Antiviral researchPub Date : 2024-07-06DOI: 10.1016/j.antiviral.2024.105958
{"title":"Prevention of post COVID-19 condition by early treatment with ensitrelvir in the phase 3 SCORPIO-SR trial","authors":"","doi":"10.1016/j.antiviral.2024.105958","DOIUrl":"10.1016/j.antiviral.2024.105958","url":null,"abstract":"<div><p>This exploratory analysis of the double-blind, phase 3, SCORPIO-SR trial assessed the effect of ensitrelvir in preventing post coronavirus disease 2019 (COVID-19) condition (PCC). Patients with mild-to-moderate COVID-19 were randomized (1:1:1) within 120 h of symptom onset; received 5-day oral ensitrelvir 125 mg (375 mg on day 1), 250 mg (750 mg on day 1), or a matching placebo once daily; and were assessed for the severity of typical PCC symptoms using a self-administered questionnaire. In total, 341, 317, and 333 patients were assessed in the ensitrelvir 125-mg, ensitrelvir 250-mg, and placebo groups, respectively (mean age, 35.6–36.5 years; men, 53.3%–58.3%). On days 85, 169, and 337, ensitrelvir 125-mg treatment showed 32.7% (95% confidence interval [CI]: 30.6, 66.1), 21.5% (95% CI: 37.3, 55.6), and 24.6% (95% CI: 43.7, 60.9) reductions versus placebo, respectively, in the risk of any of the 14 acute-phase COVID-19 symptoms (at least one mild, moderate, or severe symptom with general health not returning to the usual level). Ensitrelvir 250-mg treatment showed 10.9% (95% CI: 67.0, 52.8), 9.5% (95% CI: 56.6, 48.0), and 30.6% (95% CI: 36.2, 65.5) risk reductions versus placebo on days 85, 169, and 337, respectively. Risk reductions were observed in any of the 4 neurological symptoms and were more pronounced among patients with high acute-phase symptom scores at baseline and among those with a baseline body mass index ≥25 kg/m<sup>2</sup>. Ensitrelvir treatment in the acute phase of COVID-19 may reduce the risk of various symptoms associated with PCC.</p></div><div><h3>Trial registration number</h3><p>jRCT2031210350.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"229 ","pages":"Article 105958"},"PeriodicalIF":4.5,"publicationDate":"2024-07-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0166354224001670/pdfft?md5=7ff1c5fe8ac0d0a98196f04c4e255997&pid=1-s2.0-S0166354224001670-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141554100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-07-05DOI: 10.1016/j.antiviral.2024.105948
Brecht Bonneux , Edgar Jacoby , Martina Ceconi , Kim Stobbelaar , Peter Delputte , Florence Herschke
{"title":"Direct-acting antivirals for RSV treatment, a review","authors":"Brecht Bonneux , Edgar Jacoby , Martina Ceconi , Kim Stobbelaar , Peter Delputte , Florence Herschke","doi":"10.1016/j.antiviral.2024.105948","DOIUrl":"10.1016/j.antiviral.2024.105948","url":null,"abstract":"<div><p>Respiratory syncytial virus (RSV) causes respiratory disease and complications in infants, the elderly and the immunocompromised. While three vaccines and two prophylactic monoclonal antibodies are now available, only one antiviral, ribavirin, is currently approved for treatment. This review aims to summarize the current state of treatments directly targeting RSV. Two major viral processes are attractive for RSV-specific antiviral drug discovery and development as they play essential roles in the viral cycle: the entry/fusion process carried out by the fusion protein and the replication/transcription process carried out by the polymerase complex constituted of the L, P, N and M2-1 proteins. For each viral target resistance mutations to small molecules of different chemotypes seem to delineate definite binding pockets in the fusion proteins and in the large proteins. Elucidating the mechanism of action of these inhibitors thus helps to understand how the fusion and polymerase complexes execute their functions.</p><p>While many inhibitors have been studied, few are currently in clinical development for RSV treatment: one is in phase III, three in phase II and two in phase I. Progression was halted for many others because of strategic decisions, low enrollment, safety, but also lack of efficacy. Lessons can be learnt from the halted programs to increase the success rate of the treatments currently in development.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"229 ","pages":"Article 105948"},"PeriodicalIF":4.5,"publicationDate":"2024-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141554099","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-07-04DOI: 10.1016/j.antiviral.2024.105957
