Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Med Chem ; 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39018526

ABSTRACT

Acute respiratory viral infections, such as pneumovirus and respiratory picornavirus infections, exacerbate disease in COPD and asthma patients. A research program targeting respiratory syncytial virus (RSV) led to the discovery of GS-7682 (1), a novel phosphoramidate prodrug of a 4'-CN-4-aza-7,9-dideazaadenosine C-nucleoside GS-646089 (2) with broad antiviral activity against RSV (EC50 = 3-46 nM), human metapneumovirus (EC50 = 210 nM), human rhinovirus (EC50 = 54-61 nM), and enterovirus (EC50 = 83-90 nM). Prodrug optimization for cellular potency and lung cell metabolism identified 5'-methyl [(S)-hydroxy(phenoxy)phosphoryl]-l-alaninate in combination with 2',3'-diisobutyrate promoieties as being optimal for high levels of intracellular triphosphate formation in vitro and in vivo. 1 demonstrated significant reductions of viral loads in the lower respiratory tract of RSV-infected African green monkeys when administered once daily via intratracheal nebulized aerosol. Together, these findings support additional evaluation of 1 and its analogues as potential therapeutics for pneumo- and picornaviruses.

2.
J Med Chem ; 64(8): 5001-5017, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33835812

ABSTRACT

A discovery program targeting respiratory syncytial virus (RSV) identified C-nucleoside 4 (RSV A2 EC50 = 530 nM) as a phenotypic screening lead targeting the RSV RNA-dependent RNA polymerase (RdRp). Prodrug exploration resulted in the discovery of remdesivir (1, GS-5734) that is >30-fold more potent than 4 against RSV in HEp-2 and NHBE cells. Metabolism studies in vitro confirmed the rapid formation of the active triphosphate metabolite, 1-NTP, and in vivo studies in cynomolgus and African Green monkeys demonstrated a >10-fold higher lung tissue concentration of 1-NTP following molar normalized IV dosing of 1 compared to that of 4. A once daily 10 mg/kg IV administration of 1 in an African Green monkey RSV model demonstrated a >2-log10 reduction in the peak lung viral load. These early data following the discovery of 1 supported its potential as a novel treatment for RSV prior to its development for Ebola and approval for COVID-19 treatment.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/pharmacology , Prodrugs/pharmacology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Adenosine Monophosphate/pharmacology , Alanine/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Caco-2 Cells , Cells, Cultured , Chlorocebus aethiops , Disease Models, Animal , Dogs , Drug Evaluation, Preclinical/methods , Epithelial Cells/virology , Humans , Macaca fascicularis , Male , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats, Sprague-Dawley , Respiratory Syncytial Virus Infections/virology , Structure-Activity Relationship , Tissue Distribution , Tubercidin/analogs & derivatives , Tubercidin/chemistry , Viral Load
3.
J Infect Dis ; 222(9): 1468-1477, 2020 10 01.
Article in English | MEDLINE | ID: mdl-31971597

ABSTRACT

BACKGROUND: Presatovir is an oral respiratory syncytial virus (RSV) fusion inhibitor targeting RSV F protein. In a double-blind, placebo-controlled study in healthy adults experimentally infected with RSV (Memphis-37b), presatovir significantly reduced viral load and clinical disease severity in a dose-dependent manner. METHODS: Viral RNA from nasal wash samples was amplified and the F gene sequenced to monitor presatovir resistance. Effects of identified amino acid substitutions on in vitro susceptibility to presatovir, viral fitness, and clinical outcome were assessed. RESULTS: Twenty-eight treatment-emergent F substitutions were identified. Of these, 26 were tested in vitro; 2 were not due to lack of recombinant virus recovery. Ten substitutions did not affect presatovir susceptibility, and 16 substitutions reduced RSV susceptibility to presatovir (2.9- to 410-fold). No substitutions altered RSV susceptibility to palivizumab or ribavirin. Frequency of phenotypically resistant substitutions was higher with regimens containing lower presatovir dose and shorter treatment duration. Participants with phenotypic presatovir resistance had significantly higher nasal viral load area under the curve relative to those without, but substitutions did not significantly affect peak viral load or clinical manifestations of RSV disease. CONCLUSIONS: Emergence of presatovir-resistant RSV occurred during therapy but did not significantly affect clinical efficacy in participants with experimental RSV infection.


