Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nature ; 584(7822): 614-618, 2020 08.
Article in English | MEDLINE | ID: mdl-32612233

ABSTRACT

Oral antiretroviral agents provide life-saving treatments for millions of people living with HIV, and can prevent new infections via pre-exposure prophylaxis1-5. However, some people living with HIV who are heavily treatment-experienced have limited or no treatment options, owing to multidrug resistance6. In addition, suboptimal adherence to oral daily regimens can negatively affect the outcome of treatment-which contributes to virologic failure, resistance generation and viral transmission-as well as of pre-exposure prophylaxis, leading to new infections1,2,4,7-9. Long-acting agents from new antiretroviral classes can provide much-needed treatment options for people living with HIV who are heavily treatment-experienced, and additionally can improve adherence10. Here we describe GS-6207, a small molecule that disrupts the functions of HIV capsid protein and is amenable to long-acting therapy owing to its high potency, low in vivo systemic clearance and slow release kinetics from the subcutaneous injection site. Drawing on X-ray crystallographic information, we designed GS-6207 to bind tightly at a conserved interface between capsid protein monomers, where it interferes with capsid-protein-mediated interactions between proteins that are essential for multiple phases of the viral replication cycle. GS-6207 exhibits antiviral activity at picomolar concentrations against all subtypes of HIV-1 that we tested, and shows high synergy and no cross-resistance with approved antiretroviral drugs. In phase-1 clinical studies, monotherapy with a single subcutaneous dose of GS-6207 (450 mg) resulted in a mean log10-transformed reduction of plasma viral load of 2.2 after 9 days, and showed sustained plasma exposure at antivirally active concentrations for more than 6 months. These results provide clinical validation for therapies that target the functions of HIV capsid protein, and demonstrate the potential of GS-6207 as a long-acting agent to treat or prevent infection with HIV.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , Adolescent , Adult , Anti-HIV Agents/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cells, Cultured , Drug Resistance, Viral/genetics , Female , HIV-1/growth & development , Humans , Male , Middle Aged , Models, Molecular , Virus Replication/drug effects , Young Adult
2.
Bioorg Med Chem Lett ; 29(16): 2415-2427, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31230974

ABSTRACT

Direct-acting antiviral inhibitors have revolutionized the treatment of hepatitis C virus (HCV) infected patients. Herein is described the discovery of velpatasvir (VEL, GS-5816), a potent pan-genotypic HCV NS5A inhibitor that is a component of the only approved pan-genotypic single-tablet regimens (STRs) for the cure of HCV infection. VEL combined with sofosbuvir (SOF) is Epclusa®, an STR with 98% cure-rates for genotype 1-6 HCV infected patients. Addition of the pan-genotypic HCV NS3/4A protease inhibitor voxilaprevir to SOF/VEL is the STR Vosevi®, which affords 97% cure-rates for genotype 1-6 HCV patients who have previously failed another treatment regimen.


Subject(s)
Antiviral Agents/pharmacology , Carbamates/pharmacology , Drug Discovery , Hepacivirus/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Carbamates/chemical synthesis , Carbamates/chemistry , Dose-Response Relationship, Drug , Drug Combinations , Genotype , Hepacivirus/genetics , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Macrocyclic Compounds/chemistry , Microbial Sensitivity Tests , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Sofosbuvir/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Tablets/chemistry , Tablets/pharmacology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
3.
Bioorg Med Chem Lett ; 29(16): 2428-2436, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31133531

ABSTRACT

Treatment of hepatitis C virus (HCV) infection has been historically challenging due the high viral genetic complexity wherein there are eight distinct genotypes and at least 86 viral subtypes. While HCV NS3/4A protease inhibitors are an established treatment option for genotype 1 infection, limited coverage of genotypes 2 and/or 3 combined with serum alanine transaminase (ALT) elevations for some compounds has limited the broad utility of this therapeutic class. Our discovery efforts were focused on identifying an NS3/4A protease inhibitor with pan-genotypic antiviral activity, improved coverage of resistance associated substitutions, and a decreased risk of hepatotoxicity. Towards this goal, distinct interactions with the conserved catalytic triad of the NS3/4A protease were identified that improved genotype 3 antiviral activity. We further discovered that protein adduct formation strongly correlated with clinical ALT elevation for this therapeutic class. Improving metabolic stability and decreasing protein adduct formation through structural modifications ultimately resulted in voxilaprevir. Voxilaprevir, in combination with sofosbuvir and velpatasvir, has demonstrated pan-genotypic antiviral clinical activity. Furthermore, hepatotoxicity was not observed in Phase 3 clinical trials with voxilaprevir, consistent with our design strategy. Vosevi® (sofosbuvir, velpatasvir, and voxilaprevir) is now an approved pan-genotypic treatment option for the most difficult-to-cure individuals who have previously failed direct acting antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Carbamates/chemistry , Drug Discovery , Hepacivirus/drug effects , Heterocyclic Compounds, 4 or More Rings/chemistry , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Protease Inhibitors/pharmacology , Sofosbuvir/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Aminoisobutyric Acids , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cyclopropanes , Dose-Response Relationship, Drug , Drug Combinations , Hepacivirus/genetics , Humans , Lactams, Macrocyclic , Leucine/analogs & derivatives , Macrocyclic Compounds/chemical synthesis , Microbial Sensitivity Tests , Molecular Structure , Proline/analogs & derivatives , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Quinoxalines , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
4.
Antimicrob Agents Chemother ; 60(9): 5368-78, 2016 09.
Article in English | MEDLINE | ID: mdl-27353271

