1
|
Du S, Hu X, Menéndez-Arias L, Zhan P, Liu X. Target-based drug design strategies to overcome resistance to antiviral agents: opportunities and challenges. Drug Resist Updat 2024; 73:101053. [PMID: 38301487 DOI: 10.1016/j.drup.2024.101053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
Viral infections have a major impact in human health. Ongoing viral transmission and escalating selective pressure have the potential to favor the emergence of vaccine- and antiviral drug-resistant viruses. Target-based approaches for the design of antiviral drugs can play a pivotal role in combating drug-resistant challenges. Drug design computational tools facilitate the discovery of novel drugs. This review provides a comprehensive overview of current drug design strategies employed in the field of antiviral drug resistance, illustrated through the description of a series of successful applications. These strategies include technologies that enhance compound-target affinity while minimizing interactions with mutated binding pockets. Furthermore, emerging approaches such as virtual screening, targeted protein/RNA degradation, and resistance analysis during drug design have been harnessed to curtail the emergence of drug resistance. Additionally, host targeting antiviral drugs offer a promising avenue for circumventing viral mutation. The widespread adoption of these refined drug design strategies will effectively address the prevailing challenge posed by antiviral drug resistance.
Collapse
Affiliation(s)
- Shaoqing Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Xueping Hu
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237, PR China
| | - Luis Menéndez-Arias
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid), Madrid, Spain.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China; China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, 250012 Jinan, Shandong, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China; China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, 250012 Jinan, Shandong, PR China.
| |
Collapse
|
2
|
Lewis ME, Simpson P, Mori J, Jubb B, Sullivan J, McFadyen L, van der Ryst E, Craig C, Robertson DL, Westby M. V3-Loop genotypes do not predict maraviroc susceptibility of CCR5-tropic virus or clinical response through week 48 in HIV-1-infected, treatment-experienced persons receiving optimized background regimens. Antivir Chem Chemother 2021; 29:20402066211030380. [PMID: 34343443 PMCID: PMC8369958 DOI: 10.1177/20402066211030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Viruses from 15 of 35 maraviroc-treated participants with virologic failure and CCR5-tropic (R5) virus in the MOTIVATE studies at Week 24 had reduced maraviroc susceptibility. On-treatment amino acid changes were observed in the viral envelope glycoprotein 120 third variable (V3)-loop stems and tips and differed between viruses. No amino acid change reliably predicted reduced susceptibility, indicating that resistance was genetic context-dependent. Through Week 24, poor adherence was associated with maraviroc-susceptible virologic failure, whereas reduced maraviroc susceptibility was associated with suboptimal background regimen activity, highlighting the importance of overall regimen activity and good adherence. Predictive values of pretreatment V3-loop sequences containing these Week 24 mutations or other variants present at >3% in pretreatment viruses of participants with virologic failure at Week 48 were retrospectively assessed. Week 48 clinical outcomes were evaluated for correlates with pretreatment V3-loop CCR5-tropic sequences from 704 participants (366 responders; 338 virologic failures [83 with R5 virus with maraviroc susceptibility assessment]). Seventy-five amino acid variants with >3% prevalence were identified among 23 V3-loop residues. Previously identified variants associated with resistance in individual isolates were represented, but none were associated reliably with virologic failure alone or in combination. Univariate analysis showed virologic-failure associations with variants 4L, 11R, and 19S (P < 0.05). However, 11R is a marker for CXCR4 tropism, whereas neither 4L nor 19S was reliably associated with reduced maraviroc susceptibility in R5 failure. These findings from a large study of V3-loop sequences confirm lack of correlation between V3-loop genotype and clinical outcome in participants treated with maraviroc.Clinical trial registration numbers (ClinicalTrials.gov): NCT00098306 and NCT00098722.
Collapse
Affiliation(s)
- M E Lewis
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,The Research Network Ltd, Sandwich, Kent, UK
| | - P Simpson
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,AstraZeneca, Cambridge, UK
| | - J Mori
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,hVIVO, Queen Mary BioEnterprise Innovation Centre, London, UK
| | - B Jubb
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK
| | - J Sullivan
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,Cytel, London, UK
| | - L McFadyen
- Pfizer Inc, Pharmacometrics, Sandwich, UK
| | - E van der Ryst
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,The Research Network Ltd, Sandwich, Kent, UK
| | - C Craig
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,The Research Network Ltd, Sandwich, Kent, UK
| | - D L Robertson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - M Westby
- Pfizer Global Research and Development, Sandwich Labs, Sandwich, Kent, UK.,Centauri Therapeutics Limited, Discovery Park, Kent, UK
| |
Collapse
|
3
|
Jubb B, Lewis M, McFadyen L, Simpson P, Mori J, Chan P, Weatherley B, van der Ryst E, Westby M, Craig C. Incidence of CXCR4 tropism and CCR5-tropic resistance in treatment-experienced participants receiving maraviroc in the 48-week MOTIVATE 1 and 2 trials. Antivir Chem Chemother 2019; 27:2040206619895706. [PMID: 31856576 PMCID: PMC6931239 DOI: 10.1177/2040206619895706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Maraviroc blocks HIV-1 entry into CD4+ cells by interrupting the interaction
between viral gp120 and cell-surface CCR5. Resistance to CCR5
antagonist–mediated inhibition can develop by unmasking pre-existing CXCR4-using
virus or through selection of CCR5-tropic resistant virus, characterized by
plateaus in maximum percent inhibition <95%. Here, we examine viral escape in
maraviroc-treated participants during virologic failure through Week 48 in the
MOTIVATE 1 and 2 trials. Resistance was assessed relative to number of active
drugs in participants’ optimized background therapy, pharmacokinetic adherence
markers, Baseline demographic data, HIV-1 RNA and CD4+ counts. For participants
with R5 virus confirmed (post hoc) at Screening, Baseline
genotypic weighted optimized background therapy susceptibility scores (gwOBTSS)
were assigned where possible. Through Week 48, 219/392 (56%) participants with
an assigned gwOBTSS achieved a virologic response. Of those remaining, 48/392
(12%) had CXCR4-using virus; 58/392 (15%) had R5 virus (maraviroc sensitive:
n = 35/392, 9%; maraviroc resistant:
n = 18/392, 5%; undeterminable: n = 5/392, 1%)
and 67/392 (17%) had no failure tropism result. When optimized background
therapy provided limited support to maraviroc (gwOBTSS <2), 143/286 (50%)
responded to therapy, while 76/106 (72%) participants with gwOBTSS ≥2 responded
(p < 0.001). Resistance rates were highest for
participants with gwOBTSS <2, accounting for 45/48 (94%) of total CXCR4-using
emergence and 18/18 (100%) of total CCR5-tropic resistance. R5 viruses from
participants with gwOBTSS ≥2 (n = 10) were exclusively
maraviroc sensitive; five of these participants had pharmacokinetic and/or
pill-count markers of non-adherence. When co-administered with a fully active
background regimen, maraviroc did not readily generate resistance in the
clinical setting.
Collapse
Affiliation(s)
- Becky Jubb
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA
| | - Marilyn Lewis
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA.,The Research Network, Sandwich, UK
| | | | - Paul Simpson
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA
| | - Julie Mori
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA
| | | | | | - Elna van der Ryst
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA.,The Research Network, Sandwich, UK
| | - Mike Westby
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA
| | - Charles Craig
- Pfizer Inc, Clinical Group, Rare Disease, Groton, CT, USA.,The Research Network, Sandwich, UK
| |
Collapse
|
4
|
Cheung PK, Horhant D, Bandy LE, Zamiri M, Rabea SM, Karagiosov SK, Matloobi M, McArthur S, Harrigan PR, Chabot B, Grierson DS. A Parallel Synthesis Approach to the Identification of Novel Diheteroarylamide-Based Compounds Blocking HIV Replication: Potential Inhibitors of HIV-1 Pre-mRNA Alternative Splicing. J Med Chem 2016; 59:1869-79. [PMID: 26878150 DOI: 10.1021/acs.jmedchem.5b01357] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A 256-compound library was evaluated in an anti-HIV screen to identify structural "mimics" of the fused tetracyclic indole compound 1 (IDC16) that conserve its anti-HIV activity without associated cytotoxicity. Four diheteroarylamide-type compounds, containing a common 5-nitroisobenzothiazole motif, were identified as active. In subsequent screens, the most potent compound 9 (1C8) was active against wild-type HIV-1IIIB (subtype B, X4-tropic) and HIV-1 97USSN54 (subtype A, R5-tropic) with EC50's of 0.6 and 0.9 μM, respectively. Compound 9 also inhibited HIV strains resistant to drugs targeting HIV reverse transcriptase, protease, integrase, and coreceptor CCR5 with EC50's ranging from 0.9 to 1.5 μM. The CC50 value obtained in a cytotoxicity assay for compound 9 was >100 μM, corresponding to a therapeutic index (CC50/EC50) of approximately 100. Further comparison studies revealed that, whereas the anti-HIV activity for compound 9 and the parent molecule 1 are similar, the cytotoxic effect for compound 9 was, as planned, markedly suppressed.
Collapse
Affiliation(s)
- Peter K Cheung
- British Columbia Centre for Excellence in HIV/AIDS , 608-1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | | | | | | | | | | | | | | | - P Richard Harrigan
- British Columbia Centre for Excellence in HIV/AIDS , 608-1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Benoit Chabot
- Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke , 3201, rue Jean-Mignault, Sherbrooke, Québec J1E 4K8 Canada
| | | |
Collapse
|
5
|
Abstract
The human immunodeficiency virus-1 (HIV-1) enters target cells by binding its envelope glycoprotein gp120 to the CD4 receptor and/or coreceptors such as C-C chemokine receptor type 5 (CCR5; R5) and C-X-C chemokine receptor type 4 (CXCR4; X4), and R5-tropic viruses predominate during the early stages of infection. CCR5 antagonists bind to CCR5 to prevent viral entry. Maraviroc (MVC) is the only CCR5 antagonist currently approved by the United States Food and Drug Administration, the European Commission, Health Canada, and several other countries for the treatment of patients infected with R5-tropic HIV-1. MVC has been shown to be effective at inhibiting HIV-1 entry into cells and is well tolerated. With expanding MVC use by HIV-1-infected humans, different clinical outcomes post-approval have been observed with MVC monotherapy or combination therapy with other antiretroviral drugs, with MVC use in humans infected with dual-R5- and X4-tropic HIV-1, infected with different HIV-1 genotype or infected with HIV-2. This review discuss the role of CCR5 in HIV-1 infection, the development of the CCR5 antagonist MVC, its pharmacokinetics, pharmacodynamics, drug-drug interactions, and the implications of these interactions on treatment outcomes, including viral mutations and drug resistance, and the mechanisms associated with the development of resistance to MVC. This review also discusses available studies investigating the use of MVC in the treatment of other diseases such as cancer, graft-versus-host disease, and inflammatory diseases.
Collapse
Affiliation(s)
- Shawna M Woollard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Georgette D Kanmogne
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
6
|
Veljkovic N, Vucicevic J, Tassini S, Glisic S, Veljkovic V, Radi M. Preclinical discovery and development of maraviroc for the treatment of HIV. Expert Opin Drug Discov 2015; 10:671-84. [PMID: 25927601 DOI: 10.1517/17460441.2015.1041497] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Maraviroc is a first-in-class antiretroviral (ARV) drug acting on a host cell target (CCR5), which blocks the entry of the HIV virus into the cell. Maraviroc is currently indicated for combination ARV treatment in adults infected only with CCR5-tropic HIV-1. AREAS COVERED This drug discovery case history focuses on the key studies that led to the discovery and approval of maraviroc, as well as on post-launch clinical reports. The article is based on the data reported in published preclinical and clinical studies, conference posters and on drug package data. EXPERT OPINION The profound understanding of HIV's entry mechanisms has provided a strong biological rationale for targeting the chemokine receptor CCR5. The CCR5-antagonist mariviroc, with its unique mode of action and excellent safety profile, is an important therapeutic option for HIV patients. In general, the authors believe that targeting host factors is a useful approach for combating new and re-emerging transmissible diseases, as well as pathogens that easily become resistant to common antiviral drugs. Maraviroc, offering a potent and safe cellular receptor-mediated pharmacological response to HIV, has paved the way for the development of a new generation of host-targeting antivirals.
Collapse
Affiliation(s)
- Nevena Veljkovic
- University of Belgrade, Institute of Nuclear Sciences VINCA, Center for Multidisciplinary Research , P.O. Box 522, Belgrade , Serbia +381 11 3408154 ; + 381 11 7440100 ;
| | | | | | | | | | | |
Collapse
|
7
|
Raymond S, Maillard A, Amiel C, Peytavin G, Trabaud MA, Desbois D, Bellecave P, Delaugerre C, Soulie C, Marcelin AG, Descamps D, Izopet J, the ANRS ACll Resistance Study Group, Reigadas S, Bellecave P, Pinson-Recordon P, Fleury H, Masquelier B, Signori-Schmuck A, Morand P, Bocket L, Mouna L, Andre P, Tardy JC, Trabaud MA, Descamps D, Charpentier C, Peytavin G, Brun-Vezinet F, Haim-Boukobza S, Roques AM, Soulie C, Lambert-Niclot S, Malet I, Wirden M, Fourati S, Marcelin AG, Calvez V, Flandre P, Assoumou L, Costagliola D, Morand-Joubert L, Delaugerre C, Schneider V, Amiel C, Giraudeau G, Maillard A, Nicot F, Izopet J. Virological failure of patients on maraviroc-based antiretroviral therapy. J Antimicrob Chemother 2015; 70:1858-64. [DOI: 10.1093/jac/dkv026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/25/2015] [Indexed: 11/14/2022] Open
|
8
|
Emory JF, Seserko LA, Marzinke MA. Development and bioanalytical validation of a liquid chromatographic-tandem mass spectrometric (LC-MS/MS) method for the quantification of the CCR5 antagonist maraviroc in human plasma. Clin Chim Acta 2014; 431:198-205. [PMID: 24561264 DOI: 10.1016/j.cca.2014.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/28/2014] [Accepted: 02/12/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND Maraviroc is a CCR5 antagonist that has been utilized as a viral entry inhibitor in the management of HIV-1. Current clinical trials are pursuing maraviroc drug efficacy in both oral and topical formulations. Therefore, in order to fully understand drug pharmacokinetics, a sensitive method is required to quantify plasma drug concentrations. METHODS Maraviroc-spiked plasma was combined with acetonitrile containing an isotopically-labeled internal standard, and following protein precipitation, samples were evaporated to dryness and reconstituted for liquid chromatographic-tandem mass spectrometric (LC-MS/MS) analysis. Chromatographic separation was achieved on a Waters BEH C8, 50×2.1 mm UPLC column, with a 1.7 μm particle size and the eluent was analyzed using an API 4000 mass analyzer in selected reaction monitoring mode. The method was validated as per FDA Bioanalytical Method Validation guidelines. RESULTS The analytical measuring range of the LC-MS/MS method is 0.5-1000 ng/ml. Calibration curves were generated using weighted 1/x(2) quadratic regression. Inter-and intra-assay precision was ≤5.38% and ≤5.98%, respectively; inter-and intra-assay accuracy (%DEV) was ≤10.2% and ≤8.44%, respectively. Additional studies illustrated similar matrix effects between maraviroc and its internal standard, and that maraviroc is stable under a variety of conditions. Method comparison studies with a reference LC-MS/MS method show a slope of 0.948 with a Spearman coefficient of 0.98. CONCLUSIONS Based on the validation metrics, we have generated a sensitive and automated LC-MS/MS method for maraviroc quantification in human plasma.
Collapse
Affiliation(s)
- Joshua F Emory
- Department of Medicine, Johns Hopkins University, 600N. Wolfe St., Osler 500, Baltimore, MD 21287, United States
| | - Lauren A Seserko
- Department of Medicine, Johns Hopkins University, 600N. Wolfe St., Osler 500, Baltimore, MD 21287, United States
| | - Mark A Marzinke
- Department of Medicine, Johns Hopkins University, 600N. Wolfe St., Osler 500, Baltimore, MD 21287, United States; Department of Pathology, Johns Hopkins University, 600N. Wolfe St., Osler 500, Baltimore, MD 21287, United States.
| |
Collapse
|
9
|
Genotypic analysis of the V3 region of HIV from virologic nonresponders to maraviroc-containing regimens reveals distinct patterns of failure. Antimicrob Agents Chemother 2013; 57:6122-30. [PMID: 24080655 DOI: 10.1128/aac.01534-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Changes in HIV tropism from R5 to non-R5 or development of drug resistance is often associated with virologic failure in patients treated with maraviroc, a CCR5 antagonist. We sought to examine changes in HIV envelope sequences and inferred tropism in patients who did not respond to maraviroc-based regimens. We selected 181 patients who experienced early virologic failure on maraviroc-containing therapy in the MOTIVATE trials. All patients had R5 HIV by the original Trofile assay before entry. We used population-based sequencing methods and the geno2pheno algorithm to examine changes in tropism and V3 sequences at the time of failure. Using deep sequencing, we assessed whether V3 sequences observed at failure emerged from preexisting subpopulations. From population genotyping data at failure, 90 patients had R5 results, and 91 had non-R5 results. Of the latter group, the geno2pheno false-positive rate (FPR) value fell from a median of 20 at screening to 1.1 at failure. By deep sequencing, the median percentage of non-R5 variants in these patients rose from 1.4% to 99.5% after a median of 4 weeks on maraviroc. In 70% of cases, deep sequencing could detect a pretreatment CXCR4-using subpopulation, which emerged at failure. Overall, there were two distinct patterns of failure of maraviroc. Patients failing with R5 generally had few V3 substitutions and low non-R5 prevalence by deep sequencing. Patients with non-R5 HIV who were failing developed very-high-prevalence non-R5 HIV (median, 99%) and had very low geno2pheno values.
Collapse
|