Subject(s)
Indazoles/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Sulfonamides/therapeutic use , Viral Fusion Protein Inhibitors/therapeutic use , Adolescent , Adult , Amino Acid Substitution , Dose-Response Relationship, Drug , Double-Blind Method , Drug Resistance, Viral/genetics , Humans , Middle Aged , Respiratory Syncytial Viruses/genetics , Viral Load/drug effects , Young Adult
4.
J Med Chem ; 60(5): 1648-1661, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28124907

ABSTRACT

The recent Ebola virus (EBOV) outbreak in West Africa was the largest recorded in history with over 28,000 cases, resulting in >11,000 deaths including >500 healthcare workers. A focused screening and lead optimization effort identified 4b (GS-5734) with anti-EBOV EC50 = 86 nM in macrophages as the clinical candidate. Structure activity relationships established that the 1'-CN group and C-linked nucleobase were critical for optimal anti-EBOV potency and selectivity against host polymerases. A robust diastereoselective synthesis provided sufficient quantities of 4b to enable preclinical efficacy in a non-human-primate EBOV challenge model. Once-daily 10 mg/kg iv treatment on days 3-14 postinfection had a significant effect on viremia and mortality, resulting in 100% survival of infected treated animals [ Nature 2016 , 531 , 381 - 385 ]. A phase 2 study (PREVAIL IV) is currently enrolling and will evaluate the effect of 4b on viral shedding from sanctuary sites in EBOV survivors.


Subject(s)
Alanine/analogs & derivatives , Amides/chemistry , Hemorrhagic Fever, Ebola/drug therapy , Phosphoric Acids/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Ribonucleotides/chemistry , Virus Diseases/drug therapy , Adenosine Monophosphate/analogs & derivatives , Alanine/chemistry , Cell Line , Drug Discovery , Humans , Microbial Sensitivity Tests , Prodrugs/chemical synthesis , Structure-Activity Relationship
5.
Antiviral Res ; 140: 116-120, 2017 04.
Article in English | MEDLINE | ID: mdl-28131805

ABSTRACT

HIV-infected patients treated with certain nucleoside reverse transcriptase inhibitors (NRTIs) have experienced adverse effects due to drug-related mitochondrial toxicity. Tenofovir alafenamide (TAF) is a novel prodrug of the NRTI tenofovir (TFV) with an improved safety profile compared to tenofovir disoproxil fumarate (TDF). Prior in vitro studies have demonstrated that the parent nucleotide TFV has no significant effects on mtDNA synthesis. This study investigated whether clinically relevant TAF and TDF exposures affect mtDNA content in human lymphocytes. First, activated or resting peripheral blood mononuclear cells (PBMCs), as well as MT-2 and Jurkat T-cell lines, were continuously treated with ddC for 10 days to establish their susceptibility to mtDNA depletion. PBMCs had low sensitivity to NRTI-mediated mtDNA depletion in vitro. In contrast, ddC treatment of rapidly dividing MT-2 and Jurkat cells resulted in a dose-dependent decrease in mtDNA. Therefore, these two T-cell lines were selected for evaluating TAF and TDF treatment effects. MT-2 and Jurkat cells were pulse-treated with TAF or TDF every 24 h for 10 days to mimic pharmacologically relevant drug exposures. Pulse treatment of cells with 3.3 µM TAF or 1.1 µM TDF for 10 days resulted in 2- to 7-fold greater steady-state intracellular TFV-diphosphate (TFV-DP) levels than those observed clinically in TAF- or TDF-treated patients. At these concentrations, no significant TAF- (106.7% and 84.1% of control; p = 0.77 and 0.12 for MT-2 and Jurkat, respectively) or TDF- (100.6% and 91.0% of control; p = 0.91 and 0.37, respectively) associated reduction in mtDNA content was observed compared with untreated control cells. This study demonstrates that, despite delivering higher intracellular levels of TFV-DP than TDF, TAF does not inhibit mtDNA synthesis in vitro at concentrations exceeding the clinically relevant intracellular drug exposures. Thus, TAF has a low potential for mitochondrial toxicity in T-cells of HIV-infected patients.


Subject(s)
Anti-HIV Agents/pharmacology , DNA, Mitochondrial/metabolism , Leukocytes, Mononuclear/drug effects , T-Lymphocytes/drug effects , Tenofovir/pharmacology , Anti-HIV Agents/adverse effects , Anti-HIV Agents/analysis , Anti-HIV Agents/pharmacokinetics , Cytoplasm/chemistry , DNA, Mitochondrial/biosynthesis , HIV Infections/drug therapy , HIV-1/drug effects , Humans , Jurkat Cells , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/cytology , Prodrugs/administration & dosage , Prodrugs/analysis , Prodrugs/pharmacology , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , Tenofovir/adverse effects , Tenofovir/analysis , Tenofovir/pharmacokinetics
6.
Antimicrob Agents Chemother ; 60(12): 7086-7097, 2016 12.
Article in English | MEDLINE | ID: mdl-27645238

ABSTRACT

Bictegravir (BIC; GS-9883), a novel, potent, once-daily, unboosted inhibitor of HIV-1 integrase (IN), specifically targets IN strand transfer activity (50% inhibitory concentration [IC50] of 7.5 ± 0.3 nM) and HIV-1 integration in cells. BIC exhibits potent and selective in vitro antiretroviral activity in both T-cell lines and primary human T lymphocytes, with 50% effective concentrations ranging from 1.5 to 2.4 nM and selectivity indices up to 8,700 relative to cytotoxicity. BIC exhibits synergistic in vitro antiviral effects in pairwise combinations with tenofovir alafenamide, emtricitabine, or darunavir and maintains potent antiviral activity against HIV-1 variants resistant to other classes of antiretrovirals. BIC displayed an in vitro resistance profile that was markedly improved compared to the integrase strand transfer inhibitors (INSTIs) raltegravir (RAL) and elvitegravir (EVG), and comparable to that of dolutegravir (DTG), against nine INSTI-resistant site-directed HIV-1 mutants. BIC displayed statistically improved antiviral activity relative to EVG, RAL, and DTG against a panel of 47 patient-derived HIV-1 isolates with high-level INSTI resistance; 13 of 47 tested isolates exhibited >2-fold lower resistance to BIC than DTG. In dose-escalation experiments conducted in vitro, BIC and DTG exhibited higher barriers to resistance than EVG, selecting for HIV-1 variants with reduced phenotypic susceptibility at days 71, 87, and 20, respectively. A recombinant virus with the BIC-selected M50I/R263K dual mutations in IN exhibited only 2.8-fold reduced susceptibility to BIC compared to wild-type virus. All BIC-selected variants exhibited low to intermediate levels of cross-resistance to RAL, DTG, and EVG (<8-fold) but remained susceptible to other classes of antiretrovirals. A high barrier to in vitro resistance emergence for both BIC and DTG was also observed in viral breakthrough studies in the presence of constant clinically relevant drug concentrations. The overall virologic profile of BIC supports its ongoing clinical investigation in combination with other antiretroviral agents for both treatment-naive and -experienced HIV-infected patients.


Subject(s)
Drug Resistance, Viral/drug effects , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Amides , Anti-HIV Agents/pharmacology , Cell Line , Drug Synergism , HIV Integrase/genetics , HIV-1/genetics , HIV-1/isolation & purification , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Mutation , Oxazines , Piperazines , Pyridones , Raltegravir Potassium/pharmacology
7.
Nature ; 531(7594): 381-5, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26934220

ABSTRACT

The most recent Ebola virus outbreak in West Africa, which was unprecedented in the number of cases and fatalities, geographic distribution, and number of nations affected, highlights the need for safe, effective, and readily available antiviral agents for treatment and prevention of acute Ebola virus (EBOV) disease (EVD) or sequelae. No antiviral therapeutics have yet received regulatory approval or demonstrated clinical efficacy. Here we report the discovery of a novel small molecule GS-5734, a monophosphoramidate prodrug of an adenosine analogue, with antiviral activity against EBOV. GS-5734 exhibits antiviral activity against multiple variants of EBOV and other filoviruses in cell-based assays. The pharmacologically active nucleoside triphosphate (NTP) is efficiently formed in multiple human cell types incubated with GS-5734 in vitro, and the NTP acts as an alternative substrate and RNA-chain terminator in primer-extension assays using a surrogate respiratory syncytial virus RNA polymerase. Intravenous administration of GS-5734 to nonhuman primates resulted in persistent NTP levels in peripheral blood mononuclear cells (half-life, 14 h) and distribution to sanctuary sites for viral replication including testes, eyes, and brain. In a rhesus monkey model of EVD, once-daily intravenous administration of 10 mg kg(-1) GS-5734 for 12 days resulted in profound suppression of EBOV replication and protected 100% of EBOV-infected animals against lethal disease, ameliorating clinical disease signs and pathophysiological markers, even when treatments were initiated three days after virus exposure when systemic viral RNA was detected in two out of six treated animals. These results show the first substantive post-exposure protection by a small-molecule antiviral compound against EBOV in nonhuman primates. The broad-spectrum antiviral activity of GS-5734 in vitro against other pathogenic RNA viruses, including filoviruses, arenaviruses, and coronaviruses, suggests the potential for wider medical use. GS-5734 is amenable to large-scale manufacturing, and clinical studies investigating the drug safety and pharmacokinetics are ongoing.


Subject(s)
Alanine/analogs & derivatives , Antiviral Agents/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Macaca mulatta/virology , Ribonucleotides/therapeutic use , Adenosine Monophosphate/analogs & derivatives , Alanine/pharmacokinetics , Alanine/pharmacology , Alanine/therapeutic use , Amino Acid Sequence , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cell Line, Tumor , Ebolavirus/drug effects , Female , HeLa Cells , Hemorrhagic Fever, Ebola/prevention & control , Humans , Male , Molecular Sequence Data , Organ Specificity , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/therapeutic use , Ribonucleotides/pharmacokinetics , Ribonucleotides/pharmacology
8.
Antimicrob Agents Chemother ; 60(1): 316-22, 2016 01.
Article in English | MEDLINE | ID: mdl-26503655

ABSTRACT

Tenofovir alafenamide fumarate (TAF) is an oral phosphonoamidate prodrug of the HIV reverse transcriptase nucleotide inhibitor tenofovir (TFV). Previous studies suggested a principal role for the lysosomal serine protease cathepsin A (CatA) in the intracellular activation of TAF. Here we further investigated the role of CatA and other human hydrolases in the metabolism of TAF. Overexpression of CatA or liver carboxylesterase 1 (Ces1) in HEK293T cells increased intracellular TAF hydrolysis 2- and 5-fold, respectively. Knockdown of CatA expression with RNA interference (RNAi) in HeLa cells reduced intracellular TAF metabolism 5-fold. Additionally, the anti-HIV activity and the rate of CatA hydrolysis showed good correlation within a large set of TFV phosphonoamidate prodrugs. The covalent hepatitis C virus (HCV) protease inhibitors (PIs) telaprevir and boceprevir potently inhibited CatA-mediated TAF activation (50% inhibitory concentration [IC50] = 0.27 and 0.16 µM, respectively) in vitro and also reduced its anti-HIV activity in primary human CD4(+) T lymphocytes (21- and 3-fold, respectively) at pharmacologically relevant concentrations. In contrast, there was no inhibition of CatA or any significant effect on anti-HIV activity of TAF observed with cobicistat, noncovalent HIV and HCV PIs, or various prescribed inhibitors of host serine proteases. Collectively, these studies confirm that CatA plays a pivotal role in the intracellular metabolism of TAF, whereas the liver esterase Ces1 likely contributes to the hepatic activation of TAF. Moreover, this work demonstrates that a wide range of viral and host PIs, with the exception of telaprevir and boceprevir, do not interfere with the antiretroviral activity of TAF.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/metabolism , CD4-Positive T-Lymphocytes/drug effects , Prodrugs/metabolism , Tenofovir/metabolism , Adenine/metabolism , Adenine/pharmacology , Alanine , Anti-HIV Agents/pharmacology , Biotransformation , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/virology , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cathepsin A/antagonists & inhibitors , Cathepsin A/genetics , Cathepsin A/metabolism , Cobicistat/pharmacology , Drug Interactions , Gene Expression , HEK293 Cells , HIV-1/drug effects , HIV-1/growth & development , HeLa Cells , Host-Pathogen Interactions , Humans , Oligopeptides/pharmacology , Primary Cell Culture , Prodrugs/pharmacology , Proline/analogs & derivatives , Proline/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serine Proteinase Inhibitors/pharmacology , Tenofovir/pharmacology
9.
Antimicrob Agents Chemother ; 60(3): 1264-73, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26666922

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections in infants and young children. In addition, RSV causes significant morbidity and mortality in hospitalized elderly and immunocompromised patients. Currently, only palivizumab, a monoclonal antibody against the RSV fusion (F) protein, and inhaled ribavirin are approved for the prophylactic and therapeutic treatment of RSV, respectively. Therefore, there is a clinical need for safe and effective therapeutic agents for RSV infections. GS-5806, discovered via chemical optimization of a hit from a high-throughput antiviral-screening campaign, selectively inhibits a diverse set of 75 RSV subtype A and B clinical isolates (mean 50% effective concentration [EC50] = 0.43 nM). The compound maintained potency in primary human airway epithelial cells and exhibited low cytotoxicity in human cell lines and primary cell cultures (selectivity > 23,000-fold). Time-of-addition and temperature shift studies demonstrated that GS-5806 does not block RSV attachment to cells but interferes with virus entry. Follow-up experiments showed potent inhibition of RSV F-mediated cell-to-cell fusion. RSV A and B variants resistant to GS-5806, due to mutations in F protein (RSV A, L138F or F140L/N517I, and RSV B, F488L or F488S), were isolated and showed cross-resistance to other RSV fusion inhibitors, such as VP-14637, but remained fully sensitive to palivizumab and ribavirin. In summary, GS-5806 is a potent and selective RSV fusion inhibitor with antiviral activity against a diverse set of RSV clinical isolates. The compound is currently under clinical investigation for the treatment of RSV infection in pediatric, immunocompromised, and elderly patients.


Subject(s)
Antiviral Agents/pharmacology , Pyrazoles/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Sulfonamides/pharmacology , Bronchi/cytology , Bronchi/virology , Cell Fusion , Cell Line , Drug Evaluation, Preclinical/methods , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Humans , Indazoles , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Syncytial Virus, Human/pathogenicity , Virus Internalization/drug effects
10.
J Med Chem ; 58(4): 1630-43, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25574686

ABSTRACT

GS-5806 is a novel, orally bioavailable RSV fusion inhibitor discovered following a lead optimization campaign on a screening hit. The oral absorption properties were optimized by converting to the pyrazolo[1,5-a]-pyrimidine heterocycle, while potency, metabolic, and physicochemical properties were optimized by introducing the para-chloro and aminopyrrolidine groups. A mean EC50 = 0.43 nM was found toward a panel of 75 RSV A and B clinical isolates and dose-dependent antiviral efficacy in the cotton rat model of RSV infection. Oral bioavailability in preclinical species ranged from 46 to 100%, with evidence of efficient penetration into lung tissue. In healthy human volunteers experimentally infected with RSV, a potent antiviral effect was observed with a mean 4.2 log10 reduction in peak viral load and a significant reduction in disease severity compared to placebo. In conclusion, a potent, once daily, oral RSV fusion inhibitor with the potential to treat RSV infection in infants and adults is reported.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Pyrazoles/pharmacology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/drug effects , Sulfonamides/pharmacology , Virus Internalization/drug effects , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Humans , Indazoles , Macaca fascicularis , Microbial Sensitivity Tests , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Rats , Respiratory Syncytial Viruses/physiology , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemistry
11.
PLoS Pathog ; 10(4): e1004071, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24722454

ABSTRACT

Persistent latent reservoir of replication-competent proviruses in memory CD4 T cells is a major obstacle to curing HIV infection. Pharmacological activation of HIV expression in latently infected cells is being explored as one of the strategies to deplete the latent HIV reservoir. In this study, we characterized the ability of romidepsin (RMD), a histone deacetylase inhibitor approved for the treatment of T-cell lymphomas, to activate the expression of latent HIV. In an in vitro T-cell model of HIV latency, RMD was the most potent inducer of HIV (EC50 = 4.5 nM) compared with vorinostat (VOR; EC50 = 3,950 nM) and other histone deacetylase (HDAC) inhibitors in clinical development including panobinostat (PNB; EC50 = 10 nM). The HIV induction potencies of RMD, VOR, and PNB paralleled their inhibitory activities against multiple human HDAC isoenzymes. In both resting and memory CD4 T cells isolated from HIV-infected patients on suppressive combination antiretroviral therapy (cART), a 4-hour exposure to 40 nM RMD induced a mean 6-fold increase in intracellular HIV RNA levels, whereas a 24-hour treatment with 1 µM VOR resulted in 2- to 3-fold increases. RMD-induced intracellular HIV RNA expression persisted for 48 hours and correlated with sustained inhibition of cell-associated HDAC activity. By comparison, the induction of HIV RNA by VOR and PNB was transient and diminished after 24 hours. RMD also increased levels of extracellular HIV RNA and virions from both memory and resting CD4 T-cell cultures. The activation of HIV expression was observed at RMD concentrations below the drug plasma levels achieved by doses used in patients treated for T-cell lymphomas. In conclusion, RMD induces HIV expression ex vivo at concentrations that can be achieved clinically, indicating that the drug may reactivate latent HIV in patients on suppressive cART.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Depsipeptides/pharmacology , Gene Expression Regulation, Viral/drug effects , HIV Infections/drug therapy , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Models, Biological , Virus Activation/drug effects , Virus Latency/drug effects , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Depsipeptides/pharmacokinetics , Dose-Response Relationship, Drug , Female , HIV Infections/virology , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases/metabolism , Humans , Immunologic Memory/drug effects , Isoenzymes/metabolism , Male
12.
Bioorg Med Chem Lett ; 24(3): 995-9, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24412072

ABSTRACT

The HIV protease inhibitor (PI) ritonavir (RTV) has been widely used as a pharmacoenhancer for other PIs, which are substrates of cytochrome P450 3A (CYP3A). However the potent anti-HIV activity of ritonavir may limit its use as a pharmacoenhancer with other classes of anti-HIV agents. Ritonavir is also associated with limitations such as poor physicochemical properties. To address these issues a series of compounds with replacements at the P2 and/or P3 region was designed and evaluated as novel CYP3A inhibitors. Through these efforts, a potent and selective inhibitor of CYP3A, GS-9350 (cobicistat) with improved physiochemical properties was discovered.


Subject(s)
Carbamates/chemistry , Cytochrome P-450 CYP3A Inhibitors , Diamines/chemistry , Diamines/pharmacology , Thiazoles/chemistry , Carbamates/pharmacology , Cobicistat , Enzyme Activation/drug effects , Molecular Structure , Structure-Activity Relationship , Thiazoles/pharmacology
13.
Bioorg Med Chem Lett ; 24(3): 989-94, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24411125

ABSTRACT

Ritonavir (RTV), an HIV-1 protease inhibitor (PI), is also a potent mechanism-based inhibitor of human cytochrome P450 3A (CYP3A) and has been widely prescribed as a pharmacoenhancer. As a boosting agent for marketed PIs, it reduces pill burden, and improves compliance. Removal of the hydroxyl group from RTV reduces, but does not eliminate HIV PI activity and does not affect CYP3A inhibition. Herein we report the discovery of a novel series of CYP3A inhibitors that are devoid of antiviral activity. The synthesis and evaluation of analogs with extensive modifications of the 1,4-diamine core along with the structure activity relationships with respect to anti-HIV activity, CYP3A inhibitory activity, selectivity against other CYP enzymes and the human pregnane X receptor (PXR) will be discussed.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Diamines/chemical synthesis , Diamines/pharmacology , HIV/drug effects , Diamines/chemistry , Enzyme Activation/drug effects , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , Humans , Structure-Activity Relationship , Treatment Outcome
14.
J Virol ; 88(6): 3586-90, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371077

ABSTRACT

Insertions in the protease (PR) region of human immunodeficiency virus (HIV) represent an interesting mechanism of antiviral resistance against HIV PR inhibitors (PIs). Here, we demonstrate the improved ability of a phosphonate-containing experimental HIV PI, GS-8374, relative to that of other PIs, to effectively inhibit patient-derived recombinant HIV strains bearing PR insertions and numerous other mutations. We correlate enzyme inhibition with the catalytic activities of corresponding recombinant PRs in vitro and provide a biochemical and structural analysis of the PR-inhibitor complex.


Subject(s)
HIV Infections/virology , HIV Protease Inhibitors/chemistry , HIV Protease/genetics , HIV-1/drug effects , HIV-1/enzymology , Mutagenesis, Insertional , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Drug Resistance, Viral , HIV Infections/drug therapy , HIV Protease/chemistry , HIV Protease/metabolism , HIV Protease Inhibitors/pharmacology , HIV-1/chemistry , HIV-1/genetics , Humans , Models, Molecular , Organophosphonates/analysis
15.
PLoS One ; 8(9): e74163, 2013.
Article in English | MEDLINE | ID: mdl-24040198

ABSTRACT

HIV-1 integrase (IN) is the target for two classes of antiretrovirals: i) the integrase strand-transfer inhibitors (INSTIs) and ii) the non-catalytic site integrase inhibitors (NCINIs). NCINIs bind at the IN dimer interface and are thought to interfere primarily with viral DNA (vDNA) integration in the target cell by blocking IN-vDNA assembly as well as the IN-LEDGF/p75 interaction. Herein we show that treatment of virus-producing cells, but not of mature virions or target cells, drives NCINI antiviral potency. NCINIs target an essential late-stage event in HIV replication that is insensitive to LEDGF levels in the producer cells. Virus particles produced in the presence of NCINIs displayed normal Gag-Pol processing and endogenous reverse transcriptase activity, but were defective at initiating vDNA synthesis following entry into the target cell. NCINI-resistant virus carrying a T174I mutation in the IN dimer interface was less sensitive to the compound-induced late-stage effects, including the reverse transcription block. Wild-type, but not T174I virus, produced in the presence of NCINIs exhibited striking defects in core morphology and an increased level of IN oligomers that was not observed upon treatment of mature cell-free particles. Collectively, these results reveal that NCINIs act through a novel mechanism that is unrelated to the previously observed inhibition of IN activity or IN-LEDGF interaction, and instead involves the disruption of an IN function during HIV-1 core maturation and assembly.


Subject(s)
DNA, Viral/antagonists & inhibitors , HIV Integrase Inhibitors/pharmacology , HIV Integrase/genetics , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Virus Replication/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , DNA, Viral/genetics , Drug Resistance, Viral , Gene Expression , Genes, Reporter , Genetic Vectors , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , HIV-1/growth & development , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Luciferases/genetics , Luciferases/metabolism , Mutation , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , T-Lymphocytes/virology , Transcription Factors/genetics , Transcription Factors/metabolism , Virion/drug effects , Virion/genetics , Virus Assembly/drug effects , Virus Integration/drug effects
16.
Antimicrob Agents Chemother ; 57(10): 4982-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23896476

ABSTRACT

A once-daily single-tablet antiretroviral regimen containing tenofovir (TFV) disoproxil fumarate, emtricitabine (FTC), elvitegravir (EVG), and cobicistat (COBI) is an approved combination for the treatment of patients infected with HIV. COBI and TFV have been reported to interact with distinct transporters in renal proximal tubules; while TFV is renally eliminated by a combination of glomerular filtration and tubular secretion via anion transporters OAT1, OAT3, and MRP4, COBI inhibits renal cation transporters, particularly MATE1, resulting in a measurable decrease in the tubular secretion of creatinine. To investigate the potential for a renal drug-drug interaction between TFV and COBI in vitro, the uptake of TFV in the presence and absence of COBI was determined in fresh human renal cortex tissue and in cells expressing the relevant renal transporters. At concentrations exceeding clinical protein-unbound plasma levels, COBI did not significantly inhibit the transport of TFV by the anion transporters OAT1, OAT3, and MRP4 (50% inhibitory concentrations [IC50s] of >15, 6.6, and 8.5 µM, respectively). Conversely, TFV had little or no effect on the cation transporters OCT2 and MATE1 (IC50 > 100 µM). Consistent with studies using individual transporters, no increase in the accumulation of TFV in freshly isolated human renal cortex tissue or renal proximal tubule cells (RPTECs) was observed in the presence of COBI. Finally, COBI alone or in combination with FTC and EVG did not affect the sensitivity to TFV of cultured primary RPTECs or cells coexpressing OAT1 and MRP4. These results illustrate that COBI and TFV interact primarily with distinct renal transporters and indicate a low potential for pharmacokinetic renal drug-drug interaction.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacology , Carbamates/pharmacology , Organophosphonates/pharmacology , Thiazoles/pharmacology , Adenine/pharmacology , Adult , Cell Line , Cobicistat , Drug Interactions , Humans , In Vitro Techniques , Kidney/drug effects , Kidney/metabolism , Male , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2 , Tenofovir
17.
J Virol ; 87(1): 454-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23097440

ABSTRACT

GS-8374 is a potent HIV protease inhibitor (PI) with a unique diethyl-phosphonate moiety. Due to a balanced contribution of enthalpic and entropic components to its interaction with the protease (PR) active site, the compound retains activity against HIV mutants with high-level multi-PI resistance. We report here the in vitro selection and characterization of HIV variants resistant to GS-8374. While highly resistant viruses with multiple mutations in PR were isolated in the presence of control PIs, an HIV variant displaying moderate (14-fold) resistance to GS-8374 was generated only after prolonged passaging for >300 days. The isolate showed low-level cross-resistance to darunavir, atazanavir, lopinavir, and saquinavir, but not other PIs, and contained a single R41K mutation in PR combined with multiple genotypic changes in the Gag matrix, capsid, nucleocapsid, and SP2 domains. Mutations also occurred in the transframe peptide and p6* domain of the Gag-Pol polyprotein. Analysis of recombinant HIV variants indicated that mutations in Gag, but not the R41K in PR, conferred reduced susceptibility to GS-8374. The Gag mutations acted in concert, since they did not affect susceptibility when introduced individually. Analysis of viral particles revealed that the mutations rendered Gag more susceptible to PR-mediated cleavage in the presence of GS-8374. In summary, the emergence of resistance to GS-8374 involved a combination of substrate mutations without typical resistance mutations in PR. These substrate changes were distributed throughout Gag and acted in an additive manner. Thus, they are classified as primary resistance mutations indicating a unique mechanism and pathway of resistance development for GS-8374.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Mutation, Missense , gag Gene Products, Human Immunodeficiency Virus/genetics , Amino Acid Substitution , Cell Line , HIV-1/genetics , HIV-1/growth & development , HIV-1/isolation & purification , Humans , Selection, Genetic , Serial Passage
18.
J Med Chem ; 54(15): 5498-507, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21711054

ABSTRACT

A series of 7-aryl- and 7-hetaryl-7-deazaadenosines was prepared by the cross-coupling reactions of unprotected or protected 7-iodo-7-deazaadenosines with (het)arylboronic acids, stannanes, or zinc halides. Nucleosides bearing 5-membered heterocycles at the position 7 exerted potent in vitro antiproliferative effects against a broad panel of hematological and solid tumor cell lines. Cell cycle analysis indicated profound inhibition of RNA synthesis and induction of apoptosis in treated cells. Intracellular conversion to triphosphates has been detected with active compounds. The triphosphate metabolites showed only a weak inhibitory effect on human RNA polymerase II, suggesting potentially other mechanisms for the inhibition of RNA synthesis and quick onset of apoptosis. Initial in vivo evaluation demonstrated an effect of 7-(2-thienyl)-7-deazaadenine ribonucleoside on the survival rate in syngeneic P388D1 mouse leukemia model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cytostatic Agents/chemical synthesis , Cytostatic Agents/pharmacology , Tubercidin/analogs & derivatives , Adenosine Kinase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Humans , Mice , RNA Polymerase II/antagonists & inhibitors , Tubercidin/chemical synthesis , Tubercidin/pharmacology
19.
Antimicrob Agents Chemother ; 55(4): 1366-76, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21245449

ABSTRACT

GS-8374 is a novel bis-tetrahydrofuran HIV-1 protease (PR) inhibitor (PI) with a unique diethylphosphonate moiety. It was selected from a series of analogs containing various di(alkyl)phosphonate substitutions connected via a linker to the para position of a P-1 phenyl ring. GS-8374 inhibits HIV-1 PR with high potency (K(i) = 8.1 pM) and with no known effect on host proteases. Kinetic and thermodynamic analysis of GS-8374 binding to PR demonstrated an extremely slow off rate for the inhibitor and favorable contributions of both the enthalpic and entropic components to the total free binding energy. GS-8374 showed potent antiretroviral activity in T-cell lines, primary CD4(+) T cells (50% effective concentration [EC(50)] = 3.4 to 11.5 nM), and macrophages (EC(50) = 25.5 nM) and exhibited low cytotoxicity in multiple human cell types. The antiviral potency of GS-8374 was only moderately affected by human serum protein binding, and its combination with multiple approved antiretrovirals showed synergistic effects. When it was tested in a PhenoSense assay against a panel of 24 patient-derived viruses with high-level PI resistance, GS-8374 showed lower mean EC(50)s and lower fold resistance than any of the clinically approved PIs. Similar to other PIs, in vitro hepatic microsomal metabolism of GS-8374 was efficiently blocked by ritonavir, suggesting a potential for effective pharmacokinetic boosting in vivo. In summary, results from this broad in vitro pharmacological profiling indicate that GS-8374 is a promising candidate to be further assessed as a new antiretroviral agent with potential for clinical efficacy in both treatment-naïve and -experienced patients.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , HIV Protease/chemistry , HIV Protease/metabolism , Organophosphonates/chemistry , CD4-Positive T-Lymphocytes/drug effects , Calorimetry , Cells, Cultured , Crystallography, X-Ray , HEK293 Cells , HIV-1/drug effects , Hep G2 Cells , Humans , Molecular Structure , Proteasome Endopeptidase Complex/metabolism
20.
ACS Med Chem Lett ; 1(5): 209-13, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-24900196

ABSTRACT

Cobicistat (3, GS-9350) is a newly discovered, potent, and selective inhibitor of human cytochrome P450 3A (CYP3A) enzymes. In contrast to ritonavir, 3 is devoid of anti-HIV activity and is thus more suitable for use in boosting anti-HIV drugs without risking selection of potential drug-resistant HIV variants. Compound 3 shows reduced liability for drug interactions and may have potential improvements in tolerability over ritonavir. In addition, 3 has high aqueous solubility and can be readily coformulated with other agents.

SELECTION OF CITATIONS
SEARCH DETAIL
...