Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Rev ; 121(6): 3238-3270, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33410674

ABSTRACT

Drug resistance is prevalent across many diseases, rendering therapies ineffective with severe financial and health consequences. Rather than accepting resistance after the fact, proactive strategies need to be incorporated into the drug design and development process to minimize the impact of drug resistance. These strategies can be derived from our experience with viral disease targets where multiple generations of drugs had to be developed to combat resistance and avoid antiviral failure. Significant efforts including experimental and computational structural biology, medicinal chemistry, and machine learning have focused on understanding the mechanisms and structural basis of resistance against direct-acting antiviral (DAA) drugs. Integrated methods show promise for being predictive of resistance and potency. In this review, we give an overview of this research for human immunodeficiency virus type 1, hepatitis C virus, and influenza virus and the lessons learned from resistance mechanisms of DAAs. These lessons translate into rational strategies to avoid resistance in drug design, which can be generalized and applied beyond viral targets. While resistance may not be completely avoidable, rational drug design can and should incorporate strategies at the outset of drug development to decrease the prevalence of drug resistance.


Subject(s)
Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Pharmaceutical Preparations/chemistry , Viral Proteins/chemistry , Virus Diseases/drug therapy , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Computational Biology , Drug Design , Drug Resistance, Viral , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , HIV-1/drug effects , Hepacivirus/drug effects , Humans , Machine Learning , Mutation , Orthomyxoviridae/drug effects , Pharmaceutical Preparations/metabolism , Protein Binding , Signal Transduction , Structure-Activity Relationship
2.
Crit Rev Biochem Mol Biol ; 54(1): 11-26, 2019 02.
Article in English | MEDLINE | ID: mdl-30821513

ABSTRACT

Direct acting antivirals have dramatically increased the efficacy and tolerability of hepatitis C treatment, but drug resistance has emerged with some of these inhibitors, including nonstructural protein 3/4 A protease inhibitors (PIs). Although many co-crystal structures of PIs with the NS3/4A protease have been reported, a systematic review of these crystal structures in the context of the rapidly emerging drug resistance especially for early PIs has not been performed. To provide a framework for designing better inhibitors with higher barriers to resistance, we performed a quantitative structural analysis using co-crystal structures and models of HCV NS3/4A protease in complex with natural substrates and inhibitors. By comparing substrate structural motifs and active site interactions with inhibitor recognition, we observed that the selection of drug resistance mutations correlates with how inhibitors deviate from viral substrates in molecular recognition. Based on this observation, we conclude that guiding the design process with native substrate recognition features is likely to lead to more robust small molecule inhibitors with decreased susceptibility to resistance.


Subject(s)
Drug Resistance, Viral , Hepacivirus/drug effects , Hepatitis C/drug therapy , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Catalytic Domain/drug effects , Hepacivirus/metabolism , Hepatitis C/virology , Humans , Protease Inhibitors/chemistry , Protein Conformation/drug effects , Serine Proteases/chemistry , Serine Proteases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
3.
Nat Chem Biol ; 13(10): 1115-1122, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28825711

ABSTRACT

The HIV-1 envelope (Env) spike is a conformational machine that transitions between prefusion (closed, CD4- and CCR5-bound) and postfusion states to facilitate HIV-1 entry into cells. Although the prefusion closed conformation is a potential target for inhibition, development of small-molecule leads has been stymied by difficulties in obtaining structural information. Here, we report crystal structures at 3.8-Å resolution of an HIV-1-Env trimer with BMS-378806 and a derivative BMS-626529 for which a prodrug version is currently in Phase III clinical trials. Both lead candidates recognized an induced binding pocket that was mostly excluded from solvent and comprised of Env elements from a conserved helix and the ß20-21 hairpin. In both structures, the ß20-21 region assumed a conformation distinct from prefusion-closed and CD4-bound states. Together with biophysical and antigenicity characterizations, the structures illuminate the allosteric and competitive mechanisms by which these small-molecule leads inhibit CD4-induced structural changes in Env.


Subject(s)
HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp41/chemistry , Piperazines/chemistry , Small Molecule Libraries/chemistry , Triazoles/chemistry , Virus Internalization/drug effects , Crystallography, X-Ray , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp41/antagonists & inhibitors , Models, Molecular , Piperazines/pharmacology , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Triazoles/pharmacology
4.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28298600

ABSTRACT

The mosquito-transmitted dengue virus (DENV) infects millions of people in tropical and subtropical regions. Maturation of DENV particles requires proper cleavage of the viral polyprotein, including processing of 8 of the 13 substrate cleavage sites by dengue virus NS2B/NS3 protease. With no available direct-acting antiviral targeting DENV, NS2/NS3 protease is a promising target for inhibitor design. Current design efforts focus on the nonprime side of the DENV protease active site, resulting in highly hydrophilic and nonspecific scaffolds. However, the prime side also significantly modulates DENV protease binding affinity, as revealed by engineering the binding loop of aprotinin, a small protein with high affinity for DENV protease. In this study, we designed a series of cyclic peptides interacting with both sides of the active site as inhibitors of dengue virus protease. The design was based on two aprotinin loops and aimed to leverage both key specific interactions of substrate sequences and the entropic advantage driving aprotinin's high affinity. By optimizing the cyclization linker, length, and amino acid sequence, the tightest cyclic peptide achieved a Ki value of 2.9 µM against DENV3 wild-type (WT) protease. These inhibitors provide proof of concept that both sides of DENV protease active site can be exploited to potentially achieve specificity and lower hydrophilicity in the design of inhibitors targeting DENV.IMPORTANCE Viruses of the flaviviral family, including DENV and Zika virus transmitted by Aedes aegypti, continue to be a threat to global health by causing major outbreaks in tropical and subtropical regions, with no available direct-acting antivirals for treatment. A better understanding of the molecular requirements for the design of potent and specific inhibitors against flaviviral proteins will contribute to the development of targeted therapies for infections by these viruses. The cyclic peptides reported here as DENV protease inhibitors provide novel scaffolds that enable exploiting the prime side of the protease active site, with the aim of achieving better specificity and lower hydrophilicity than those of current scaffolds in the design of antiflaviviral inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Peptides, Cyclic/pharmacology , Protease Inhibitors/pharmacology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acid Sequence , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Aprotinin/chemistry , Aprotinin/metabolism , Aprotinin/pharmacology , Catalytic Domain , Computer Simulation , Dengue Virus/chemistry , Dengue Virus/enzymology , Drug Discovery/methods , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Peptides, Cyclic/chemical synthesis , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Viral Nonstructural Proteins/chemistry
5.
J Am Chem Soc ; 138(36): 11850-9, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27512818

ABSTRACT

Hepatitis C virus (HCV), affecting an estimated 150 million people worldwide, is the leading cause of viral hepatitis, cirrhosis and hepatocellular carcinoma. HCV is genetically diverse with six genotypes (GTs) and multiple subtypes of different global distribution and prevalence. Recent development of direct-acting antivirals against HCV including NS3/4A protease inhibitors (PIs) has greatly improved treatment outcomes for GT-1. However, all current PIs exhibit significantly lower potency against GT-3. Lack of structural data on GT-3 protease has limited our ability to understand PI failure in GT-3. In this study the molecular basis for reduced potency of current inhibitors against GT-3 NS3/4A protease is elucidated with structure determination, molecular dynamics simulations and inhibition assays. A chimeric GT-1a3a NS3/4A protease amenable to crystallization was engineered to recapitulate decreased sensitivity of GT-3 protease to PIs. High-resolution crystal structures of this GT-1a3a bound to 3 PIs, asunaprevir, danoprevir and vaniprevir, had only subtle differences relative to GT-1 despite orders of magnitude loss in affinity. In contrast, hydrogen-bonding interactions within and with the protease active site and dynamic fluctuations of the PIs were drastically altered. The correlation between loss of intermolecular dynamics and inhibitor potency suggests a mechanism where polymorphisms between genotypes (or selected mutations) in the drug target confer resistance through altering the intermolecular dynamics of the protein-inhibitor complex.


Subject(s)
Drug Resistance, Viral/genetics , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Viral Nonstructural Proteins/genetics , Catalytic Domain , Hepacivirus/enzymology , Molecular Dynamics Simulation , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Static Electricity , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
6.
ACS Chem Biol ; 11(4): 900-9, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26682473

ABSTRACT

Recent advances in direct-acting antivirals against Hepatitis C Virus (HCV) have led to the development of potent inhibitors, including MK-5172, that target the viral NS3/4A protease with relatively low susceptibility to resistance. MK-5172 has a P2-P4 macrocycle and a unique binding mode among current protease inhibitors where the P2 quinoxaline packs against the catalytic residues H57 and D81. However, the effect of macrocyclization on this binding mode is not clear, as is the relation between macrocyclization, thermodynamic stabilization, and susceptibility to the resistance mutation A156T. We have determined high-resolution crystal structures of linear and P1-P3 macrocyclic analogs of MK-5172 bound to WT and A156T protease and compared these structures, their molecular dynamics, and experimental binding thermodynamics to the parent compound. We find that the "unique" binding mode of MK-5172 is conserved even when the P2-P4 macrocycle is removed or replaced with a P1-P3 macrocycle. While beneficial to decreasing the entropic penalty associated with binding, the constraint exerted by the P2-P4 macrocycle prevents efficient rearrangement to accommodate the A156T mutation, a deficit alleviated in the linear and P1-P3 analogs. Design of macrocyclic inhibitors against NS3/4A needs to achieve the best balance between exerting optimal conformational constraint for enhancing potency, fitting within the substrate envelope and allowing adaptability to be robust against resistance mutations.


Subject(s)
Antiviral Agents/pharmacology , Quinoxalines/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Amides , Antiviral Agents/chemistry , Carbamates , Cyclopropanes , Quinoxalines/chemistry , Sulfonamides , Thermodynamics
7.
ACS Chem Biol ; 9(11): 2485-90, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25243902

ABSTRACT

Asunaprevir (ASV), an isoquinoline-based competitive inhibitor targeting the hepatitis C virus (HCV) NS3/4A protease, is very potent in vivo. However, the potency is significantly compromised by the drug resistance mutations R155K and D168A. In this study three crystal structures of ASV and an analogue were determined to analyze the structural basis of drug resistance susceptibility. These structures revealed that ASV makes extensive contacts with Arg155 outside the substrate envelope. Arg155 in turn is stabilized by Asp168, and thus when either residue is mutated, the enzyme's interaction with ASV's P2* isoquinoline is disrupted. Adding a P1-P3 macrocycle to ASV enhances the inhibitor's resistance barrier, likely due to poising the inhibitor to its bound conformation. Macrocyclic inhibitors with P2* extension moieties avoiding interaction with the protease S2 residues including Arg155 must be chosen for future design of more robust protease inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Carrier Proteins/chemistry , Isoquinolines/pharmacology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/chemistry , Drug Resistance, Viral/genetics , Hepacivirus/drug effects , Intracellular Signaling Peptides and Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...