Margaret Gartland , Eugene Stewart , Nannan Zhou , Zhufang Li , Ronald Rose , Jagadish Beloor , Andrew Clark , Allan R. Tenorio , Mark Krystal
{"title":"Characterization of clinical envelopes with lack of sensitivity to the HIV-1 inhibitors temsavir and ibalizumab","authors":"Margaret Gartland , Eugene Stewart , Nannan Zhou , Zhufang Li , Ronald Rose , Jagadish Beloor , Andrew Clark , Allan R. Tenorio , Mark Krystal","doi":"10.1016/j.antiviral.2024.105957","DOIUrl":"10.1016/j.antiviral.2024.105957","url":null,"abstract":"<div><p>Previous data suggest a lack of cross-resistance between the gp120-directed attachment inhibitor temsavir (active moiety of fostemsavir) and the CD4-directed post-attachment inhibitor ibalizumab. Recently, analysis of HIV-1 envelopes with reduced sensitivity to both inhibitors was undertaken to determine whether they shared genotypic correlates of resistance. Sequences from 2 envelopes with reduced susceptibility to both agents were mapped onto a temsavir-bound gp120 structure. Residues within 5.0 Å of the temsavir binding site were evaluated using reverse genetics. Broader applicability and contextual determinants of key substitutions were further assessed using envelopes from participants in the phase 3 BRIGHTE study. Temsavir sensitivity was measured by half-maximal inhibitory concentration (IC<sub>50</sub>) and ibalizumab sensitivity by IC<sub>50</sub> and maximum percent inhibition (MPI). One envelope required substitutions of E113D and T434M for full restoration of temsavir susceptibility. Neither substitution nor their combination affected ibalizumab sensitivity. However, in the second envelope, an E202 substitution (HXB2, T202) was sufficient for observed loss of susceptibility to both inhibitors. One BRIGHTE participant with no ibalizumab exposure had an emergent K202E substitution at protocol-defined virologic failure, with reduced sensitivity to both inhibitors. Introducing T202E into previously susceptible clinical isolates reduced temsavir potency by ≥ 40-fold and ibalizumab MPI from >99% to ∼80%. Interestingly, introduction of the gp120 V5 region from a highly ibalizumab-susceptible envelope mitigated the E202 effect on ibalizumab but not temsavir. A rare HIV-1 gp120 E202 mutation reduced temsavir susceptibility, and depending on sequence context, could result in reduced susceptibility to ibalizumab.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"228 ","pages":"Article 105957"},"PeriodicalIF":4.5,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0166354224001669/pdfft?md5=17ee0f31fad80cee3289685131c8ea9d&pid=1-s2.0-S0166354224001669-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141544448","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-07-04DOI: 10.1016/j.antiviral.2024.105956
{"title":"Detection of influenza A(H3N2) viruses with polymerase acidic subunit substitutions after and prior to baloxavir marboxil treatment during the 2022–2023 influenza season in Japan","authors":"","doi":"10.1016/j.antiviral.2024.105956","DOIUrl":"10.1016/j.antiviral.2024.105956","url":null,"abstract":"<div><p>Baloxavir marboxil (baloxavir), approved as an anti-influenza drug in Japan in March 2018, can induce reduced therapeutic effectiveness due to PA protein substitutions. We assessed PA substitutions in clinical samples from influenza-infected children and adults pre- and post-baloxavir treatment, examining their impact on fever and symptom duration. During the 2022–2023 influenza season, the predominant circulating influenza subtype detected by cycling-probe RT-PCR was A(H3N2) (n = 234), with a minor circulation of A(H1N1)pdm09 (n = 10). Of the 234 influenza A(H3N2) viruses collected prior to baloxavir treatment, 2 (0.8%) viruses carry PA/I38T substitution. One virus was collected from a toddler and one from an adult, indicating the presence of viruses with reduced susceptibility to baloxavir, without prior exposure to the drug. Of the 54 paired influenza A(H3N2) viruses collected following baloxavir treatment, 8 (14.8%) viruses carried E23 K/G, or I38 M/T substitutions in PA. Variant calling through next-generation sequencing (NGS) showed varying proportions (6–100 %), a polymorphism and a mixture of PA/E23 K/G, and I38 M/T substitutions in the clinical samples. These eight viruses were obtained from children aged 7–14 years, with a median fever duration of 16.7 h and a median symptom duration of 93.7 h, which were similar to those of the wild type. However, the delayed viral clearance associated with the emergence of PA substitutions was observed. No substitutions conferring resistance to neuraminidase inhibitors were detected in 37 paired samples collected before and following oseltamivir treatment. These findings underscore the need for ongoing antiviral surveillance, informing public health strategies and clinical antiviral recommendations for seasonal influenza.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"229 ","pages":"Article 105956"},"PeriodicalIF":4.5,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0166354224001657/pdfft?md5=30544dd33875cb69979324d5c964b41c&pid=1-s2.0-S0166354224001657-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141537403","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-07-02DOI: 10.1016/j.antiviral.2024.105954
{"title":"Boosting the immune response in COVID-19 vaccines via an Alum:CpG complex adjuvant","authors":"","doi":"10.1016/j.antiviral.2024.105954","DOIUrl":"10.1016/j.antiviral.2024.105954","url":null,"abstract":"<div><p>Selecting appropriate adjuvants is crucial for developing an effective vaccine. However, studies on the immune responses triggered by different adjuvants in COVID-19 inactivated vaccines are scarce. Herein, we evaluated the efficacy of Alum, CpG HP021, Alum combined with CpG HP021 (Alum/CpG), or MF-59 adjuvants with COVID-19 inactivated vaccines in K18-hACE2 mice, and compared the different immune responses between K18-hACE2 and BALB/c mice. In K18-hACE2 mice, the Alum/CpG group produced a 6.5-fold increase in anti-receptor-binding domain (RBD) IgG antibody titers compared to the Alum group, and generated a comparable level of antibodies even when the antigen amount was reduced by two-thirds, possibly due to the significant activation of germinal center (GC) structures in the central region of the spleen. Different adjuvants induced a variety of binding antibody isotypes. CpG HP021 and Alum/CpG were biased towards Th1/IgG2c, while Alum and MF-59 were biased toward Th2/IgG1. Cytokines IFN-γ, IL-2, and TNF-α were significantly increased in the culture supernatants of splenocytes specifically stimulated in the Alum/CpG group. The antibody responses in BALB/c mice were similar to those in K18-hACE2 mice, but with lower levels of neutralizing antibodies (NAbs). Notably, the Alum/CpG-adjuvanted inactivated vaccine induced a higher number of T cells secreting IFN-γ and IL-2, increased the percentage of effector memory T (TEM) cells among CD8<sup>+</sup> T cells, and effectively protected K18-hACE2 mice from Delta variant challenge. Our results showed that Alum/CpG complex adjuvant significantly enhanced the immune response to inactivated COVID-19 antigens and could induce a long-lasting immune response.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"229 ","pages":"Article 105954"},"PeriodicalIF":4.5,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141533461","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-07-02DOI: 10.1016/j.antiviral.2024.105955
Biplav Shrestha , Sisi Yang , Lauren Griffith , Julia Ma , Fuxuan Wang , Hui Liu , Qiong Zhao , Yanming Du , Jiming Zhang , Jinhong Chang , Ju-Tao Guo
{"title":"Discovery of hepatitis B virus subviral particle biogenesis inhibitors from a bioactive compound library","authors":"Biplav Shrestha , Sisi Yang , Lauren Griffith , Julia Ma , Fuxuan Wang , Hui Liu , Qiong Zhao , Yanming Du , Jiming Zhang , Jinhong Chang , Ju-Tao Guo","doi":"10.1016/j.antiviral.2024.105955","DOIUrl":"10.1016/j.antiviral.2024.105955","url":null,"abstract":"<div><p>High levels of hepatitis B virus (HBV) surface antigen (HBsAg) in the blood of chronic HBV carriers are considered to drive the exhaustion of antigen-specific T and B lymphocytes and thus responsible for the persistence of infection. Accordingly, therapeutic elimination of HBsAg may facilitate the activation of adaptive antiviral immune responses against HBV and achieve a functional cure of chronic hepatitis B. We discovered recently that an amphipathic alpha helix spanning W156 to R169 of HBV small envelope (S) protein plays an essential role in the morphogenesis of subviral particles (SVPs) and metabolism of S protein. We thus hypothesized that pharmacological disruption of SVP morphogenesis may induce intracellular degradation of S protein and reduce HBsAg secretion. To identify inhibitors of SVP biogenesis, we screened 4417 bioactive compounds with a HepG2-derived cell line expressing HBV S protein and efficiently secreting small spherical SVPs. The screen identified 24 compounds that reduced intracellular SVPs and secreted HBsAg in a concentration-dependent manner. However, 18 of those compounds inhibited the secretion of HBsAg and HBeAg in HBV replicon transfected HepG2 cells at similar efficiency, suggesting each of those compounds may disrupt a common cellular function required for the synthesis and/or secretion of these viral proteins. Interestingly, lycorine more efficiently inhibited the secretion of HBsAg in HepG2 cells transfected with HBV replicons, HepG2.2.15 cells and HBV infected - HepG2 cells expressing sodium taurocholate cotransporting polypeptide (NTCP). The structure activity relationship and antiviral mechanism of lycorine against HBV have been determined.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"228 ","pages":"Article 105955"},"PeriodicalIF":4.5,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141533462","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-07-01DOI: 10.1016/j.antiviral.2024.105953
{"title":"The sensitivity of HIV-1 gp120 polymorphs to inhibition by temsavir correlates to temsavir binding on-rate","authors":"","doi":"10.1016/j.antiviral.2024.105953","DOIUrl":"10.1016/j.antiviral.2024.105953","url":null,"abstract":"<div><p>Temsavir binds directly to the HIV-1 envelope glycoprotein gp120 and selectively inhibits interactions between HIV-1 and CD4 receptors. Previous studies identified gp120 amino acid positions where substitutions are associated with reduced susceptibility to temsavir. The mechanism by which temsavir susceptibility is altered in these envelope glycoproteins was evaluated. Pseudoviruses encoding gp120 substitutions alone (S375H/I/M/N, M426L, M434I, M475I) or in combination (S375H + M475I) were engineered on a wild-type JRFL background. Temsavir-gp120 and CD4-gp120 binding kinetics and ability of temsavir to block CD4-gp120 binding were evaluated using the purified polymorphic gp120 proteins and a Creoptix® WAVE Delta grating-coupled interferometry system. Fold-change in half-maximal inhibitory concentration (IC<sub>50</sub>) in JRFL-based pseudoviruses containing the aforementioned polymorphisms relative to that of wild-type ranged from 4-fold to 29,726-fold, while temsavir binding affinity for the polymorphic gp120 proteins varied from 0.7-fold to 73.7-fold relative to wild-type gp120. Strong correlations between temsavir IC<sub>50</sub> and temsavir binding affinity (<em>r</em> = 0.7332; <em>P</em> = 0.0246) as well as temsavir binding on-rate (<em>r</em> = −0.8940; <em>P</em> = 0.0011) were observed. Binding affinity of gp120 proteins for CD4 varied between 0.4-fold and 3.1-fold compared with wild-type gp120; no correlations between temsavir IC<sub>50</sub> and CD4 binding kinetic parameters were observed. For all polymorphic gp120 proteins, temsavir was able to fully block CD4 binding; 3 polymorphs required higher temsavir concentrations. Loss of susceptibility to temsavir observed for gp120 polymorphisms strongly correlated with reductions in temsavir binding on-rate. Nonetheless, temsavir retained the ability to fully block CD4-gp120 engagement given sufficiently high concentrations.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"229 ","pages":"Article 105953"},"PeriodicalIF":4.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141496977","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-06-28DOI: 10.1016/j.antiviral.2024.105952
Jonna B. Westover , Kevin W. Bailey , Samantha R. Wasson , Kirsten M. Boardman , Kurt H. Lustig , Sean M. Amberg , Brian B. Gowen
{"title":"Coadministration of LHF-535 and favipiravir protects against experimental Junín virus infection and disease","authors":"Jonna B. Westover , Kevin W. Bailey , Samantha R. Wasson , Kirsten M. Boardman , Kurt H. Lustig , Sean M. Amberg , Brian B. Gowen","doi":"10.1016/j.antiviral.2024.105952","DOIUrl":"10.1016/j.antiviral.2024.105952","url":null,"abstract":"<div><p>Argentine hemorrhagic fever, caused by Junín virus (JUNV), is the most common of the South American arenaviral hemorrhagic fevers. The disease has a case fatality rate of 15–30% in untreated patients. Although early intervention with immune plasma is effective, diminishing stocks and limited availability outside of Argentina underscores the need for new therapeutics. Ideally, these would be broadly active agents effective against all the pathogenic arenaviruses. The fusion inhibitor LHF-535 and the nucleoside analog favipiravir have shown promise in animal models of Lassa fever, a disease endemic in parts of Africa and the most prominent of the arenaviral hemorrhagic fevers. Against JUNV, a high dose of favipiravir is required to achieve protection in the gold-standard guinea pig infection model. Here, we demonstrate a synergistic effect by the coadministration of LHF-535 with a sub-optimal dose of favipiravir in guinea pigs challenged with JUNV. Administered individually, LHF-535 and sub-optimal favipiravir only delayed the onset of severe disease. However, combined dosing of the drugs afforded complete protection against lethal JUNV infection in guinea pigs. The benefits of the drug combination were also evident by the absence of viremia and infectious virus in tissues compared to guinea pigs treated with only the placebos. Thus, combined targeting of JUNV-endosomal membrane fusion and the viral polymerase with pan-arenaviral LHF-535 and favipiravir may expand their indication beyond Lassa fever, providing a significant barrier to drug resistance.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"229 ","pages":"Article 105952"},"PeriodicalIF":4.5,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141465811","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-06-28DOI: 10.1016/j.antiviral.2024.105951
Nathalia Williams , Filo Silva , Mirco Schmolke
{"title":"Harnessing host enhancers of SARS-CoV-2 entry as novel targets for antiviral therapy","authors":"Nathalia Williams , Filo Silva , Mirco Schmolke","doi":"10.1016/j.antiviral.2024.105951","DOIUrl":"10.1016/j.antiviral.2024.105951","url":null,"abstract":"<div><p>The WHO declared the official end of the SARS-CoV-2 caused public health emergency on May 5th, 2023, after two years in which the virus infected approximately 750 Mio individuals causing estimated up to 7 Mio deaths. Likely, the virus will continue to evolve in the human population as a seasonal respiratory pathogen. To now prevent severe infection outcomes in vulnerable individuals, effective antivirals are urgently needed to complement the protection provided by vaccines. SARS-CoV-2 enters its host cell via ACE2 mediated membrane fusion, either at the plasma membrane, if the protease TMPRSS2 is present or via the endosome, in a cathepsin dependent fashion. A small number of positive regulators of viral uptake were described in the literature, which are potentially useful targets for host directed antiviral therapy or biomarkers indicating increased or diminished susceptibility to infection. We identified here by cell surface proximity ligation novel proteins, required for efficient virion uptake. Importantly, chemical inhibition of one of these factors, SLC3A2, resulted in robust reduction of viral replication, to that achieved with a TMPRSS2 inhibitor. Our screen identified new host dependency factors for SARS-CoV-2 entry, which could be targeted by novel antiviral therapies.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"228 ","pages":"Article 105951"},"PeriodicalIF":4.5,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0166354224001608/pdfft?md5=2a1e82f68b70f456466351d6a90af298&pid=1-s2.0-S0166354224001608-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141465813","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Antiviral researchPub Date : 2024-06-27DOI: 10.1016/j.antiviral.2024.105950
Shuxuan Zheng , Georges M.G.M. Verjans , Anouk Evers , Ellen van den Wittenboer , Jeroen H.T. Tjhie , Robert Snoeck , Emmanuel J.H.J. Wiertz , Graciela Andrei , Jeroen J.A. van Kampen , Robert Jan Lebbink
{"title":"CRISPR/Cas9-mediated genome editing of the thymidine kinase gene in a clinical HSV-1 isolate identifies F289S as novel acyclovir-resistant mutation","authors":"Shuxuan Zheng , Georges M.G.M. Verjans , Anouk Evers , Ellen van den Wittenboer , Jeroen H.T. Tjhie , Robert Snoeck , Emmanuel J.H.J. Wiertz , Graciela Andrei , Jeroen J.A. van Kampen , Robert Jan Lebbink","doi":"10.1016/j.antiviral.2024.105950","DOIUrl":"10.1016/j.antiviral.2024.105950","url":null,"abstract":"<div><p>Herpes simplex virus type 1 (HSV-1) is a neurotropic alphaherpesvirus that establishes a lifelong infection in sensory neurons of infected individuals, accompanied with intermittent reactivation of latent virus causing (a)symptomatic virus shedding. Whereas acyclovir (ACV) is a safe and highly effective antiviral to treat HSV-1 infections, long-term usage can lead to emergence of ACV resistant (ACV<sup>R</sup>) HSV-1 and subsequently ACV refractory disease. Here, we isolated an HSV-1 strain from a patient with reactivated herpetic eye disease that did not respond to ACV treatment. The isolate carried a novel non-synonymous F289S mutation in the viral <em>UL23</em> gene encoding the thymidine kinase (TK) protein. Because ACV needs conversion by viral TK and subsequently cellular kinases to inhibit HSV-1 replication, the <em>UL23</em> gene is commonly mutated in ACV<sup>R</sup> HSV-1 strains. The potential role of the F289S mutation causing ACV<sup>R</sup> was investigated using CRISPR/Cas9-mediated HSV-1 genome editing. Reverting the F289S mutation in the original clinical isolate to the wild-type sequence S289F resulted in an ACV-sensitive (ACV<sup>S</sup>) phenotype, and introduction of the F289S substitution in an ACV<sup>S</sup> HSV-1 reference strain led to an ACV<sup>R</sup> phenotype. In summary, we identified a new HSV-1 TK mutation in the eye of a patient with ACV refractory herpetic eye disease, which was identified as the causative ACV<sup>R</sup> mutation with the aid of CRISPR/Cas9-mediated genome engineering technology. Direct editing of clinical HSV-1 isolates by CRISPR/Cas9 is a powerful strategy to assess whether single residue substitutions are causative to a clinical ACV<sup>R</sup> phenotype.</p></div>","PeriodicalId":8259,"journal":{"name":"Antiviral research","volume":"228 ","pages":"Article 105950"},"PeriodicalIF":4.5,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0166354224001591/pdfft?md5=31495184122f2c62f08dbf159f6733bb&pid=1-s2.0-S0166354224001591-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141465812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}