Antiviral Research xxx (2005) xxx xxx. Short communication

Similar documents
Flu, Avian Flu and emerging aspects (H1N1 resistance)

Medicinal Chemistry Feature Molecule

J.A.L. Ives a, *, J.A. Carr a, D.B. Mendel b, C.Y. Tai b, R. Lambkin c,1, L. Kelly c, J.S. Oxford c, F.G. Hayden d, N.A. Roberts a. 1.

Influenza A Virus Hemagglutinin Antibody Escape Promotes Neuraminidase Antigenic Variation and Drug Resistance

A study of oseltamivir-resistant influenza viruses in Thailand,

Review. The antiviral resistance of influenza virus. Vanessa Escuret 1,2, Olivier Ferraris 2 & Bruno Lina 1,2

Influenza virus.

Micro-plaque assay of influenza virus sensitivity to neuraminidase inhibitors

Emergence of Multidrug-Resistant Influenza A(H1N1)pdm09 Virus Variants in an Immunocompromised Child Treated With Oseltamivir and Zanamivir

Influenza Virus Genotypes Circulating In Central Greece During And Vaccine Strain Match

10 3 pfu A B 2004/ FluA. Viral Shedding. Viral Shedding. Frank Houston Family Study 2005/

INFLUENZA VIRUS. Influenza virus

Antiviral-resistant B viruses with a novel mutation detected by antiviral resistance surveillance in Japan

Detection and management of antiviral resistance for influenza viruses

Ralf Wagner Paul-Ehrlich-Institut

Study of Specific Oligosaccharide Structures Related with Swine Flu (H1N1) and Avian Flu, and Tamiflu as Their Remedy

Palindromes drive the re-assortment in Influenza A

TREATING MEXICAN (NOVEL) INFLUENZA IN AN IMMUNOCOMPROMISED PATIENT

Existence of reassortant A (H1N2) swine influenza viruses in Saitama Prefecture, Japan

INFLUENZA. Rob Young (James. J. Reid) Faculty of Medicine University of Auckland (Otago)

ECDC TECHNICAL REPORT

Influenza antiviral susceptibility: methods and challenges to detect resistant virus

Influenza virus.

Reassortment of influenza A virus genes linked to PB1 polymerase gene

NEXT GENERATION SEQUENCING OPENS NEW VIEWS ON VIRUS EVOLUTION AND EPIDEMIOLOGY. 16th International WAVLD symposium, 10th OIE Seminar

Evolution of influenza

on September 26, 2018 by guest

INFLUENZA WEEKLY UPDATE

Characterization of drug-resistant influenza A(H1N1) and A(H3N2) variants selected in vitro with laninamivir.

INFLUENZA VIRUS. INFLUENZA VIRUS CDC WEBSITE

Balanced Hemagglutinin and Neuraminidase Activities Are Critical for Efficient Replication of Influenza A Virus

In Vitro Generation of Neuraminidase Inhibitor Resistance in A(H5N1) Influenza Viruses

The Anti-Influenza Virus Agent 4-GU-DANA (Zanamivir) Inhibits Cell Fusion Mediated by Human Parainfluenza Virus and Influenza Virus HA

Alanine-scanning mutagenesis for revealing the role of highly conserved regions of influenza A virus neuraminidase

Characterization of two distinct neuraminidases from avian-origin human-infecting H7N9 influenza viruses

Hemagglutinin Receptor Binding Avidity Drives Influenza A Virus Antigenic Drift

Design and antiviral properties of influenza virus neuraminidase inhibitors

Identification of New Influenza B Virus Variants by Multiplex Reverse Transcription-PCR and the Heteroduplex Mobility Assay

Public health relevant virological features of Influenza A(H7N9) causing human infection in China

Review Influenza virus susceptibility and resistance to oseltamivir

Michael M. Kaminski, Annette Ohnemus, Peter Staeheli *, and Dennis Rubbenstroth *

Zanamivir Susceptibility Monitoring and Characterization of Influenza Virus Clinical Isolates Obtained during Phase II Clinical Efficacy Studies

Journal of Ethnic Foods

Orthomyxoviridae and Paramyxoviridae. Lecture in Microbiology for medical and dental medical students

Report on virological surveillance of influenza activity in the Romania 2014/15 season

What s New in Flu? An Update on Influenza Prevention and Treatment

Alkaline Phosphatase Assay Kit (Fluorometric)

Virological surveillance of the Influenza A(H1N1)2009 pandemic: the role of the Belgian National Influenza Centre

Influenza Update N 157

NIH Public Access Author Manuscript Science. Author manuscript; available in PMC 2010 December 1.

Prophylaxis and Treatment for Influenza among the Elderly

ESwab challenges influenza virus propagation in cell cultures

Original Article Development and Sequence Analysis of a Cold-Adapted Strain of Influenza A/New Caledonia/20/1999(H1N1) Virus

Spread of Influenza A(H1N1) oseltamivir-resistant viruses in Africa in 2008 confirmed by multiple introductions in Senegal

Ab initio and DFT investigation of C4 & C7 position of sialidase antiviral inhibitor

A Novel Duplex Real-Time Reverse-Transcription PCR Assay for the Detection of Influenza A and the Novel Influenza A(H1N1) Strain

INFLUENZA VACCINATION AND MANAGEMENT SUMMARY

Neuraminidase Inhibitors for Influenza

Attaching zanamivir to a polymer markedly enhances its activity against drug-resistant strains of influenza a virus

AGLOBAL EMERGENCE AND

The Cold, the Flu or INFLUENZA!

Influenza 2009: Not Yet The Perfect Storm

Fluorometric Measurement of Neuraminidase Activity of Influenza Viruses*)

Weekly influenza surveillance overview

Diagnosing and managing

External quality assessment for influenza antiviral susceptibility for the European Reference Laboratory Network for human influenza

An Exploration to Determine if Fab Molecules are Efficacious in Neutralizing Influenza H1 and H3 Subtypes. Nick Poulton June September 2012

Influenza surveillance and pandemic preparedness - a global challenge Anne Kelso

Influenza A H1N1 HA ELISA Pair Set

Technology Overview. Summary

2009 H1N1 Influenza ( Swine Flu ) Hemagglutinin ELISA kit

The war against influenza: discovery and development of sialidase inhibitors

ph1n1 H3N2: A Novel Influenza Virus Reassortment

Avian Influenza A H5N8

University of Groningen. Computational studies of influenza hemagglutinin Boonstra, Sander

in control group 7, , , ,

Lipoprotein Lipase Activity Assay Kit (Fluorometric)

Preparing for the Fall Flu Season. Jonathan Gubbay Medical Microbiologist Public Health Laboratory OAHPP

Emergence and Fixing of Antiviral Resistance in Influenza A Via Recombination and Hitch Hiking. Henry L Niman

Antigenic and genetic diversity among swine inf luenza viruses in Europe

دکتر بهروز نقیلی استاد بیماریهای عفونی مرکس تحقیقات بیماریهای عفونی و گرمسیری پاییس 88

Influenza A H1N1 (Swine Flu 2009) Hemagglutinin / HA ELISA Pair Set

WHO NIC at Research Institute of Influenza and D.I. Ivanovsky Institute of Virology INTEGRATED DATA OF INFLUENZA MORBIDITY AND DIAGNOSIS

Isolation of an Egg-Adapted Influenza A(H3N2) Virus without Amino Acid Substitutions at the Antigenic Sites of Its Hemagglutinin

November 5 to 11, 2017 (Week 45)

INFLUENZA VIRUS. Influenza virus

Influenza A Virus Neuraminidase: Regions of the Protein Potentially Involved in Virus Host Interactions

Antiviral Therapy 2015; 20:49 55 (doi: /IMP2798)

Review Article A Critical Prospect of Structural Designing of Avian Influenza A/H5N1 Neuraminidase Inhibitors That Evade Tamiflu Resistance

Influenza viruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Detection of neuraminidase-inhibiting antibodies for measurement of Influenza vaccine immunogenicity

Alberta Health. Seasonal Influenza in Alberta. 2012/2013 Season. Surveillance and Assessment Branch. November Government of Alberta 1

Competitive Fitness of Oseltamivir-Sensitive and -Resistant Highly Pathogenic H5N1 Influenza Viruses in a Ferret Model

6.2 Pandemic influenza

Protein Modeling Event

Avian Influenza: Armageddon or Hype? Bryan E. Bledsoe, DO, FACEP The George Washington University Medical Center

December 3 to 9, 2017 (Week 49)

Q: If antibody to the NA and HA are protective, why do we continually get epidemics & pandemics of flu?

1918 Influenza; Influenza A, H1N1. Basic agent information. Section I- Infectious Agent. Section II- Dissemination

Transcription:

Antiviral Research xxx (2005) xxx xxx 3 4 5 6 7 8 Short communication Sensitivity of influenza viruses to zanamivir and oseltamivir: A study performed on viruses circulating in France prior to the introduction of neuraminidase inhibitors in clinical practice O. Ferraris, N. Kessler, B. Lina Laboratoire de Virologie, Centre National de Référence pour la Grippe (France-Sud), Domaine Rockefeller, 8Avenue Rockefeller, 69373 Lyon cedex 08, France Received 18 March 2005; accepted 13 July 2005 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 Abstract Influenza virus neuraminidase inhibitors (NAIs) were introduced in clinical practice in various parts of the world since 1999 but were only scarcely distributed in France. Prior to the generalization of zanamivir and oseltamivir utilization in our country, we decided to test a large panel of influenza strains to establish the baseline sensitivity of these viruses to anti-neuraminidase drugs, based upon a fluorometric neuraminidase enzymatic test. Our study was performed on clinical samples collected by practitioners of the GROG network (Groupe Régional d Observation de la Grippe) in the south of France during the 2002 2003 influenza season. Out of 355 isolates tested in the fluorometric neuraminidase activity assay, 267 isolates could be included in inhibition assay against anti-neuraminidase drugs. Differences in IC 50 range were found according to the subtype and the anti-neuraminidase drug. Influenza B and A/H1N1 viruses appeared to be more sensitive to zanamivir than to oseltamivir (mean B IC 50 values: 4.19 nm versus 13 nm; mean H1N1 IC 50 values: 0.92 nm versus 1.34 nm), while A/H1N2 and A/H3N2 viruses were more sensitive to oseltamivir than to zanamivir (mean H3N2 IC 50 values: 0.67 nm versus 2.28 nm; mean H1N2 IC 50 values: 0.9 nm versus 3.09 nm). Out of 128 N2 carrying isolates, 10 isolates had zanamivir or oseltamivir IC 50 values in upper limits compared to their respective data range. Sequencing of the neuraminidase of these outliers N2 highlighted several mutations, but none of them were associated with resistance to neuraminidase inhibitors. 2005 Published by Elsevier B.V. Keywords: Influenza; Neuraminidase inhibitors; Resistance Influenza viruses contain two glycoproteins on their surface, hemagglutinin (HA) and neuraminidase (NA), which recognize sialic acid on host cell glycoconjugates. HA binds to terminal sialic acid groups on cell surface glycoconjugates leading to attachment and subsequent penetration of the virus into cells (Wiley and Skehel, 1987), while NA possesses an enzymatic activity that removes sialic acid from glycoconjugates, facilitating the release of progeny virions from infected cells and preventing the aggregation of progeny virions (Griffin et al., 1983; Liu et al., 1995). The emergence of annual influenza epidemics and sporadic influenza pandemics are mainly a consequence of rapid rates of evolution in the genes that encode HA and NA, the two major antigenic Corresponding author. Tel.: +33 4 78 77 70 29; fax: +33 4 78 01 48 87. E-mail address: Ferraris@sante.univ-lyon1.fr (O. Ferraris). 1 0166-3542/$ see front matter 2005 Published by Elsevier B.V. 2 doi:10.1016/j.antiviral.2005.07.004 determinants of influenza virus. These antigenic variations 39 drift/shift of influenza viruses produce new virus strains 40 that may not be recognized by antibodies to earlier influenza 41 strains. In order to avoid failure of immune protection in vac- 42 cinated people, the influenza vaccine is updated on a yearly 43 basis to keep up with changes in influenza viruses. 44 Antiviral agents can be useful during epidemics and pan- 45 demics. New drug design strategies allowed the development 46 and subsequent licensing of two anti-influenza A and B drugs, 47 zanamivir (von Itzstein et al., 1993) and oseltamivir (Li et 48 al., 1998) that inhibit virus replication in vitro and in vivo. 49 These drugs act by inhibiting neuraminidase activity, prevent- 50 ing release of progeny virions budding out from cell surface. 51 Neuraminidase was chosen as a suitable drug target because 52 NA plays a major role in influenza virus propagation, and 53 the amino acids residues of the active site interacting directly 54

2 O. Ferraris et al. / Antiviral Research xxx (2005) xxx xxx 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 with the substrate or surrounding the central active site of the enzyme are strictly conserved (Burmeister et al., 1991; Taylor and von Itzstein, 1994). Introduction of a new antiviral drug in clinical practice requires suitable tools to monitor antiviral susceptibility to take account of the probable emergence of drug-resistant variants. Viruses resistant to zanamivir and oseltamivir carboxylate have been generated in vitro, enabling the characterization of three mechanisms involved in the resistance: substitution in the NA, in HA or in both glycoproteins. NA variants can be found after in vitro or in vivo studies (see review by Gubareva, 2004), NA mutations such as R292K, E119G, and H274Y have been selected after several passages of influenza viruses in cell culture in the presence of either zanamivir or oseltamivir (Tai et al., 1998; Barnett et al., 2000; Gubareva et al., 2001). Other NA variants like R152K were isolated from patients (Gubareva et al., 1998). Neuraminidase inhibitor (NAI)-resistant mutants can be detected in the NA inhibition assays. Mutations in HA led to a reduced efficiency of virus binding to sialic acids and then to a decrease in dependence of virus on NA function (Staschke et al., 1995; McKimm-Breschkin et al., 1996; Gubareva et al., 1996). Such resistant variants can be detected in cell based assays using MDCK SIAT1 cells which have been transfected for overexpressing Sia 2 6 receptors (Matrosovich et al., 2003), and by the analysis of HA gene sequences. Another mechanism of resistance involves substitutions in the conserved residues in the NA enzyme active site with compensatory mutations on the HA glycoprotein (McKimm-Breschkin et al., 1996; Gubareva et al., 1996). Virus replication in MDCK cells of resistant variants with NA mutations is reduced, and can be partly rescued by HA compensatory mutations (McKimm-Breschkin et al., 1998). However, most of these viruses showed reduced infectivity and virulence in animal models (Tai et al., 1998; Blick et al., 1998), suggesting that this type of viruses would be severely compromised with a very low clinical impact in man (Carr et al., 2002). The present study on sensitivity of influenza viruses to zanamivir and oseltamivir was initiated prior to the generalization of the use of anti-neuraminidase compounds in the French population. For this aim, 355 clinical isolates obtained by culture of throat and nasal swabs on MDCK cells (Lina et al., 1996), were provided by the GROG network for further analysis in fluorometric neuraminidase test. Out of 355 isolates, 267 viruses were found positive for NA activity and were subsequently introduced in fluorometric NA inhibition test (Tisdale, 2000). Such a test, contrary to plaque reduction assay, was shown uniformly sensitive independently of the cell substrate used for growing virus isolates (Abed et al., 2002; Barnett et al., 2000). The fluorometric NA inhibition test restricted the detection of potential resistant viruses to those viruses mutated in the NA gene. Indeed, the accurate detection of HA-mediated resistant viruses was not considered due to the lack of reliable cell culture based assay when starting this study. Recently, Matrosovich et al. (2003) reported an improvement in the sensitivity of human Table 1 Neuraminidase activity (nmol/h/ml) of influenza isolates in the fluorometric assay N isolates Mean Max Min S.D. B 80 962.13 3076 179 616.55 A/H3N2 128 658.54 2485 71 407.98 A/H1N2 50 764.1 2267 216 415.58 A/H1N1 97 32.6 31 0 39.8 A/H1N1 a 9 14.3 31 0 12.2 A/H1N1 a + BSA 5 mg/ml 9 64.2 107.5 43.5 19.4 a Nine isolates out of 97 A/H1N1 isolates were chosen at random and analyzed in parallel with and without addition of BSA in the virus harvest. influenza A and B viruses to the neuraminidase inhibitor 111 oseltamivir carboxylate, when using MDCK cells transfected 112 for over-expressing Sia 2 6 virus receptors. The fluoromet- 113 ric NA test that we used in this study was a modification of 114 the technique previously described by Potier et al. (1979). 115 Briefly, serial two-fold dilutions of reference strains and 116 isolates (HA titer 16) in 32.5 mm MES (2-(N-morpholino) 117 ethanesulfonic acid, sodium salt, Mw 217.2, Sigma Aldrich) 118 ph 5.8, 4 mm CaCl 2 buffer were prepared in a 96-well flat 119 bottom plates. Plates were gently shaked on a mechanical 120 vibrator after the addition of a 100 M working solution 121 of 2 -(4-methylumbelliferyl)- -d-n-acetylneuraminic acid, 122 sodium salt (MUN, MW 489.4, Sigma Aldrich, St. Louis, 123 MO) prepared in 32.5 mm MES ph 5.8, 4 mm CaCl 2 buffer 124 and were then incubated for 1 h at 37 C. The reaction was 125 stopped by adding 150 l per well of 50 mm glycine buffer 126 ph 10.4. Fluorescence was measured in a DYNEX, MRX 127 fluorometer with an excitation wavelength of 355 nm and 128 an emission wavelength of 460 nm. Both reference sensitive 129 and reference resistant control viruses were included in each 130 assay. These controls were kindly provided by Glaxo Smith 131 Kline (B/Beijing/1/87) and Roche (A/Sydney/05/97). Resis- 132 tant viruses have a specific mutation in the NA gene, position 133 E119G (Barnett et al., 2000) or R292K (Gubareva et al., 1997; 134 McKimm-Breschkin et al., 2003), respectively. 135 Relative fluorescent units (RFUs) of samples were mea- 136 sured and quantification of NA activity was deduced from 137 comparison of sample RFUs values with a standard curve 138 established by using 4-methylumbelliferone (4-Mu) sodium 139 salt. