Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Ann Intern Med ; 172(2): 86-95, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31887743

ABSTRACT

Background: MIV-711 is a novel selective cathepsin K inhibitor with beneficial effects on bone and cartilage in preclinical osteoarthritis models. Objective: To evaluate the efficacy, safety, and tolerability of MIV-711 in participants with symptomatic, radiographic knee osteoarthritis. Design: 26-week randomized, double-blind, placebo-controlled phase 2a study with a 26-week open-label safety extension substudy. (EudraCT: 2015-003230-26 and 2016-001096-73). Setting: Six European sites. Participants: 244 participants with primary knee osteoarthritis, Kellgren-Lawrence grade 2 or 3, and pain score of 4 to 10 on a numerical rating scale (NRS). Intervention: MIV-711, 100 (n = 82) or 200 (n = 81) mg daily, or matched placebo (n = 77). Participants (46 who initially received 200 mg/d and 4 who received placebo) received 200 mg of MIV-711 daily during the extension substudy. Measurements: The primary outcome was change in NRS pain score. The key secondary outcome was change in bone area on magnetic resonance imaging (MRI). Other secondary end points included cartilage thickness on quantitative MRI and type I and II collagen C-telopeptide biomarkers. Outcomes were assessed over 26 weeks. Results: Changes in NRS pain scores with MIV-711 were not statistically significant (placebo, -1.4; MIV-711, 100 mg/d, -1.7; MIV-711, 200 mg/d, -1.5). MIV-711 significantly reduced medial femoral bone area progression (P = 0.002 for 100 mg/d and 0.004 for 200 mg/d) and medial femoral cartilage thinning (P = 0.023 for 100 mg/d and 0.125 for 200 mg/d) versus placebo and substantially reduced bone and cartilage biomarker levels. Nine serious adverse events occurred in 6 participants (1 in the placebo group, 3 in the 100 mg group, and 2 in the 200 mg group); none were considered to be treatment-related. Limitation: The trial was relatively short. Conclusion: MIV-711 was not more effective than placebo for pain, but it significantly reduced bone and cartilage progression with a reassuring safety profile. This treatment may merit further evaluation as a disease-modifying osteoarthritis drug. Primary Funding Source: Medivir.


Subject(s)
Cathepsin K/antagonists & inhibitors , Organic Chemicals/therapeutic use , Osteoarthritis, Knee/drug therapy , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Double-Blind Method , Europe , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Pain Measurement
3.
Antimicrob Agents Chemother ; 59(6): 3189-96, 2015.
Article in English | MEDLINE | ID: mdl-25779585

ABSTRACT

Compound A is a novel nucleotide-competing HIV-1 reverse transcriptase (RT) inhibitor (NcRTI) that selects for a unique W153L substitution that confers hypersusceptibility to tenofovir, while the K65R substitution in RT confers resistance against tenofovir and enhances susceptibility to NcRTIs. Although the K65R substitution is more common in subtype C viruses, the impact of subtype variability on NcRTI susceptibility has not been studied. In the present study, we performed experiments with compound A by using purified recombinant RT enzymes and viruses of subtypes B and C and circulating recombinant form CRF_A/G. We confirmed the hypersusceptibility of K65R substitution-containing RTs to compound A for subtype C, CRF_A/G, and subtype B. Steady-state kinetic analysis showed that K65R RTs enhanced the susceptibility to compound A by increasing binding of the inhibitor to the nucleotide binding site of RT in a subtype-independent manner, without significantly discriminating against the natural nucleotide substrate. These data highlight the potential utility of NcRTIs, such as compound A, for treatment of infections with K65R substitution-containing viruses, regardless of HIV-1 subtype.


Subject(s)
HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Amino Acid Substitution , Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans , Kinetics
4.
Antimicrob Agents Chemother ; 58(8): 4515-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24867966

ABSTRACT

A W153L substitution in HIV-1 reverse transcriptase (RT) was recently identified by selection with a novel nucleotide-competing RT inhibitor (NcRTI) termed compound A that is a member of the benzo[4,5]furo[3,2,d]pyrimidin-2-one NcRTI family of drugs. To investigate the impact of W153L, alone or in combination with the clinically relevant RT resistance substitutions K65R (change of Lys to Arg at position 65), M184I, K101E, K103N, E138K, and Y181C, on HIV-1 phenotypic susceptibility, viral replication, and RT enzymatic function, we generated recombinant RT enzymes and viruses containing each of these substitutions or various combinations of them. We found that W153L-containing viruses were impaired in viral replicative capacity and were hypersusceptible to tenofovir (TFV) while retaining susceptibility to most nonnucleoside RT inhibitors. The nucleoside 3TC retained potency against W153L-containing viruses but not when the M184I substitution was also present. W153L was also able to reverse the effects of the K65R substitution on resistance to TFV, and K65R conferred hypersusceptibility to compound A. Biochemical assays demonstrated that W153L alone or in combination with K65R, M184I, K101E, K103N, E138K, and Y181C impaired enzyme processivity and polymerization efficiency but did not diminish RNase H activity, providing mechanistic insights into the low replicative fitness associated with these substitutions. We show that the mechanism of the TFV hypersusceptibility conferred by W153L is mainly due to increased efficiency of TFV-diphosphate incorporation. These results demonstrate that compound A and/or derivatives thereof have the potential to be important antiretroviral agents that may be combined with tenofovir to achieve synergistic results.


Subject(s)
Amino Acid Substitution , Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Alkynes , Benzoxazines/pharmacology , Cyclopropanes , HEK293 Cells , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , Humans , Lamivudine/pharmacology , Microbial Sensitivity Tests , Mutagenesis, Site-Directed , Mutation , Nevirapine/pharmacology , Organophosphonates/pharmacology , Pyrimidines/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tenofovir , Virus Replication
5.
Antimicrob Agents Chemother ; 58(6): 3233-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24663024

ABSTRACT

BI 224436 is an HIV-1 integrase inhibitor with effective antiviral activity that acts through a mechanism that is distinct from that of integrase strand transfer inhibitors (INSTIs). This 3-quinolineacetic acid derivative series was identified using an enzymatic integrase long terminal repeat (LTR) DNA 3'-processing assay. A combination of medicinal chemistry, parallel synthesis, and structure-guided drug design led to the identification of BI 224436 as a candidate for preclinical profiling. It has antiviral 50% effective concentrations (EC50s) of <15 nM against different HIV-1 laboratory strains and cellular cytotoxicity of >90 µM. BI 224436 also has a low, ∼2.1-fold decrease in antiviral potency in the presence of 50% human serum and, by virtue of a steep dose-response curve slope, exhibits serum-shifted EC95 values ranging between 22 and 75 nM. Passage of virus in the presence of inhibitor selected for either A128T, A128N, or L102F primary resistance substitutions, all mapping to a conserved allosteric pocket on the catalytic core of integrase. BI 224436 also retains full antiviral activity against recombinant viruses encoding INSTI resistance substitutions N155S, Q148H, and E92Q. In drug combination studies performed in cellular antiviral assays, BI 224436 displays an additive effect in combination with most approved antiretrovirals, including INSTIs. BI 224436 has drug-like in vitro absorption, distribution, metabolism, and excretion (ADME) properties, including Caco-2 cell permeability, solubility, and low cytochrome P450 inhibition. It exhibited excellent pharmacokinetic profiles in rat (clearance as a percentage of hepatic flow [CL], 0.7%; bioavailability [F], 54%), monkey (CL, 23%; F, 82%), and dog (CL, 8%; F, 81%). Based on the excellent biological and pharmacokinetic profile, BI 224436 was advanced into phase 1 clinical trials.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/enzymology , Amino Acid Substitution/genetics , Amino Acid Substitution/physiology , Animals , Anti-HIV Agents/pharmacology , Caco-2 Cells , Cloning, Molecular , Cytochrome P-450 Enzyme Inhibitors/pharmacology , DNA, Viral/drug effects , Drug Resistance, Viral , HIV Integrase/biosynthesis , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/metabolism , HIV Integrase Inhibitors/pharmacokinetics , Hepatocytes/metabolism , Humans , Mice , Rats , Serum/virology , Virus Replication/drug effects
6.
J Transl Med ; 12: 217, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25179432

ABSTRACT

BACKGROUND: Coexpression of CD160 and PD-1 on HIV-specific CD8+ T-cells defines a highly exhausted T-cell subset. CD160 binds to Herpes Virus Entry Mediator (HVEM) and blocking this interaction with HVEM antibodies reverses T-cell exhaustion. As HVEM binds both inhibitory and activatory receptors, our aim in the current study was to assess the impact of CD160-specific antibodies on the enhancement of T-cell activation. METHODS: Expression of the two CD160 isoforms; glycosylphosphatidylinositol-anchored (CD160-GPI) and the transmembrane isoforms (CD160-TM) was assessed in CD4 and CD8 primary T-cells by quantitative RT-PCR and Flow-cytometry. Binding of these isoforms to HVEM ligand and the differential capacities of CD160 and HVEM specific antibodies to inhibit this binding were further evaluated using a Time-Resolved Fluorescence assay (TRF). The impact of both CD160 and HVEM specific antibodies on enhancing T-cell functionality upon antigenic stimulation was performed in comparative ex vivo studies using primary cells from HIV-infected subjects stimulated with HIV antigens in the presence or absence of blocking antibodies to the key inhibitory receptor PD-1. RESULTS: We first show that both CD160 isoforms, CD160-GPI and CD160-TM, were expressed in human primary CD4+ and CD8+ T-cells. The two isoforms were also recognized by the HVEM ligand, although this binding was less pronounced with the CD160-TM isoform. Mechanistic studies revealed that although HVEM specific antibodies blocked its binding to CD160-GPI, surprisingly, these antibodies enhanced HVEM binding to CD160-TM, suggesting that potential antibody-mediated HVEM multimerization and/or induced conformational changes may be required for optimal CD160-TM binding. Triggering of CD160-GPI over-expressed on Jurkat cells with either bead-bound HVEM-Fc or anti-CD160 monoclonal antibodies enhanced cell activation, consistent with a positive co-stimulatory role for CD160-GPI. However, CD160-TM did not respond to this stimulation, likely due to the lack of optimal HVEM binding. Finally, ex vivo assays using PBMCs from HIV viremic subjects showed that the use of CD160-GPI-specific antibodies combined with blockade of PD-1 synergistically enhanced the proliferation of HIV-1 specific CD8+ T-cells upon antigenic stimulation. CONCLUSIONS: Antibodies targeting CD160-GPI complement the blockade of PD-1 to enhance HIV-specific T-cell responses and warrant further investigation in the development of novel immunotherapeutic approaches.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Lymphocyte Activation/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Adult , Animals , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Cricetinae , Cricetulus , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , HIV Infections/genetics , HIV Infections/metabolism , Humans , Jurkat Cells , Male , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor, Member 14/metabolism
7.
Bioorg Med Chem Lett ; 24(21): 4984-8, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25288185

ABSTRACT

Previous investigations identified 2'-C-Me-branched ribo-C-nucleoside adenosine analogues, 1, which contains a pyrrolo[2,1-f][1,2,4]triazin-4-amine heterocyclic base, and 2, which contains an imidazo[2,1-f][1,2,4]triazin-4-amine heterocyclic base as two compounds with promising anti-HCV in vitro activity. This Letter describes the synthesis and evaluation of a series of novel analogues of these compounds substituted at the 2-, 7-, and 8-positions of the heterocyclic bases. A number of active new HCV inhibitors were identified but most compounds also demonstrated unacceptable cytotoxicity. However, the 7-fluoro analogue of 1 displayed good potency with a promising cytotherapeutic margin.


Subject(s)
Antiviral Agents/pharmacology , Cell Proliferation/drug effects , Hepacivirus/drug effects , Imidazoles/chemistry , Nucleosides/pharmacology , Pyrroles/chemistry , Triazines/chemistry , Virus Replication/drug effects , Antiviral Agents/chemistry , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/virology , Molecular Structure , Nucleosides/chemistry , RNA, Viral/genetics , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Antimicrob Agents Chemother ; 57(10): 4928-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23877706

ABSTRACT

Faldaprevir (BI 201335) is a selective NS3/4A protease inhibitor under development for the treatment of chronic hepatitis C virus (HCV) infection. NS3/4A genotyping and NS3 protease phenotyping analyses were performed to monitor the emergence of resistance in patients with HCV genotype 1 infection receiving faldaprevir alone or combined with pegylated interferon alfa 2a and ribavirin (PegIFN-RBV) during a phase 1b study. Among all baseline variants, a maximum 7-fold reduction in in vitro sensitivity to faldaprevir was observed for a rare NS3 (V/I)170T polymorphism. During faldaprevir monotherapy in treatment-naive patients, virologic breakthrough was common (77%, 20/26) and was associated with the emergence of resistance mutations predominantly carrying NS3 substitutions R155K in GT1a and D168V in GT1b. D168V conferred a greater reduction in faldaprevir sensitivity (1,800-fold) than R155K (330-fold); however, D168V was generally less fit than R155K in the absence of selective drug pressure. Treatment-experienced patients treated with faldaprevir-PegIFN-RBV triple therapy showed higher viral load reductions, lower rates of breakthrough (8%, 5/62), and less frequent emergence of resistance-associated variants compared with faldaprevir monotherapy. (This study has been registered at ClinicalTrials.gov under registration no. NCT00793793.).


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Oligopeptides/therapeutic use , Thiazoles/therapeutic use , Aminoisobutyric Acids , Cell Line , Genotype , Genotyping Techniques/methods , Humans , Leucine/analogs & derivatives , Proline/analogs & derivatives , Protease Inhibitors/therapeutic use , Quinolines , Viral Nonstructural Proteins/antagonists & inhibitors
9.
J Virol ; 86(21): 11595-607, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22896614

ABSTRACT

Phosphatidylinositol-4-kinase IIIα (PI4KIIIα) is an essential host cell factor for hepatitis C virus (HCV) replication. An N-terminally truncated 130-kDa form was used to reconstitute an in vitro biochemical lipid kinase assay that was optimized for small-molecule compound screening and identified potent and specific inhibitors. Cell culture studies with PI4KIIIα inhibitors demonstrated that the kinase activity was essential for HCV RNA replication. Two PI4KIIIα inhibitors were used to select cell lines harboring HCV replicon mutants with a 20-fold loss in sensitivity to the compounds. Reverse genetic mapping isolated an NS4B-NS5A segment that rescued HCV RNA replication in PIK4IIIα-deficient cells. HCV RNA replication occurs on specialized membranous webs, and this study with PIK4IIIα inhibitor-resistant mutants provides a genetic link between NS4B/NS5A functions and PI4-phosphate lipid metabolism. A comprehensive assessment of PI4KIIIα as a drug target included its evaluation for pharmacologic intervention in vivo through conditional transgenic murine lines that mimic target-specific inhibition in adult mice. Homozygotes that induce a knockout of the kinase domain or knock in a single amino acid substitution, kinase-defective PI4KIIIα, displayed a lethal phenotype with a fairly widespread mucosal epithelial degeneration of the gastrointestinal tract. This essential host physiologic role raises doubt about the pursuit of PI4KIIIα inhibitors for treatment of chronic HCV infection.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Hepacivirus/physiology , Host-Pathogen Interactions , Virus Replication , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Animals , Antiviral Agents/pharmacology , Cell Line , DNA Mutational Analysis , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Female , Genes, Essential , Hepatocytes/enzymology , Hepatocytes/virology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mutant Proteins/genetics , Viral Nonstructural Proteins/genetics
10.
Bioorg Med Chem Lett ; 23(9): 2781-6, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23545107

ABSTRACT

A HTS screen led to the identification of a benzofurano[3,2-d]pyrimidin-2-one core structure which upon further optimization resulted in 1 as a potent HIV-1 nucleotide competing reverse transcriptase inhibitor (NcRTI). Investigation of the SAR at N-1 allowed significant improvements in potency and when combined with the incorporation of heterocycles at C-8 resulted in potent analogues not requiring a basic amine to achieve antiviral activity. Additional modifications at N-1 resulted in 33 which demonstrated excellent antiviral potency and improved physicochemical properties.


Subject(s)
Benzofurans/chemistry , HIV Reverse Transcriptase/antagonists & inhibitors , Nucleotides/chemistry , Pyrimidinones/chemistry , Reverse Transcriptase Inhibitors/chemistry , Caco-2 Cells , Cell Membrane Permeability/drug effects , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/enzymology , Humans , Microsomes, Liver/metabolism , Nucleotides/metabolism , Protein Binding , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacology , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacology , Stereoisomerism , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 23(13): 3967-75, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23673016

ABSTRACT

Recently, a new class of HIV reverse transcriptase (HIV-RT) inhibitors has been reported. The novel mechanism of inhibition by this class involves competitive binding to the active site of the RT enzyme and has been termed Nucleotide-Competing Reverse Transcriptase Inhibitors (NcRTIs). In this publication we describe the optimization of a novel benzofurano[3,2-d]pyrimidin-2-one series of NcRTIs. The starting point for the current study was inhibitor 2, which had high biochemical and antiviral potency but only moderate permeability in a Caco-2 assay and high B-to-A efflux, resulting in moderate rat bioavailability and low Cmax. We present herein the results and strategies we employed to optimize both the potency as well as the permeability, metabolic stability and pharmacokinetic profile of this series. One of the key observations of the present study was the importance of shielding polar functionality, at least in the context of the current chemotype, to enhance permeability. These studies led to the identification of inhibitors 39 and 45, which display sub-nanomolar antiviral potency in a p24 ELISA assay with significantly reduced efflux ratios (ratios <1.5). These inhibitors also display excellent rat pharmacokinetic profiles with high bioavailabilities and low clearance.


Subject(s)
Antiviral Agents/pharmacology , Benzofurans/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV/drug effects , Pyrimidinones/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Benzofurans/chemistry , Biological Availability , Caco-2 Cells , Dose-Response Relationship, Drug , HIV Reverse Transcriptase/metabolism , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Pyrimidinones/administration & dosage , Pyrimidinones/chemistry , Rats , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 23(9): 2775-80, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23511023

ABSTRACT

Screening of our sample collection led to the identification of a set of benzofurano[3,2-d]pyrimidine-2-one hits acting as nucleotide-competing HIV-1 reverse transcriptase inhibitiors (NcRTI). Significant improvement in antiviral potency was achieved when substituents were introduced at positions N1, C4, C7 and C8 on the benzofuranopyrimidone scaffold. The series was optimized from low micromolar enzymatic activity against HIV-1 RT and no antiviral activity to low nanomolar antiviral potency. Further profiling of inhibitor 30 showed promising overall in vitro properties and also demonstrated that its potency was maintained against viruses resistant to the other major classes of HIV-1 RT inhibitors.


Subject(s)
Benzofurans/chemistry , HIV Reverse Transcriptase/antagonists & inhibitors , Nucleotides/chemistry , Pyrimidinones/chemistry , Reverse Transcriptase Inhibitors/chemistry , Animals , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/enzymology , Humans , Microsomes, Liver/metabolism , Nucleotides/metabolism , Protein Binding , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacology , Rats , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship
13.
Xenobiotica ; 43(7): 592-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23244592

ABSTRACT

1. The present study evaluates which factors should be incorporated into a simplified approach to reasonably predict CYP3A-mediated drug-drug interaction (DDI) at an early drug discovery stage. 2. CYP3A IC50 values were obtained using human liver microsomes (HLM) and hepatocytes. Plasma and microsomal protein binding and in vitro hepatocyte partition coefficient (Kp) were also determined for 10 drugs. Therapeutic human maximum plasma concentrations (Cmax) were retrieved from the literature. DDI predictions were performed using an equation incorporating the fraction of the substrate metabolized by CYP3A with the total or free plasma Cmax, with or without correction for hepatocyte Kp. 3. Based on the Ki data from HLM, the use of total Cmax provided a prediction of DDI within 2-fold of the observed clinical values for 9 out of 10 drugs. 4. In comparison, free drug corrections for both Cmax and Ki values from HLM led to an underprediction of DDI (>3-fold error for five drugs). 5. Data from hepatocytes showed, in general, lower prediction accuracy than data from HLM. 6. CYP3A-mediated DDIs can be predicted with a high level of accuracy based on Ki estimates from HLM data and the total therapeutic plasma Cmax of the inhibitors. This approach should be widely applicable to the assessment of clinically significant DDIs risk in early drug discovery programs.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Pharmaceutical Preparations/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Drug Interactions , Hepatocytes/metabolism , Humans , Kinetics , Microsomes, Liver/metabolism , Protein Binding
14.
Antimicrob Agents Chemother ; 56(10): 5381-6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22869578

ABSTRACT

Pharmacokinetic-pharmacodynamic (PK-PD) data analyses from early hepatitis C virus (HCV) clinical trials failed to show a good correlation between the plasma inhibitory quotient (IQ) and antiviral activity of different classes of directly acting antiviral agents (DAAs). The present study explored whether use of the liver partition coefficient-corrected IQ (LCIQ) could improve the PK-PD relationship. Animal liver partition coefficients (Kp(liver)) were calculated from liver to plasma exposure ratios. In vitro hepatocyte partition coefficients (Kp(hep)) were determined by the ratio of cellular to medium drug concentrations. Human Kp(liver) was predicted using an in vitro-in vivo proportionality method: the species-averaged animal Kp(liver) multiplied by the ratio of human Kp(hep) over those in animals. LCIQ was calculated using the IQ multiplied by the predicted human Kp(liver). Our results demonstrated that the in vitro-in vivo proportionality approach provided the best human Kp(liver) prediction, with prediction errors of <45% for all 5 benchmark drugs evaluated (doxorubicin, verapamil, digoxin, quinidine, and imipramine). Plasma IQ values correlated poorly (r(2) of 0.48) with maximum viral load reduction and led to a corresponding 50% effective dose (ED(50)) IQ of 42, with a 95% confidence interval (CI) of 0.1 to 148534. In contrast, the LCIQ-maximum VLR relationship fit into a typical sigmoidal curve with an r(2) value of 0.95 and an ED(50) LCIQ of 121, with a 95% CI of 83 to 177. The present study provides a novel human Kp(liver) prediction model, and the LCIQ correlated well with the viral load reductions observed in short-term HCV monotherapy of different DAAs and provides a valuable tool to guide HCV drug discovery.


Subject(s)
Antiviral Agents/pharmacokinetics , Hepacivirus/drug effects , Liver/metabolism , Animals , Cells, Cultured , Digoxin/pharmacokinetics , Doxorubicin/pharmacokinetics , Hepatocytes/virology , Humans , Imipramine/pharmacokinetics , Male , Mice , Quinidine/pharmacokinetics , Rats , Rats, Sprague-Dawley , Verapamil/pharmacokinetics
15.
Xenobiotica ; 42(2): 164-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21988548

ABSTRACT

The present study describes the cross-species absorption, metabolism, distribution and pharmacokinetics of BI 201335, a potent HCV protease inhibitor currently in phase III clinical trials. BI 201335 showed a good Caco-II permeability (8.7 × 10(-6) cm/sec) and in vitro metabolic stability (predicted hepatic clearence (CL(hep)) <19% Q(h) in all species tested). Single dose PK revealed a clearance of 17, 3.0 and 2.6 mL/min/kg in rat, monkey and dog respectively, with a corresponding oral bioavailability of 29.1, 25.5 and 35.6%. Comparative plasma and liver PK profile in rodents showed a high liver Kp in the rat (42-fold), suggesting high target tissue distribution. Simple allometry based on animal PK predicted a human oral CL/F of 168 mL/min, within two-fold of the observed value (118 mL/min) at 240 mg in healthy volunteers. Allometry of volume of distribution generated a low exponent of 0.59, and a much lower predicted Vss/F (5-fold less than observed). Several different approaches of Vss/F prediction were evaluated and compared with the value observed in human. The averaged Vss/F from preclinical animals provides the best estimation of the observed human value (169 L vs. 175 L). Corresponding human "effective" t(1/2) values were also compared. The predicted human t(1/2) based on the CL from allometry with metabolic corrections and the averaged animal Vss represented the best estimation of the clinical data (12.1 vs. 17.2 hr). The present study demonstrated that the good preclinical ADMEPK profile of BI 201335 is consistent with that observed in the clinic. While preclinical data accurately predicted the human CL, the prediction of human Vss seems to be more challenging. The averaged Vss/F from all tested preclinical animals provided the best prediction of human Vss and the resulting "effective" t(1/2).


Subject(s)
Antiviral Agents/pharmacokinetics , Oligopeptides/pharmacokinetics , Protease Inhibitors/pharmacokinetics , Thiazoles/pharmacokinetics , Viral Nonstructural Proteins/antagonists & inhibitors , Absorption , Aminoisobutyric Acids , Animals , Biological Availability , Caco-2 Cells , Dogs , Drug Evaluation, Preclinical , Hepacivirus/enzymology , Humans , Leucine/analogs & derivatives , Macaca mulatta , Male , Microsomes, Liver , Oligopeptides/chemistry , Proline/analogs & derivatives , Quinolines , Rats , Rats, Sprague-Dawley , Thiazoles/chemistry , Tissue Distribution
16.
Bioorg Med Chem Lett ; 21(1): 398-404, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21087861

ABSTRACT

The discovery of a 1,5-dihydrobenzo[b][1,4]diazepine-2,4-dione series of inhibitors of HIV-1 capsid assembly is described. Synthesis of analogs of the 1,5-dihydrobenzo[b][1,4]diazepine-2,4-dione hit established structure-activity relationships. Replacement of the enamine functionality of the hit series with either an imidazole or a pyrazole ring led to compounds that inhibited both capsid assembly and reverse transcriptase. Optimization of the bicyclic benzodiazepine scaffold to include a 3-phenyl substituent led to lead compound 48, a pure capsid assembly inhibitor with improved antiviral activity.


Subject(s)
Anti-HIV Agents/chemistry , Benzodiazepinones/chemistry , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Benzodiazepinones/chemical synthesis , Benzodiazepinones/pharmacology , Capsid Proteins/metabolism , Drug Evaluation, Preclinical , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , Humans , Imidazoles/chemistry , Pyrazoles/chemistry , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship
17.
Antimicrob Agents Chemother ; 54(11): 4611-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823284

ABSTRACT

BI 201335 is a hepatitis C virus (HCV) NS3-NS4A (NS3 coexpressed with NS4A) protease inhibitor that has been shown to have potent clinical antiviral activity. It is a highly optimized noncovalent competitive inhibitor of full-length NS3-NS4A proteases of HCV genotypes 1a and 1b with K(i) values of 2.6 and 2.0 nM, respectively. K(i) values of 2 to 230 nM were measured against the NS3-NS4A proteases of HCV genotypes 2 to 6, whereas it was a very weak inhibitor of cathepsin B and showed no measurable inhibition of human leukocyte elastase. BI 201335 was also shown to be a potent inhibitor of HCV RNA replication in vitro with 50% effective concentrations (EC(50)s) of 6.5 and 3.1 nM obtained in genotype 1a and 1b replicon assays. Combinations of BI 201335 with either interferon or ribavirin had additive effects in replicon assays. BI 201335 had good permeability in Caco-2 cell assays and high metabolic stability after incubation with human, rat, monkey, and dog liver microsomes. Its good absorption, distribution, metabolism, and excretion (ADME) profile in vitro, as well as in rat, monkey, and dog, predicted good pharmacokinetics (PK) in humans. Furthermore, drug levels were significantly higher in rat liver than in plasma, suggesting that distribution to the target organ may be especially favorable. BI 201335 is a highly potent and selective NS3-NS4A protease inhibitor with good in vitro and animal ADME properties, consistent with its good human PK profile, and shows great promise as a treatment for HCV infection.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Hepacivirus/enzymology , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Thiazoles/pharmacology , Thiazoles/pharmacokinetics , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Aminoisobutyric Acids , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Caco-2 Cells , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins , Leucine/analogs & derivatives , Male , Proline/analogs & derivatives , Proline/pharmacology , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Quinolines , Rats , Viral Nonstructural Proteins/drug effects , Virus Replication/drug effects
18.
Antiviral Res ; 76(1): 68-74, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17628710

ABSTRACT

Several nucleoside reverse transcriptase inhibitors are associated with mitochondrial toxicity resulting from inhibition of DNA polymerase-gamma. This study compared the effects on mitochondrial DNA of apricitabine (previously referred to as AVX754 or SPD754), a novel cytidine analogue under development for the treatment of human immunodeficiency virus (HIV)-1 infection, and other reverse transcriptase inhibitors. Human HepG2 hepatoblastoma were cultured for up to 16 days with test compounds at concentrations of 0.3-300 microM. Mitochondrial DNA replication was assessed by means of a duplex nucleic acid sequence-based amplification technique, which measures the ratio of the number of mitochondrial DNA copies to the number of genomic DNA copies. Apricitabine and tenofovir had no effect on the mitochondrial DNA content. In contrast, alovudine, zalcitabine, didanosine and stavudine markedly reduced mitochondrial DNA content, whereas abacavir, emtricitabine, lamivudine and zidovudine produced slight increases in mitochondrial DNA, which may reflect an adaptive cellular response to mitochondrial dysfunction. These results suggest that apricitabine shows a favorable mitochondrial toxicity profile, which is important for long-term clinical use. Further studies are warranted to define the clinical implications of these findings.


Subject(s)
Deoxycytidine/analogs & derivatives , Reverse Transcriptase Inhibitors/toxicity , Adenine/analogs & derivatives , Adenine/toxicity , DNA Replication , DNA, Mitochondrial/biosynthesis , Deoxycytidine/toxicity , Hepatocytes , Humans , Organophosphonates/toxicity , Tenofovir
19.
Antivir Chem Chemother ; 18(2): 61-70, 2007.
Article in English | MEDLINE | ID: mdl-17542150

ABSTRACT

Existing nucleoside reverse transcriptase inhibitors for HIV disease are limited by problems of resistance and, in some cases, long-term toxicity. Apricitabine (ATC; formerly BCH10618, SPD754 and AVX754) is a deoxycytidine analogue nucleoside reverse transcriptase inhibitor in clinical development. ATC retains substantial in vitro activity against HIV-1 containing many mutations associated with nucleoside reverse transcriptase inhibitor resistance, showing a less than twofold reduction in susceptibility in the presence of either up to five thymidine analogue mutations or the M184V mutation. ATC showed a low potential for cellular or mitochondrial toxicity in vitro. ATC is well absorbed orally, with a bioavailability of 65-80%. Its plasma elimination half-life (approximately 3 h), and the intracellular half-life of its triphosphate (TP) metabolite (6-7 h) support twice-daily dosing. Intracellular ATC-TP levels are markedly reduced in the presence of lamivudine or emtricitabine, indicating that clinical co-administration of ATC together with these agents will not be possible. The drug is renally eliminated, giving a low potential for hepatic drug interactions. In a double-blind, randomized, placebo-controlled Phase II monotherapy trial in antiretroviral-naive patients, ATC doses of 1,200 and 1,600 mg/day reduced plasma viral load levels by 1.65 and 1.58 log10 HIV RNA copies/ml, respectively, after 10 days of treatment (P<0.0001 versus placebo). ATC showed a low propensity to select for resistance mutants in vitro and during clinical monotherapy. ATC was well tolerated in volunteers and in HIV-infected patients. This promising profile suggests that ATC may be useful in treating patients who have failed previous lamivudine- or emtricitabine-containing regimens. Further studies to evaluate the long-term efficacy and tolerability of ATC are underway.


Subject(s)
Anti-HIV Agents/therapeutic use , Deoxycytidine/analogs & derivatives , HIV Infections/drug therapy , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Resistance, Viral , Humans
20.
AIDS ; 20(9): 1261-8, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16816554

ABSTRACT

OBJECTIVE: Apricitabine (formerly AVX754 and SPD754) is a deoxycytidine analogue nucleoside reverse transcriptase inhibitor in clinical development for patients with HIV disease. This study evaluated the antiretroviral efficacy, tolerability and safety of apricitabine monotherapy, administered for 10 days in antiretroviral-naive, HIV-1 infected adults. METHODS: Adult patients (> or = 18 years) with HIV infection (CD4 count > or = 250 cells/microl; plasma HIV-1 RNA level 5000-100 000 copies/ml) were randomized to 10 days' double-blind oral therapy with placebo or apricitabine 400 mg/day, 800 mg/day, 1200 mg/day, or 1600 mg/day. RESULTS: At 7 days, all apricitabine doses produced statistically significant log10 reductions in plasma HIV RNA levels from baseline relative to placebo (n = 13; P < 0.0001), as follows: -1.16 (400 mg; n = 11), -1.28 (800 mg; n = 12), -1.44 (1200 mg; n = 14), -1.30 (1600 mg; n = 13). After 10 days, the log10 viral load reductions with apricitabine 1200 mg (-1.65; P = 0.01) and 1600 mg/day (-1.58; P = 0.04) were significantly greater than that with the 400-mg dose (-1.18). No clinically relevant changes were observed in CD4 or CD8 cell indices. Apricitabine was well tolerated and showed no tendency to select any particular resistance mutation. CONCLUSION: Apricitabine monotherapy showed promising antiretroviral efficacy, good tolerability and a low propensity for resistance selection in antiretroviral-naive HIV-infected patients treated for 10 days. These results warrant further evaluation of the long-term clinical efficacy and tolerability of apricitabine.


Subject(s)
Deoxycytidine/analogs & derivatives , HIV Infections/drug therapy , HIV-1 , Reverse Transcriptase Inhibitors/therapeutic use , Administration, Oral , Adult , Argentina , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , HIV Infections/blood , HIV Infections/immunology , HIV-1/genetics , Humans , Lymphocyte Count , Male , Mutation , RNA, Viral/blood , South Africa , Thailand , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL