.
Brain Struct Funct ; 223(6): 2609-2625, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29550938

ABSTRACT

Serotonin 1A (5-HT1A) receptors mediate serotonin trophic role in brain neurogenesis. Gray matter volume (GMV) loss and 5-HT1A receptor binding alterations have been identified in major depressive disorder (MDD). Here we investigated the relationship between 5-HT1A receptor binding and GMV in 40 healthy controls (HCs) and, for the first time, 47 antidepressant-free MDD patients using Voxel-Based Morphometry and [11C]WAY100635 Positron Emission Tomography. Values of GMV and 5-HT1A binding (expressed as BPF, one of the types of binding potentials that refer to displaceable or specific binding that can be quantified in vivo with PET) were obtained in 13 regions of interest, including raphe, and at the voxel level. We used regression analysis within each group to predict GMV from BPF, while covarying for age, sex, total gray matter volume and medication status. In the HCs group, we found overall a positive correlation between terminal field 5-HT1A receptor binding and GMV, which reached statistical significance in regions such as hippocampus, insula, orbital prefrontal cortex, and parietal lobe. We observed a trend towards inverse correlation between raphe 5-HT1A autoreceptor binding and anterior cingulate GMV in both groups, and a statistically significant positive correlation between raphe 5-HT1A binding and temporal GMV in MDD. Analysis of covariance at the voxel-level revealed a trend towards interaction between diagnosis and raphe 5-HT1A binding in predicting GMV in cerebellum and supramarginal gyrus (higher correlation in HCs compared with MDD). Our results replicated previous findings in the normative brain, but did not extend them to the brain in MDD, and indicated a trend towards dissociation between MDD and HCs in the relationship of raphe 5-HT1A binding with postsynaptic GMV. These results suggest that 5-HT1A receptors contribute to altered neuroplasticity in MDD, possibly via effects predating depression onset.


Subject(s)
Brain/pathology , Depressive Disorder, Major/pathology , Gray Matter/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Adolescent , Adult , Aged , Brain/diagnostic imaging , Brain/drug effects , Brain Mapping , Carbon Isotopes/pharmacokinetics , Cyclohexanes/pharmacokinetics , Female , Gray Matter/diagnostic imaging , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Piperazines/pharmacokinetics , Positron-Emission Tomography , Protein Binding/drug effects , Young Adult
9.
Drug Metab Dispos ; 46(5): 493-502, 2018 May.
Article in English | MEDLINE | ID: mdl-29475834

ABSTRACT

Maraviroc (MVC) is a CCR5 coreceptor antagonist indicated in combination with other antiretroviral agents for the treatment of CCR5-tropic human immunodefinciency virus-1 infection. In this study, the metabolism of MVC was investigated in human liver microsomes to delineate the relative roles of CYP3A4 and CYP3A5. MVC is metabolized to five hydroxylated metabolites, all of which were biosynthesized and identified using mass and NMR spectroscopy. The sites of metabolism were the 2- and 3-positions of the 4,4-difluorocyclohexyl moiety and the methyl of the triazole moiety. Absolute configurations were ultimately ascertained by comparison to authentic standards. The biosynthesized metabolites were used for quantitative in vitro experiments in liver microsomes using cyp3cide, a selective inactivator of CYP3A4. (1S,2S)-2-OH-MVC was the main metabolite representing approximately half of the total metabolism, and CYP3A5 contributed approximately 40% to that pathway in microsomes from CYP3A5*1/*1 donors. The other four metabolites were almost exclusively metabolized by CYP3A4. (1S,2S)-2-hydroxylation also correlated to T-5 N-oxidation, a CYP3A5-specific activity. These data are consistent with clinical pharmacokinetic data wherein CYP3A5 extensive metabolizer subjects showed a modestly lower exposure to MVC.


Subject(s)
Cyclohexanes/metabolism , Cytochrome P-450 CYP3A/metabolism , Triazoles/metabolism , Cyclohexanes/pharmacokinetics , Humans , Hydroxylation/physiology , Kinetics , Maraviroc , Microsomes, Liver/metabolism , Oxidation-Reduction , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics
10.
J Antimicrob Chemother ; 72(11): 3167-3171, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28961979

ABSTRACT

BACKGROUND: Sub-optimal penetration of antiretroviral drugs in genital compartments might promote local HIV persistence and increase the risk of HIV transmission. OBJECTIVES: To describe the penetration of maraviroc, raltegravir, raltegravir glucuronide and rilpivirine in seminal plasma and cervico-vaginal secretions (CVS) and to assess local antiretroviral efficacy in HIV-1-positive patients. METHODS: This was a prospective, multicentre study. Inclusion criteria were HIV-1 positive, age >18 years, receiving regimens containing maraviroc and/or raltegravir and/or rilpivirine for >1 month, and good self-reported adherence. Paired blood and genital samples were collected 12 h (raltegravir and maraviroc) or 24 h (rilpivirine) post-dose. These concentrations were determined (UPLC-MS/MS) in blood and seminal plasma (total and unbound) and CVS (total, dried spots) and HIV-RNA was quantified in paired blood and genital samples. RESULTS: Among the 54 enrolled patients, 15 received maraviroc (6 men), 27 received raltegravir (14 men) and 20 received rilpivirine (10 men), corresponding to 54 total and 52 unbound plasma concentrations, 29 total CVS samples and 23 total and 18 unbound seminal plasma samples. Maraviroc and raltegravir displayed a ratio of genital fluids/plasma concentrations >0.5 in both male and female genital tracts. Conversely, rilpivirine displayed a low ratio. Antiretroviral free fractions were consistent with historical data. Nine patients had blood plasma HIV-RNA >50 copies/mL (2/9 had sub-optimal antiretroviral blood plasma exposure) and two other patients had detectable HIV-RNA in genital fluids. CONCLUSIONS: Maraviroc and raltegravir demonstrated good penetration in genital compartments, yielding good local virological response in genital compartments, whereas rilpivirine presented a low penetration profile but good local response.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Body Fluids/chemistry , HIV Infections/drug therapy , Semen/chemistry , Adult , Anti-HIV Agents/administration & dosage , Cervix Uteri/chemistry , Cervix Uteri/virology , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacokinetics , Cyclohexanes/therapeutic use , Female , HIV Infections/metabolism , HIV Infections/transmission , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Maraviroc , Middle Aged , Prospective Studies , Raltegravir Potassium/administration & dosage , Raltegravir Potassium/pharmacokinetics , Raltegravir Potassium/therapeutic use , Rilpivirine/administration & dosage , Rilpivirine/metabolism , Rilpivirine/pharmacokinetics , Rilpivirine/therapeutic use , Semen/virology , Triazoles/administration & dosage , Triazoles/pharmacokinetics , Triazoles/therapeutic use , Vagina/chemistry , Vagina/virology , Viral Load
11.
J Pharm Pharmacol ; 69(9): 1099-1109, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28590055

ABSTRACT

OBJECTIVES: Nateglinide, an Antidiabetic drug (BCS II), shows pH-dependent solubility and variable bioavailability. The purpose of study was to increase dissolution and bioavailability of Nateglinide by development of its microenvironmental pH-regulated ternary solid dispersion (MeSD). METHODS: MeSD formulation of Nateglinide, poloxamer-188 and Na2 CO3 was prepared by melt dispersion in 1 : 2 : 0.2 w/w ratio and further characterised for solubility, In-vitro dissolution, microenvironmental pH, crystallinity/amorphism, physicochemical interactions, bioavailability in Wistar rats. KEY FINDINGS: Solubility of Nateglinide was increased notably in MeSD, and its in-vitro dissolution study showed fourfold increase in the dissolution, particularly in 1.2 pH buffer. Prominent reduction in the peak intensity of X-ray powder diffraction (XRPD) and absence of endotherm in DSC thermogram confirmed the amorphism of Nateglinide in MeSD. Attenuated total reflectance Fourier transform infrared spectra revealed the hydrogen bond interactions between Nateglinide and poloxamer-188. In-vivo study indicated that MeSD exhibited fourfold increase in area under curve over Nateglinide. Tmax of MeSD was observed at 0.25 h, which is beneficial for efficient management of postprandial sugar. Instead of mere transformation of the Nateglinide to its amorphous form as evidenced by DSC and XRPD, formation of a soluble carboxylate compound of Nateglinide in MeSD was predominantly responsible for dissolution and bioavailability enhancement. CONCLUSIONS: The study demonstrates the utility of MeSD in achieving pH-independent dissolution, reduced Tmax and enhanced bioavailability of Nateglinide.


Subject(s)
Cyclohexanes/administration & dosage , Drug Compounding/methods , Excipients/chemistry , Hypoglycemic Agents/administration & dosage , Phenylalanine/analogs & derivatives , Animals , Biological Availability , Carbonates/chemistry , Chemistry, Pharmaceutical , Cyclohexanes/chemistry , Cyclohexanes/pharmacokinetics , Drug Liberation , Female , Hydrogen-Ion Concentration , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Male , Nateglinide , Phenylalanine/administration & dosage , Phenylalanine/chemistry , Phenylalanine/pharmacokinetics , Poloxamer/chemistry , Rats , Rats, Wistar , Solubility
12.
Neuropharmacology ; 115: 100-114, 2017 03 15.
Article in English | MEDLINE | ID: mdl-26748052

ABSTRACT

Metabotropic glutamate 2/3 (mGlu2/3) receptors are of considerable interest owing to their role in modulating glutamate transmission via presynaptic, postsynaptic and glial mechanisms. As part of our ongoing efforts to identify novel ligands for these receptors, we have discovered (1S,2R,3S,4S,5R,6R)-2-amino-3-[(3,4-difluorophenyl)sulfanylmethyl]-4-hydroxy-bicyclo[3.1.0]hexane-2,6-dicarboxylic acid; (LY3020371), a potent and selective orthosteric mGlu2/3 receptor antagonist. In this account, we characterize the effects of LY3020371 in membranes and cells expressing human recombinant mGlu receptor subtypes as well as in native rodent and human brain tissue preparations, providing important translational information for this molecule. In membranes from cells expressing recombinant human mGlu2 and mGlu3 receptor subtypes, LY3020371.HCl competitively displaced binding of the mGlu2/3 agonist ligand [3H]-459477 with high affinity (hmGlu2 Ki = 5.26 nM; hmGlu3 Ki = 2.50 nM). In cells expressing hmGlu2 receptors, LY3020371.HCl potently blocked mGlu2/3 agonist (DCG-IV)-inhibited, forskolin-stimulated cAMP formation (IC50 = 16.2 nM), an effect that was similarly observed in hmGlu3-expressing cells (IC50 = 6.21 nM). Evaluation of LY3020371 in cells expressing the other human mGlu receptor subtypes revealed high mGlu2/3 receptor selectivity. In rat native tissue assays, LY3020371 demonstrated effective displacement of [3H]-459477 from frontal cortical membranes (Ki = 33 nM), and functional antagonist activity in cortical synaptosomes measuring both the reversal of agonist-suppressed second messenger production (IC50 = 29 nM) and agonist-inhibited, K+-evoked glutamate release (IC50 = 86 nM). Antagonism was fully recapitulated in both primary cultured cortical neurons where LY3020371 blocked agonist-suppressed spontaneous Ca2+ oscillations (IC50 = 34 nM) and in an intact hippocampal slice preparation (IC50 = 46 nM). Functional antagonist activity was similarly demonstrated in synaptosomes prepared from epileptic human cortical or hippocampal tissues, suggesting a translation of the mGlu2/3 antagonist pharmacology from rat to human. Intravenous dosing of LY3020371 in rats led to cerebrospinal fluid drug levels that are expected to effectively block mGlu2/3 receptors in vivo. Taken together, these results establish LY3020371 as an important new pharmacological tool for studying mGlu2/3 receptors in vitro and in vivo. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.


Subject(s)
Cyclohexanes/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Animals , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclohexanes/chemistry , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/chemistry , Humans , Male , Rats , Rats, Sprague-Dawley
13.
J Acquir Immune Defic Syndr ; 73(3): 252-257, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27727157

ABSTRACT

To investigate the pharmacokinetics/pharmacodynamics of single-dose maraviroc 300 mg in HIV-1 exposure compartments. Maraviroc concentrations in blood, secretions (vaginal, urethral, oral, and rectal), and tissue (vaginal and rectal) were measured, and ex vivo challenge was performed in 54 healthy volunteers to study protection from HIV infection. Maraviroc Cmax occurred within 4 hours in most compartments. Concentrations from 4 to 72 hours were above intracellular (IC) IC90 in all compartments, range 15-8095 ng/mL. Mean AUC0-72 compartment-to-plasma ratios were highest in the rectum (45-819) and urethra (144) compared with the female genital tract (1.6-4.8) and saliva (0.2). No sex differences in AUC0-72 or Cmax were observed. No ex vivo protection from HIV-1BaL occurred in rectal or vaginal tissue. Despite high and sustained concentrations, single-dose maraviroc was not protective against ex vivo challenge of vaginal/rectal tissue.


Subject(s)
CCR5 Receptor Antagonists/pharmacokinetics , Cyclohexanes/pharmacokinetics , HIV Fusion Inhibitors/pharmacokinetics , HIV Infections/prevention & control , HIV Infections/virology , Pre-Exposure Prophylaxis , Triazoles/pharmacokinetics , Administration, Oral , Adult , CCR5 Receptor Antagonists/administration & dosage , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacology , Female , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Healthy Volunteers , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/virology , Male , Maraviroc , Models, Biological , Randomized Controlled Trials as Topic , Rectum/drug effects , Rectum/virology , Saliva/drug effects , Saliva/virology , Treatment Outcome , Triazoles/administration & dosage , Triazoles/pharmacology , Urethra/drug effects , Urethra/virology , Vagina/drug effects , Vagina/virology
14.
Biol Blood Marrow Transplant ; 22(10): 1829-1835, 2016 10.
Article in English | MEDLINE | ID: mdl-27498124

ABSTRACT

Maraviroc is an allosteric small molecule antagonist of chemokine receptor type 5 (CCR5) and has been used in adult allogeneic hematopoietic stem cell transplant (HSCT) recipients to prevent acute graft-versus-host disease (GVHD) of the gastrointestinal (GI) tract and liver. The goal of this study was to establish feasibility and pharmacokinetic and pharmacodynamic profiles of maraviroc in pediatric HSCT recipients. Children ages 2 to 12 years were enrolled and maraviroc was added to standard GVHD prophylaxis, which included a calcineurin inhibitor and either steroids or mycophenolate mofetil. Maraviroc was started on day -3 and administered at a dose of approximately 300 mg/m(2) orally twice daily until day +30 after stem cell infusion. On days 0 and day +10, samples for pharmacokinetic analysis were collected before the dose and 1, 2, 4, 6, 8, and 12 hours after maraviroc administration. Additional trough concentrations were collected on days +7, 14, and 21. Patients were followed until day +100 for acute GVHD. Functional blockade of CCR5 was assessed in a pharmacodynamic assay by flow cytometry. Thirteen patients, median age of 4 years (range, 2 to 11 years), were prospectively enrolled. Underlying diagnoses included a primary immune deficiency (n = 6), hemoglobinopathy (n = 4), metabolic disorder (n = 1), and bone marrow failure syndrome (n = 2). Patients received either a myeloablative preparative regimen (n = 7) or a reduced-intensity conditioning regimen (n = 6). Cyclosporine and methylprednisolone (n = 7) was the predominant GVHD prophylactic regimen, followed by tacrolimus and mycophenolate mofetil (n = 4) and tacrolimus and steroids (n = 2). Two formulations of maraviroc (150-mg tablets and 20-mg/mL solution) were used on study. Mean (± SD) area under the concentration-time curve from 0 to 12 hours was 4805 ± 3265 hour * ng/mL on day 0 and 5917 ± 4048 hour * ng/mL on day +10. Four patients developed grade 1 or 2 acute skin GVHD before day +100 and were successfully treated. Two patients developed grade 3 acute GI GVHD on days +23 and +24 after HSCT and both had discontinued maraviroc before development of GI GVHD. No adverse effects attributable to maraviroc were observed and administration by enteral tubes was well tolerated by children and accepted by parents. All evaluable patients demonstrated functional CCR5 blockade on day 0. Administration of maraviroc is feasible in most pediatric HSCT recipients with good safety and tolerability profile.


Subject(s)
Cyclohexanes/administration & dosage , Graft vs Host Disease/prevention & control , Triazoles/administration & dosage , Acute Disease , Child , Child, Preschool , Cyclohexanes/pharmacokinetics , Drug Administration Schedule , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Maraviroc , Premedication/methods , Prospective Studies , Receptors, CCR5/drug effects , Transplantation Conditioning/methods , Treatment Outcome , Triazoles/pharmacokinetics , Viscera/pathology
15.
Medicine (Baltimore) ; 95(28): e4174, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27428211

ABSTRACT

BACKGROUND: The ex vivo challenge assay is a bio-indicator of drug efficacy and was utilized in this randomized, placebo controlled trial as one of the exploratory endpoints. Fresh and cryopreserved tissues were evaluated for human immunodeficiency virus (HIV) infection and pharmacokinetic (PK)/pharmacodynamic (PD) relationships. METHODS: HIV-negative women used vaginal rings containing 25 mg dapivirine (DPV)/100 mg maraviroc (MVC) (n = 12), DPV only (n = 12), MVC only (n = 12), or placebo (n = 12) for 28 days. Blood plasma, cervicovaginal fluid (CVF), and cervical biopsies were collected for drug quantification and the ex vivo challenge assay; half (fresh) were exposed immediately to HIV while the other half were cryopreserved, thawed, then exposed to HIV. HIV replication was monitored by p24 enzyme-linked immunosorbent assay from culture supernatant. Data were log-transformed and analyzed by linear least squared regression, nonlinear Emax dose-response model and Satterthwaite t test. RESULTS: HIV replication was greater in fresh compared to cryopreserved tissue (P = 0.04). DPV was detected in all compartments, while MVC was consistently detected only in CVF. Significant negative correlations between p24 and DPV levels were observed in fresh cervical tissue (P = 0.01) and CVF (P = 0.03), but not plasma. CVF MVC levels showed a significant negative correlation with p24 levels (P = 0.03); drug levels in plasma and tissue were not correlated with HIV suppression. p24 levels from cryopreserved tissue did not correlate to either drug from any compartment. CONCLUSION: Fresh tissue replicated HIV to greater levels and defined PK/PD relationships while cryopreserved tissue did not. The ex vivo challenge assay using fresh tissue could prioritize drugs being considered for HIV prevention.


Subject(s)
Contraceptive Devices, Female , Cyclohexanes/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV Infections/prevention & control , HIV-1/drug effects , Pyrimidines/pharmacology , Sexually Transmitted Diseases, Viral/prevention & control , Triazoles/pharmacology , Administration, Intravaginal , Adult , Biopsy , Cervix Uteri/virology , Cryopreservation , Cyclohexanes/pharmacokinetics , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , HIV Fusion Inhibitors/pharmacokinetics , Humans , In Vitro Techniques , Maraviroc , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics , United States
16.
Neuroimage ; 138: 134-140, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27211474

ABSTRACT

Efflux transporters at the blood-brain barrier can decrease the entry of drugs and increase the removal of those molecules able to bypass the transporter. We previously hypothesized that (18)F-FCWAY, a radioligand for the serotonin 5-HT1A receptor, is a weak substrate for permeability glycoprotein (P-gp) based on its very early peak and rapid washout from human brain. To determine whether (18)F-FCWAY is a substrate for P-gp, breast cancer resistance protein (BCRP), and multidrug resistance protein (MRP1) - the three most prevalent efflux transporters at the blood-brain barrier - we performed three sets of experiments. In vitro, we conducted fluorescence-activated cell sorting (FACS) flow cytometry studies in cells over-expressing P-gp, BCRP, and MRP1 treated with inhibitors specific to each transporter and with FCWAY. Ex vivo, we measured (18)F-FCWAY concentration in plasma and brain homogenate of transporter knockout mice using γ-counter and radio-HPLC. In vivo, we conducted positron emission tomography (PET) studies to assess changes in humans who received (18)F-FCWAY during an infusion of tariquidar (2-4mg/kg iv), a potent and selective P-gp inhibitor. In vitro studies showed that FCWAY allowed fluorescent substrates to get into the cell by competitive inhibition of all three transporters at the cell membrane. Ex vivo measurements in knockout mice indicate that (18)F-FCWAY is a substrate only for P-gp and not BCRP. In vivo, tariquidar increased (18)F-FCWAY brain uptake in seven of eight subjects by 60-100% compared to each person's baseline. Tariquidar did not increase brain uptake via some peripheral mechanism, given that it did not significantly alter concentrations in plasma of the parent radioligand (18)F-FCWAY or its brain-penetrant radiometabolite (18)F-FC. These results show that (18)F-FCWAY is a weak substrate for efflux transport at the blood-brain barrier; some radioligand can enter brain, but its removal is hastened by P-gp. Although (18)F-FCWAY is not ideal for measuring 5-HT1A receptors, it demonstrates that weak substrate radioligands can be useful for measuring both increased and decreased function of efflux transporters, which is not possible with currently available radioligands such as (11)C-loperamide and (11)C-verapamil that are avid substrates for transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Blood-Brain Barrier/metabolism , Cyclohexanes/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Piperazines/pharmacokinetics , Positron-Emission Tomography/methods , Receptor, Serotonin, 5-HT1A/metabolism , Adult , Capillary Permeability/physiology , Female , Humans , Male , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
17.
Antimicrob Agents Chemother ; 60(6): 3759-66, 2016 06.
Article in English | MEDLINE | ID: mdl-27067321

ABSTRACT

Preexposure prophylaxis (PrEP) against HIV using oral regimens based on the nucleoside reverse transcriptase inhibitor tenofovir disoproxil fumarate (TDF) has been effective to various degrees in multiple clinical trials, and the CCR5 receptor antagonist maraviroc (MVC) holds potential for complementary efficacy. The effectiveness of HIV PrEP is highly dependent on adherence. Incorporation of the TDF-MVC combination into intravaginal rings (IVRs) for sustained mucosal delivery could increase product adherence and efficacy compared with oral and vaginal gel formulations. A novel pod-IVR technology capable of delivering multiple drugs is described. The pharmacokinetics and preliminary local safety characteristics of a novel pod-IVR delivering a combination of TDF and MVC were evaluated in the ovine model. The device exhibited sustained release at controlled rates over the 28-day study and maintained steady-state drug levels in cervicovaginal fluids (CVFs). Dilution of CVFs during lavage sample collection was measured by ion chromatography using an inert tracer, allowing corrected drug concentrations to be measured for the first time. Median, steady-state drug levels in vaginal tissue homogenate were as follows: for tenofovir (TFV; in vivo hydrolysis product of TDF), 7.3 × 10(2) ng g(-1) (interquartile range [IQR], 3.0 × 10(2), 4.0 × 10(3)); for TFV diphosphate (TFV-DP; active metabolite of TFV), 1.8 × 10(4) fmol g(-1) (IQR, 1.5 × 10(4), 4.8 × 10(4)); and for MVC, 8.2 × 10(2) ng g(-1) (IQR, 4.7 × 10(2), 2.0 × 10(3)). No adverse events were observed. These findings, together with previous pod-IVR studies, have allowed several lead candidates to advance into clinical evaluation.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Cyclohexanes/pharmacokinetics , Delayed-Action Preparations/pharmacokinetics , HIV Infections/prevention & control , Tenofovir/pharmacokinetics , Triazoles/pharmacokinetics , Vagina/chemistry , Administration, Intravaginal , Animals , Contraceptive Devices, Female , Drug Combinations , Female , Humans , Maraviroc , Models, Animal , Primary Prevention , Sheep , Vagina/drug effects
18.
J Acquir Immune Defic Syndr ; 72(5): 498-506, 2016 08 15.
Article in English | MEDLINE | ID: mdl-26999532

ABSTRACT

BACKGROUND: Quantifying tissue drug concentrations can yield important information during drug development, but complicates pharmacokinetic study design. Mucosal fluids collected by direct aspiration (cervicovaginal fluid; CVF) or swab (rectal fluid; RF) might be used as tissue concentration surrogates, but these relationships are not well characterized. METHODS: Forty-nine healthy women, given a single oral dose of tenofovir, maraviroc, emtricitabine, or raltegravir at 50%-200% of the treatment dose, provided 13 plasma, 12 CVF, 12 RF and one cervical, vaginal and rectal tissue biopsy over 48 hours. Relationships between these paired samples were characterized by linear and multiple linear regression. Adjusted r values were used to select the final predictive models. RESULTS: CVF exposure increased linearly with dose for all antiretrovirals (r(2) ≥ 0.23, P ≤ 0.02) except raltegravir (r(2) = 0.08, P = 0.19). In RF, only emtricitabine increased linearly with dose (r(2) = 0.27, P = 0.01). For all antiretrovirals, CVF and RF concentrations significantly correlated with mucosal tissue concentrations (female genital tract r(2) ≥ 0.37, rectal tissue (2)r ≥ 0.50, P ≤ 0.001). In the final multivariate models, plasma and fluid concentrations were both associated with FGT concentrations for all antiretrovirals (r(2) ≥ 0.81, P < 0.001). The same was noted for rectal tissue (r(2) ≥ 0.58, P < 0.001) except for tenofovir, for which RF alone was predictive of tissue concentration (r(2) = 0.91, P < 0.001). CONCLUSIONS: Mucosal fluids were positively correlated with tissue concentrations and including plasma concentrations improved the regression models in most cases. Dose linearity in CVF, but not RF, suggests a saturation process in lower gastrointestinal tract tissue. These findings suggest that mucosal fluid and plasma concentrations may be used for qualitative inference of tissue concentrations for these antiretrovirals.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Body Fluids/metabolism , Cervix Uteri/metabolism , Clinical Trials as Topic/methods , Rectum/metabolism , Vagina/metabolism , Adult , Biomarkers/analysis , Body Fluids/drug effects , Cervix Uteri/drug effects , Cyclohexanes/pharmacokinetics , Emtricitabine/pharmacokinetics , Female , Healthy Volunteers , Humans , Maraviroc , Raltegravir Potassium/pharmacokinetics , Rectum/drug effects , Tenofovir/pharmacokinetics , Triazoles/pharmacokinetics , Vagina/drug effects
19.
Drug Res (Stuttg) ; 66(2): 74-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26158794

ABSTRACT

OBJECTIVE: Tofogliflozin is an oral hypoglycemic agent with a novel mechanism of action that reduces blood glucose levels by promoting glucose excretion in urine, achieved by selectively inhibiting sodium-glucose co-transporter 2 (SGLT2). We evaluated the effects of several selected anti-type 2 diabetes mellitus (T2DM) drugs-glimepiride, metformin, sitagliptin, pioglitazone, miglitol, nateglinide, and voglibose-on the pharmacokinetics and pharmacodynamics of tofogliflozin, and the effects of tofogliflozin on the pharmacokinetics of these anti-T2DM drugs in healthy male volunteers. METHODS: A single dose of either tofogliflozin alone, one of the anti-T2DM drugs alone, or co-administration of tofogliflozin and the anti-T2DM drug was administered to 108 healthy men. Cmax, AUCinf, and cumulative urine glucose excretion after co-administration of tofogliflozin and each of the anti-T2DM drugs was evaluated relative to the values of those parameters after administration of each drug alone. RESULTS: None of the anti-T2DM drugs had any effect on tofogliflozin exposure. Tofogliflozin had no or little effect on the exposure of any anti-T2DM drug. No anti-T2DM drug had any major effect on the cumulative urine glucose excretion induced by tofogliflozin. There were no safety concerns evident after administration of any drug alone or in co-administration. CONCLUSIONS: Neither the pharmacokinetics nor the pharmacodynamics of tofogliflozin was affected by any of the anti-T2DM drugs evaluated in this study, nor was the pharmacokinetics of any of the anti-T2DM drugs affected by tofogliflozin in healthy male volunteers.


Subject(s)
Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/pharmacokinetics , Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Glucosides/pharmacokinetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacokinetics , 1-Deoxynojirimycin/pharmacology , Adult , Cyclohexanes/pharmacokinetics , Cyclohexanes/pharmacology , Diabetes Mellitus, Type 2/blood , Drug Interactions , Glucose/analysis , Healthy Volunteers , Humans , Inositol/analogs & derivatives , Inositol/pharmacokinetics , Inositol/pharmacology , Male , Metformin/pharmacokinetics , Metformin/pharmacology , Middle Aged , Nateglinide , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacokinetics , Phenylalanine/pharmacology , Pioglitazone , Sitagliptin Phosphate/pharmacokinetics , Sitagliptin Phosphate/pharmacology , Sulfonylurea Compounds/pharmacokinetics , Sulfonylurea Compounds/pharmacology , Thiazolidinediones/pharmacokinetics , Thiazolidinediones/pharmacology , Urine/chemistry , Young Adult
20.
AAPS PharmSciTech ; 17(3): 673-81, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26314243

ABSTRACT

The aim of the present work was to prepare a co-amorphous mixture (COAM) of Nateglinide and Metformin hydrochloride to enhance the dissolution rate of poorly soluble Nateglinide. Nateglinide (120 mg) and Metformin hydrochloride (500 mg) COAM, as a dose ratio, were prepared by ball-milling technique. COAMs were characterized for saturation solubility, amorphism and physicochemical interactions (X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), Fourier transform infrared spectroscopy (FTIR)), SEM, in vitro dissolution, and stability studies. Solubility studies revealed a sevenfold rise in solubility of Nateglinide from 0.061 to 0.423 mg/ml in dose ratio of COAM. Solid-state characterization of COAM suggested amorphization of Nateglinide after 6 h of ball milling. XRPD and DSC studies confirmed amorphism in Nateglinide, whereas FTIR elucidated hydrogen interactions (proton exchange between Nateglinide and Metformin hydrochloride). Interestingly, due to low energy of fusion, Nateglinide was completely amorphized and stabilized by Metformin hydrochloride. Consequently, in vitro drug release showed significant increase in dissolution of Nateglinide in COAM, irrespective of dissolution medium. However, little change was observed in the solubility and dissolution profile of Metformin hydrochloride, revealing small change in its crystallinity. Stability data indicated no traces of devitrification in XRPD of stability sample of COAM, and % drug release remained unaffected at accelerated storage conditions. Amorphism of Nateglinide, proton exchange with Metformin hydrochloride, and stabilization of its amorphous form have been noted in ball-milled COAM of Nateglinide-Metformin hydrochloride, revealing enhanced dissolution of Nateglinide. Thus, COAM of Nateglinide-Metformin hydrochloride system is a promising approach for combination therapy in diabetic patients.


Subject(s)
Cyclohexanes/analysis , Cyclohexanes/chemical synthesis , Metformin/analysis , Metformin/chemical synthesis , Phenylalanine/analogs & derivatives , Chemistry, Pharmaceutical , Cyclohexanes/pharmacokinetics , Drug Combinations , Metformin/pharmacokinetics , Nateglinide , Phenylalanine/analysis , Phenylalanine/chemical synthesis , Phenylalanine/pharmacokinetics , Solubility , Spectroscopy, Fourier Transform Infrared/methods , X-Ray Diffraction/methods
SELECTION OF CITATIONS
SEARCH DETAIL