ABSTRACT

Velpatasvir (VEL, GS-5816) is a novel pan-genotypic hepatitis C virus (HCV) nonstructural protein 5A (NS5A) inhibitor with activity against genotype 1 (GT1) to GT6 HCV replicons. In a phase 1b 3-day monotherapy study, patients treated with a 150-mg dose of GS-5816 had a mean maximal HCV RNA decline of ≥3.3 log10 IU/ml in GT1a, -1b, -2, -3, and -4. This report characterizes virologic resistance to VEL in these patients. NS5A resistance-associated substitutions (RASs) were detected by deep sequencing (1% cutoff) pretreatment in 22/70 patients, i.e., 10/35 (29%) patients with GT1a, 1/8 (13%) with GT1b, 4/8 (50.0%) with GT2, 5/17 (29.4%) with GT3, and 2/2 (100.0%) with GT4. In GT1a and GT3 patients, pretreatment RASs were associated with a slightly reduced HCV RNA response compared to that of patients without pretreatment RASs; among patients with GT1b, GT2, and GT4, no significant difference in response was observed in those with or without pretreatment RASs. Following treatment, the pattern of emergent RASs was more complex for GT1a than for the other genotypes. In GT1a, substitutions emerged at positions M28, Q30, L31, P32, H58, E92, and Y93, with the most prevalent substitutions at positions Y93, M28, and L31. RASs were observed at two positions in GT1b and GT2 (Y93 and L31), three positions in GT3 (Y93, L31, and E92), and four positions in GT4 (L28, M31, P32L, and Y93). RASs that were present pretreatment persisted through the 48-week follow-up period; however, RASs emerging during treatment were more likely to decline both in prevalence and in frequency within the viral population during follow-up. (This study has been registered at ClinicalTrials.gov under registration no. NCT01740791.).


Subject(s)
Antiviral Agents/therapeutic use , Carbamates/therapeutic use , Drug Resistance, Viral/genetics , Hepatitis C, Chronic/drug therapy , Heterocyclic Compounds, 4 or More Rings/therapeutic use , RNA, Viral/genetics , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Double-Blind Method , Drug Administration Schedule , Gene Expression , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatitis C, Chronic/virology , Humans , Mutation , RNA, Viral/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors
5.
Antimicrob Agents Chemother ; 60(3): 1847-1853, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26824950

ABSTRACT

Ledipasvir (LDV; GS-5885), a component of Harvoni (a fixed-dose combination of LDV with sofosbuvir [SOF]), is approved to treat chronic hepatitis C virus (HCV) infection. Here, we report key preclinical antiviral properties of LDV, including in vitro potency, in vitro resistance profile, and activity in combination with other anti-HCV agents. LDV has picomolar antiviral activity against genotype 1a and genotype 1b replicons with 50% effective concentration (EC50) values of 0.031 nM and 0.004 nM, respectively. LDV is also active against HCV genotypes 4a, 4d, 5a, and 6a with EC50 values of 0.11 to 1.1 nM. LDV has relatively less in vitro antiviral activity against genotypes 2a, 2b, 3a, and 6e, with EC50 values of 16 to 530 nM. In vitro resistance selection with LDV identified the single Y93H and Q30E resistance-associated variants (RAVs) in the NS5A gene; these RAVs were also observed in patients after a 3-day monotherapy treatment. In vitro antiviral combination studies indicate that LDV has additive to moderately synergistic antiviral activity when combined with other classes of HCV direct-acting antiviral (DAA) agents, including NS3/4A protease inhibitors and the nucleotide NS5B polymerase inhibitor SOF. Furthermore, LDV is active against known NS3 protease and NS5B polymerase inhibitor RAVs with EC50 values equivalent to those for the wild type.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Fluorenes/pharmacology , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Viral Nonstructural Proteins/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Cell Line, Tumor , Drug Combinations , Drug Resistance, Viral/genetics , Drug Synergism , Genotype , HeLa Cells , Hepacivirus/genetics , Humans , Intracellular Signaling Peptides and Proteins , Microbial Sensitivity Tests , Sofosbuvir/pharmacology
6.
ACS Med Chem Lett ; 15(4): 546-554, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38628802

ABSTRACT

Chronic hepatitis B (CHB) virus infection afflicts hundreds of millions of people and causes nearly one million deaths annually. The high levels of circulating viral surface antigen (HBsAg) that characterize CHB may lead to T-cell exhaustion, resulting in an impaired antiviral immune response in the host. Agents that suppress HBsAg could help invigorate immunity toward infected hepatocytes and facilitate a functional cure. A series of dihydropyridoisoquinolizinone (DHQ) inhibitors of human poly(A) polymerases PAPD5/7 were reported to suppress HBsAg in vitro. An example from this class, RG7834, briefly entered the clinic. We set out to identify a potent, orally bioavailable, and safe PAPD5/7 inhibitor as a potential component of a functional cure regimen. Our efforts led to the identification of a dihydropyridophthalazinone (DPP) core with improved pharmacokinetic properties. A conformational restriction strategy and optimization of core substitution led to GS-8873, which was projected to provide deep HBsAg suppression with once-daily dosing.

7.
J Hepatol ; 57(1): 24-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22314425

ABSTRACT

BACKGROUND & AIMS: GS-5885 is an inhibitor of the hepatitis C virus (HCV) NS5A protein and exhibits potent suppression of genotype 1 HCV replicons. The safety, tolerability, pharmacokinetics, antiviral activity, and resistance profile of once-daily GS-5885 doses of 1-90 mg were evaluated in patients with chronic genotype 1 HCV. METHODS: Genotype 1 HCV-infected patients were randomized to 3 days of once-daily (QD) dosing with placebo (n=12) or GS-5885 1 mg (n=10), 3 mg (n=10), 10 mg (n=20), 30 mg (n=10), or 90 mg (n=10). Plasma samples for pharmacokinetics, HCV RNA, and NS5A sequencing were collected through day 14. RESULTS: GS-5885 was well tolerated and resulted in median maximal reductions in HCV RNA ranging from 2.3 log(10) IU/ml (1 mg QD) to 3.3 log(10) IU/ml (10 mg QD in genotype 1b and 30 mg QD). E(max) modeling indicated GS-5885 30 mg was associated with>95% of maximal antiviral response to HCV genotype 1a. HCV RNA reductions were generally more sustained among patients with genotype 1b vs. 1a. Three of 60 patients had a reduced response and harbored NS5A-resistant virus at baseline. NS5A sequencing identified residues 30 and 31 in genotype 1a, and 93 in genotype 1b as the predominant sites of mutation following GS-5885 dosing. Plasma pharmacokinetics was consistent with QD dosing. CONCLUSIONS: During 3 days of monotherapy, low doses of GS-5885 demonstrated significant antiviral activity in genotype 1a and 1b HCV-infected patients. GS-5885 is currently being evaluated in combination with direct antiviral regimens with and without peginterferon.


Subject(s)
Antiviral Agents/administration & dosage , Benzimidazoles/administration & dosage , Fluorenes/administration & dosage , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Viral Nonstructural Proteins/antagonists & inhibitors , Adolescent , Adult , Aged , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Dose-Response Relationship, Drug , Drug Resistance, Viral/genetics , Female , Fluorenes/adverse effects , Fluorenes/pharmacokinetics , Genotype , Hepacivirus/genetics , Humans , Male , Middle Aged , Placebos , RNA, Viral/metabolism , Treatment Outcome , Young Adult
8.
Nat Med ; 25(9): 1377-1384, 2019 09.
Article in English | MEDLINE | ID: mdl-31501601

ABSTRACT

People living with HIV (PLWH) have expressed concern about the life-long burden and stigma associated with taking pills daily and can experience medication fatigue that might lead to suboptimal treatment adherence and the emergence of drug-resistant viral variants, thereby limiting future treatment options1-3. As such, there is strong interest in long-acting antiretroviral (ARV) agents that can be administered less frequently4. Herein, we report GS-CA1, a new archetypal small-molecule HIV capsid inhibitor with exceptional potency against HIV-2 and all major HIV-1 types, including viral variants resistant to the ARVs currently in clinical use. Mechanism-of-action studies indicate that GS-CA1 binds directly to the HIV-1 capsid and interferes with capsid-mediated nuclear import of viral DNA, HIV particle production and ordered capsid assembly. GS-CA1 selects in vitro for unfit GS-CA1-resistant capsid variants that remain fully susceptible to other classes of ARVs. Its high metabolic stability and low solubility enabled sustained drug release in mice following a single subcutaneous dosing. GS-CA1 showed high antiviral efficacy as a long-acting injectable monotherapy in a humanized mouse model of HIV-1 infection, outperforming long-acting rilpivirine. Collectively, these results demonstrate the potential of ultrapotent capsid inhibitors as new long-acting agents for the treatment of HIV-1 infection.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV Infections/drug therapy , HIV-1/drug effects , Indazoles/pharmacology , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Anti-HIV Agents/therapeutic use , Capsid/drug effects , Capsid/metabolism , Capsid Proteins/genetics , DNA, Viral/drug effects , Delayed-Action Preparations , Drug Resistance, Viral/drug effects , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , HIV-2/drug effects , HIV-2/pathogenicity , Humans , Indazoles/therapeutic use , Medication Adherence , Mice , Pyridines/therapeutic use
9.
Curr Opin Drug Discov Devel ; 9(4): 471-82, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16889230

ABSTRACT

Cathepsin S is expressed in antigen-presenting cells and plays a role in invariant chain processing and major histocompatibility complex class II (MHCII) antigen presentation leading to CD4+ T-cell activation. An oral cathepsin S inhibitor that blocks MHCII antigen presentation could result in a T-cell-selective immunosuppressant agent with improved safety over the current standard of care for the treatment of rheumatoid arthritis, psoriasis, multiple sclerosis and other autoimmune-based inflammatory diseases. This review focuses on advances in cathepsin S inhibitor utility and design since January of 2004.


Subject(s)
Cathepsins/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Technology, Pharmaceutical/trends , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/enzymology , Cathepsins/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Technology, Pharmaceutical/methods
10.
PLoS One ; 10(4): e0122844, 2015.
Article in English | MEDLINE | ID: mdl-25856426

ABSTRACT

Ledipasvir, a direct acting antiviral agent (DAA) targeting the Hepatitis C Virus NS5A protein, exhibits picomolar activity in replicon cells. While its mechanism of action is unclear, mutations that confer resistance to ledipasvir in HCV replicon cells are located in NS5A, suggesting that NS5A is the direct target of ledipasvir. To date co-precipitation and cross-linking experiments in replicon or NS5A transfected cells have not conclusively shown a direct, specific interaction between NS5A and ledipasvir. Using recombinant, full length NS5A, we show that ledipasvir binds directly, with high affinity and specificity, to NS5A. Ledipasvir binding to recombinant NS5A is saturable with a dissociation constant in the low nanomolar range. A mutant form of NS5A (Y93H) that confers resistance to ledipasvir shows diminished binding to ledipasvir. The current study shows that ledipasvir inhibits NS5A through direct binding and that resistance to ledipasvir is the result of a reduction in binding affinity to NS5A mutants.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Drug Resistance, Viral/genetics , Fluorenes/pharmacology , Hepacivirus/drug effects , Mutation , Viral Nonstructural Proteins/genetics , Amino Acid Sequence , Antiviral Agents/chemistry , Benzimidazoles/chemistry , Cell Line, Tumor , Fluorenes/chemistry , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Humans , Kinetics , Molecular Sequence Data , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replicon , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Virus Replication
11.
J Med Chem ; 57(5): 2033-46, 2014 03 13.
Article in English | MEDLINE | ID: mdl-24320933

ABSTRACT

A new class of highly potent NS5A inhibitors with an unsymmetric benzimidazole-difluorofluorene-imidazole core and distal [2.2.1]azabicyclic ring system was discovered. Optimization of antiviral potency and pharmacokinetics led to the identification of 39 (ledipasvir, GS-5885). Compound 39 (GT1a replicon EC50 = 31 pM) has an extended plasma half-life of 37-45 h in healthy volunteers and produces a rapid >3 log viral load reduction in monotherapy at oral doses of 3 mg or greater with once-daily dosing in genotype 1a HCV-infected patients. 39 has been shown to be safe and efficacious, with SVR12 rates up to 100% when used in combination with direct-acting antivirals having complementary mechanisms.


Subject(s)
Antiviral Agents/therapeutic use , Benzimidazoles/therapeutic use , Fluorenes/therapeutic use , Hepatitis C/drug therapy , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Base Sequence , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , DNA Primers , Double-Blind Method , Fluorenes/pharmacokinetics , Fluorenes/pharmacology , Half-Life , Humans , Macaca fascicularis , Male , Placebos , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL