Assessing the fitness of a dual-antiviral drug resistant human influenza virus in the ferret model

Harry L Stannard, Edin J Mifsud, Steffen Wildum, Sook Kwan Brown, Paulina Koszalka, Takao Shishido, Satoshi Kojima, Shinya Omoto, Keiko Baba, Klaus Kuhlbusch, Aeron C Hurt, Ian G Barr, Harry L Stannard, Edin J Mifsud, Steffen Wildum, Sook Kwan Brown, Paulina Koszalka, Takao Shishido, Satoshi Kojima, Shinya Omoto, Keiko Baba, Klaus Kuhlbusch, Aeron C Hurt, Ian G Barr

Abstract

Influenza antivirals are important tools in our fight against annual influenza epidemics and future influenza pandemics. Combinations of antivirals may reduce the likelihood of drug resistance and improve clinical outcomes. Previously, two hospitalised immunocompromised influenza patients, who received a combination of a neuraminidase inhibitor and baloxavir marboxil, shed influenza viruses resistant to both drugs. Here-in, the replicative fitness of one of these A(H1N1)pdm09 virus isolates with dual resistance mutations (NA-H275Y and PA-I38T) was similar to wild type virus (WT) in vitro, but reduced in the upper respiratory tracts of challenged ferrets. The dual-mutant virus transmitted well between ferrets in an airborne transmission model, but was outcompeted by the WT when the two viruses were co-administered. These results indicate the dual-mutant virus had a moderate loss of viral fitness compared to the WT virus, suggesting that while person-to-person transmission of the dual-resistant virus may be possible, widespread community transmission is unlikely.

Trial registration: ClinicalTrials.gov NCT03684044.

Conflict of interest statement

These authors declare the following competing interests: K.K, S.W., and A.C.H are employees of F. Hoffmann-La Roche. All other authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Virus growth in single and…
Fig. 1. Virus growth in single and multi-stage replication cycles.
MDCK cells were infected with virus isolates containing one or both NA-H275Y and PA-I38T mutations. a Single-stage replication assay (MOI 5) with sampling every 2 h for a total of 12 h. b Multi-stage replication assay (MOI 0.01) and sampled at 6, 24, 30, 48, 54, 72, 78, 96, 102 h. Infectious virus in the supernatant was quantified by TCID50. Each symbol represents the mean amount of virus present at each time point and the error bars depict one standard deviation (SD). Data in each graph is the combination of three independently conducted experiments performed in triplicate. Limit of detection (LOD) is shown as a dotted horizontal line, all values below LOD equal two log10TCID50/mL. No statistically significant differences (adjusted p < 0.05) were observed (Two-way ANOVA with Sidak’s multiple comparisons).
Fig. 2. Viral shedding and lung lobe…
Fig. 2. Viral shedding and lung lobe viral load of WT and NA-H275Y + PA-I38T virus isolate infected ferrets.
Ferrets (n = 8) infected by the intranasal route with 4 log10TCID50/mL (in 500 µL of PBS) with WT (d1 isolate, black) or NA-H275Y + PA-I38T (d10 isolate, red) purified virus isolates were nasally washed daily for 9 days. Half of the ferrets were culled at day five post-inoculation to harvest the five major lung lobes. a The viral shedding from the ferret nasal washes was measured by TCID50 assay. b Viral titre of each homogenised lung lobe aliquot wase enumerated using a TCID50 assay, the fraction representing the number of major lung lobes with detectable viral titres (above LOD) per ferret. Each dot represents the titre in a single lung lobe and the line shows the mean. Error bars represent one SD. LOD is shown as a dotted horizontal line, all values beyond LOD equal zero viral titre. Adjusted p-values and asterisks show statistically significant (p < 0.05) differences (Two-way ANOVA with Sidak’s multiple comparisons).
Fig. 3. Infectious viral shedding in nasal…
Fig. 3. Infectious viral shedding in nasal washes from ferrets infected with WT or NA-H275Y + PA-I38T clinical isolates and in ferrets infected by airborne virus exposure.
Donor ferrets (n = 4) were infected by the intranasal route with 5 log10TCID50/mL of pure WT or NA-H275Y + PA-I38T clinically isolated virus and co-housed with naïve recipient ferret 1 (RF1) separated by an airborne virus-permeable barrier at day 1 (D1) post inoculation. Infected RF1s were then co-housed with RF2 ferrets, enabling a second airborne transmission event to occur. Viral titre mean from nasal washes from donor (a), RF1 (b) and RF2 ferrets (c), by TCID50 assay. Fractions above each symbol represent the number of ferrets shedding virus above LOD (dotted horizontal line), all values below LOD equal zero. No statistically significant (adjusted p < 0.05) differences was observed (Sidak’s Two-way ANOVA multiple comparisons). d Area under the curve analysis was performed for each individual ferret (n = 4) and overall viral shedding was compared between WT (black) and NA-H275Y + PA-I38T (D.Mt, red). e Duration of infectious viral shedding above LOD was tallied for each ferret. f Estimation plot of the combined duration of viral shedding for all donor, RF1 and RF2 ferrets was analysed by Unpaired t-test to display a 95% confidence interval of the difference between the mean duration of shedding for WT and NA-H275Y + PA-I38T infected ferrets. Error bars indicate one SD. Statistically significant differences (p < 0.05) were compared by Tukey’s Ordinary One-way ANOVA.
Fig. 4. Infectious virus titres and pyrosequencing…
Fig. 4. Infectious virus titres and pyrosequencing analysis of ferret nasal washes from 50:50 WT:NA-H275Y + PA-I38T clinical isolate inoculated donor and airborne transmission infected recipient ferrets.
Donor ferrets (n = 4) were infected via the intranasal route with 5 log10TCID50/mL with 50:50 WT: NA-H275Y + PA-I38T clinically isolated virus and co-housed with naïve recipient ferrets (RF1) virus separated by an airborne virus permeable barrier, infected RF1s were then co-housed with RF2 ferrets. Nasal wash samples were titrated by TCID50 assay for each Group A-D Donor-RF1-RF2 transmission pairs, LOD is shown as a dotted horizontal line, and all values beyond LOD equal zero viral titre (a). The relative proportion of virus encoding WT PA (white bars) and PA-I38T (dark blue bars) and WT NA (white bars) and NA-H275Y (light blue bars) were determined by pyrosequencing analysis and are shown side-by-side in bar charts for each donor (b), RF1 (c) and RF2 (d) ferret.
Fig. 5. Summary of whole-genome sequencing on…
Fig. 5. Summary of whole-genome sequencing on ferret inoculum and nasal wash samples from pure WT, pure NA-H275Y + PA-I38T and 50% WT: 50%NA-H275Y + PA-I38T isolate infected ferrets.
Ferret inoculum and nasal wash samples post inoculation/exposure (PI/E) from pure WT, pure NA-H275Y + PA-I38T or 50% WT: 50%NA-H275Y + PA-I38T virus infected ferrets were sequenced using WGS. Five amino acid (AA) changes (across the HA, NA, PA and PB1 genes) distinguished ferret inoculum WT and NA-H275Y + PA-I38T virus genotypes. The bar charts display the proportion of AA codon changes detected by NGS for each sample associated with the WT (white) and NA-H275Y + PA-I38T (black) virus inoculum for each gene loci (along X-axis; HA-294, NA-275, PA-38, PA-325, PB1-129). Percentage frequency of AA codons is determined from variant bases called with a read depth over 1000 and frequency of greater than 5%. Created with BioRender.com.

References

    1. Iuliano AD, et al. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018;391:1285–1300. doi: 10.1016/S0140-6736(17)33293-2.
    1. WHO. Up to 650000 People Die of Respiratory Diseases Linked to Seasonal Flu Each Year. (2017).
    1. Patel TS, et al. Oseltamivir for pandemic influenza preparation: Maximizing the use of an existing stockpile. Am. J. Infect. control. 2017;45:303–305. doi: 10.1016/j.ajic.2016.09.024.
    1. Harrington WN, Kackos CM, Webby RJ. The evolution and future of influenza pandemic preparedness. Exp. Mol. Med. 2021;53:737–749. doi: 10.1038/s12276-021-00603-0.
    1. Lakdawala SS, et al. Eurasian-origin gene segments contribute to the transmissibility, aerosol release, and morphology of the 2009 pandemic H1N1 influenza virus. PLoS Pathog. 2011;7:e1002443–e1002443. doi: 10.1371/journal.ppat.1002443.
    1. Dong G, et al. Adamantane-resistant influenza a viruses in the world (1902-2013): frequency and distribution of M2 gene mutations. PLoS One. 2015;10:e0119115. doi: 10.1371/journal.pone.0119115.
    1. Lackenby A, et al. Global update on the susceptibility of human influenza viruses to neuraminidase inhibitors and status of novel antivirals, 2016-2017. Antivir. Res. 2018;157:38–46. doi: 10.1016/j.antiviral.2018.07.001.
    1. Okomo-Adhiambo M, et al. Neuraminidase inhibitor susceptibility testing in human influenza viruses: a laboratory surveillance perspective. Viruses. 2010;2:2269–2289. doi: 10.3390/v2102269.
    1. Dharan NJ, et al. Infections with Oseltamivir-resistant influenza A(H1N1) virus in the United States. JAMA. 2009;301:1034–1041. doi: 10.1001/jama.2009.294.
    1. Hayden FG, et al. Baloxavir marboxil for uncomplicated influenza in adults and adolescents. N. Engl. J. Med. 2018;379:913–923. doi: 10.1056/NEJMoa1716197.
    1. Mifsud EJ, Hayden FG, Hurt AC. Antivirals targeting the polymerase complex of influenza viruses. Antivir. Res. 2019;169:104545. doi: 10.1016/j.antiviral.2019.104545.
    1. Hirotsu N, et al. Baloxavir marboxil in Japanese pediatric patients with influenza: safety and clinical and virologic outcomes. Clin. Infect. Dis.: Off. Publ. Infect. Dis. Soc. Am. 2020;71:971–981. doi: 10.1093/cid/ciz908.
    1. Ikematsu H, et al. Baloxavir marboxil for prophylaxis against influenza in household contacts. N. Engl. J. Med. 2020;383:309–320. doi: 10.1056/NEJMoa1915341.
    1. Ison MG, et al. Early treatment with baloxavir marboxil in high-risk adolescent and adult outpatients with uncomplicated influenza (CAPSTONE-2): a randomised, placebo-controlled, phase 3 trial. Lancet Infect. Dis. 2020;20:1204–1214. doi: 10.1016/S1473-3099(20)30004-9.
    1. Omoto S, et al. Characterization of influenza virus variants induced by treatment with the endonuclease inhibitor baloxavir marboxil. Sci. Rep. 2018;8:9633. doi: 10.1038/s41598-018-27890-4.
    1. Uehara T, et al. Treatment-emergent influenza variant viruses with reduced baloxavir susceptibility: Impact on clinical and virologic outcomes in uncomplicated influenza. J. Infect. Dis. 2020;221:346–355.
    1. Kawai N, et al. Clinical effectiveness of Oseltamivir and Zanamivir for treatment of Influenza A virus subtype H1N1 with the H274Y mutation: a Japanese, multicenter study of the 2007–2008 and 2008–2009 influenza seasons. Clin. Infect. Dis. 2009;49:1828–1835. doi: 10.1086/648424.
    1. Matsuzaki Y, et al. A two-year survey of the oseltamivir-resistant influenza A(H1N1) virus in Yamagata, Japan and the clinical effectiveness of oseltamivir and zanamivir. Virol. J. 2010;7:53. doi: 10.1186/1743-422X-7-53.
    1. Holmes, E. C. et al. Understanding the impact of resistance to influenza antivirals. Clin Microbiol Rev.34, e00224–00220, (2021).
    1. Macesic N, et al. Baloxavir treatment of oseltamivir-resistant influenza A/H1pdm09 in two immunocompromised patients. Transpl. Infect. Dis.: Off. J. Transplant. Soc. 2021;23:e13542. doi: 10.1111/tid.13542.
    1. Salvatore, M. et al. Baloxavir for the treatment of Influenza in allogeneic hematopoietic stem cell transplant recipients previously treated with oseltamivir. Transpl Infect Dis.22, e13336, (2020).
    1. Seki M, Sakai-Tagawa Y, Yasuhara A, Watanabe Y. Adult influenza A (H3N2) with reduced susceptibility to baloxavir or peramivir cured after switching anti-influenza agents. IDCases. 2019;18:e00650. doi: 10.1016/j.idcr.2019.e00650.
    1. Govorkova EA, et al. Global update on the susceptibilities of human influenza viruses to neuraminidase inhibitors and the cap-dependent endonuclease inhibitor baloxavir, 2018–2020. Antivir. Res. 2022;200:105281. doi: 10.1016/j.antiviral.2022.105281.
    1. WHO. Recommended Composition of Influenza Virus Vaccines for Use in the 2022 Southern Hemisphere Influenza Season (WHO, 2021).
    1. WHO. Recommended Composition of Influenza Virus Vaccines for Use in the 2022–23 Northern Hemisphere Influenza Season (WHO, 2022).
    1. Bartlett JA, DeMasi R, Quinn J, Moxham C, Rousseau F. Overview of the effectiveness of triple combination therapy in antiretroviral-naive HIV-1 infected adults. AIDS. 2001;15:1369–1377. doi: 10.1097/00002030-200107270-00006.
    1. Lumby CK, et al. Favipiravir and Zanamivir cleared infection with influenza B in a severely immunocompromised child. Clin. Infect. Dis. 2020;71:e191–e194. doi: 10.1093/cid/ciaa023.
    1. Terrier, O. & Slama-Schwok, A. in Antiviral Drug Discovery and Development (eds Liu, X. et al.) 195–218 (Springer Singapore, 2021).
    1. Park J-H, et al. Baloxavir-oseltamivir combination therapy inhibits the emergence of resistant substitutions in influenza A virus PA gene in a mouse model. Antivir. Res. 2021;193:105126. doi: 10.1016/j.antiviral.2021.105126.
    1. Koszalka, P., George, A., Dhanasekaran, V., Hurt, A. C. & Subbarao, K. Effect of baloxavir and oseltamivir in combination on infection with influenza viruses with PA/I38T or PA/E23K substitutions in the ferret model. mBio 13, e01056–01022, (2022).
    1. Kumar, D. et al. Combining baloxavir marboxil with standard-of-care neuraminidase inhibitor in patients hospitalised with severe influenza (FLAGSTONE): a randomised, parallel-group, double-blind, placebo-controlled, superiority trial. Lancet Infect. Dis.10.1016/S1473-3099(21)00469-2 (2022).
    1. Karlsson EA, et al. Visualizing real-time influenza virus infection, transmission and protection in ferrets. Nat. Commun. 2015;6:6378–6378. doi: 10.1038/ncomms7378.
    1. Moore IN, et al. Severity of clinical disease and pathology in ferrets experimentally infected with influenza viruses is influenced by inoculum volume. J. Virol. 2014;88:13879–13891. doi: 10.1128/JVI.02341-14.
    1. Munster VJ, et al. Pathogenesis and transmission of swine-origin 2009 A(H1N1) influenza virus in ferrets. Science. 2009;325:481–483. doi: 10.1126/science.1177127.
    1. Cowling BJ, et al. Aerosol transmission is an important mode of influenza A virus spread. Nat. Commun. 2013;4:1935. doi: 10.1038/ncomms2922.
    1. Tellier R. Aerosol transmission of influenza A virus: a review of new studies. J. R. Soc. Interface. 2009;6:S783–790. doi: 10.1098/rsif.2009.0302.focus.
    1. Belser JA, Eckert AM, Tumpey TM, Maines TR. Complexities in ferret influenza virus pathogenesis and transmission models. Microbiol. Mol. Biol. Rev. 2016;80:733–744. doi: 10.1128/MMBR.00022-16.
    1. Memoli MJ, et al. Multidrug-resistant 2009 pandemic influenza A(H1N1) viruses maintain fitness and transmissibility in ferrets. J. Infect. Dis. 2011;203:348–357. doi: 10.1093/infdis/jiq067.
    1. Kiso M, et al. Characterization of oseltamivir-resistant 2009 H1N1 pandemic influenza A viruses. PLoS Pathog. 2010;6:e1001079. doi: 10.1371/journal.ppat.1001079.
    1. Duan S, et al. Oseltamivir-resistant pandemic H1N1/2009 influenza virus possesses lower transmissibility and fitness in ferrets. PLoS Pathog. 2010;6:e1001022. doi: 10.1371/journal.ppat.1001022.
    1. Hamelin ME, et al. Reduced airborne transmission of oseltamivir-resistant pandemic A/H1N1 virus in ferrets. Antivir. Ther. 2011;16:775–779. doi: 10.3851/IMP1794.
    1. Imai M, et al. Influenza A variants with reduced susceptibility to baloxavir isolated from Japanese patients are fit and transmit through respiratory droplets. Nat. Microbiol. 2020;5:27–33. doi: 10.1038/s41564-019-0609-0.
    1. Jones JC, et al. Influenza A and B viruses with reduced baloxavir susceptibility display attenuated in vitro fitness but retain ferret transmissibility. Proc. Natl. Acad. Sci. USA. 2020;117:8593–8601. doi: 10.1073/pnas.1916825117.
    1. Lee LY, et al. Evaluating the fitness of PA/I38T-substituted influenza A viruses with reduced baloxavir susceptibility in a competitive mixtures ferret model. PLoS Pathog. 2021;17:e1009527–e1009527. doi: 10.1371/journal.ppat.1009527.
    1. Takashita E, et al. Human-to-human transmission of influenza A(H3N2) virus with reduced susceptibility to Baloxavir, Japan, February 2019. Emerg. Infect. Dis. 2019;25:2108–2111. doi: 10.3201/eid2511.190757.
    1. Hurt AC, et al. Characteristics of a widespread community cluster of H275Y Oseltamivir-resistant A(H1N1)pdm09 influenza in Australia. J. Infect. Dis. 2012;206:148–157. doi: 10.1093/infdis/jis337.
    1. Takashita E, et al. Characterization of a large cluster of influenza A(H1N1)pdm09 viruses cross-resistant to oseltamivir and peramivir during the 2013-2014 influenza season in Japan. Antimicrob. Agents Chemother. 2015;59:2607–2617. doi: 10.1128/AAC.04836-14.
    1. Mohan T, et al. Cluster of Oseltamivir-resistant and hemagglutinin antigenically drifted influenza A(H1N1)pdm09 viruses, Texas, USA, January 2020. Emerg. Infect. Dis. 2021;27:1953–1957. doi: 10.3201/eid2707.204593.
    1. Varble A, et al. Influenza A virus transmission bottlenecks are defined by infection route and recipient host. Cell Host Microbe. 2014;16:691–700. doi: 10.1016/j.chom.2014.09.020.
    1. Lee, L. Y. et al. Evaluating the fitness of PA/I38T-substituted influenza A viruses with reduced baloxavir susceptibility in a competitive mixtures ferret model. PLoS Pathog.17, e1009527 (2020).
    1. Checkmahomed L, et al. Impact of the Baloxavir-resistant polymerase acid I38T substitution on the fitness of contemporary Influenza A(H1N1)pdm09 and A(H3N2) strains. J. Infect. Dis. 2019;221:63–70. doi: 10.1093/infdis/jiz418.
    1. Hurt, A. C. et al. Assessing the viral fitness of Oseltamivir-resistant influenza viruses in Ferrets, using a competitive-mixtures model.J.Virol.84, 9427–9438, (2010).
    1. Seibert CW, et al. Oseltamivir-resistant variants of the 2009 pandemic H1N1 influenza A virus are not attenuated in the guinea pig and ferret transmission models. J. Virol. 2010;84:11219–11226. doi: 10.1128/JVI.01424-10.
    1. Goldhill DH, et al. Favipiravir-resistant influenza A virus shows potential for transmission. PLoS Pathog. 2021;17:e1008937. doi: 10.1371/journal.ppat.1008937.
    1. Richard, M., Herfst, S., Tao, H., Jacobs, N. T. & Lowen, A. C. Influenza A virus reassortment is limited by anatomical compartmentalization following coinfection via distinct routes. J. Virol.10.1128/jvi.02063-17 (2018).
    1. Kalil AC, Thomas PG. Influenza virus-related critical illness: pathophysiology and epidemiology. Crit. Care. 2019;23:258. doi: 10.1186/s13054-019-2539-x.
    1. Nguyen, T. Q., Rollon, R. & Choi, Y. K. Animal models for influenza research: strengths and weaknesses. Viruses10.3390/v13061011 (2021).
    1. Finberg RW, et al. Phase 2b study of Pimodivir (JNJ-63623872) as monotherapy or in combination with Oseltamivir for treatment of acute uncomplicated seasonal influenza A: TOPAZ trial. J. Infect. Dis. 2019;219:1026–1034. doi: 10.1093/infdis/jiy547.
    1. Baz M, Abed Y, McDonald J, Boivin G. Characterization of multidrug-resistant influenza A/H3N2 viruses shed during 1 year by an immunocompromised child. Clin. Infect. Dis. 2006;43:1555–1561. doi: 10.1086/508777.
    1. Ison MG, Gubareva LV, Atmar RL, Treanor J, Hayden FG. Recovery of drug-resistant influenza virus from immunocompromised patients: a case series. J. Infect. Dis. 2006;193:760–764. doi: 10.1086/500465.
    1. van der Vries E, et al. Prolonged influenza virus shedding and emergence of antiviral resistance in immunocompromised patients and ferrets. PLoS Pathog. 2013;9:e1003343. doi: 10.1371/journal.ppat.1003343.
    1. Oh DY, Barr IG, Hurt AC. A novel video tracking method to evaluate the effect of influenza infection and antiviral treatment on ferret activity. PLoS One. 2015;10:e0118780. doi: 10.1371/journal.pone.0118780.
    1. Reed LJ, Muench H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 1938;27:493–497. doi: 10.1093/oxfordjournals.aje.a118408.
    1. Zhou, B. et al. Single-reaction genomic amplification accelerates sequencing and vaccine production for classical and Swine origin human influenza A viruses. J.Virol.83, 10309–10313, (2009).
    1. Shepard SS, et al. Viral deep sequencing needs an adaptive approach: IRMA, the iterative refinement meta-assembler. BMC Genom. 2016;17:708. doi: 10.1186/s12864-016-3030-6.
    1. Leang, S.-K. & Hurt, A. C. Fluorescence-based neuraminidase inhibition assay to assess the susceptibility of influenza viruses to the neuraminidase inhibitor class of antivirals. J .Vis. Exp. 10.3791/55570 (2017).
    1. Koszalka P, Tilmanis D, Roe M, Vijaykrishna D, Hurt AC. Baloxavir marboxil susceptibility of influenza viruses from the Asia-Pacific, 2012–2018. Antivir. Res. 2019;164:91–96. doi: 10.1016/j.antiviral.2019.02.007.
    1. Koszalka P, Farrukee R, Mifsud E, Vijaykrishna D, Hurt AC. A rapid pyrosequencing assay for the molecular detection of influenza viruses with reduced baloxavir susceptibility due to PA/I38X amino acid substitutions. Influenza Other Respir. Viruses. 2020;14:460–464. doi: 10.1111/irv.12725.
    1. Stannard, H. et al. Assessing the fitness of a dual-antiviral drug resistant human influenza virus in the ferret model. figshare 10.6084/m9.figshare.21008614 (2022).

Source: PubMed

3
Předplatit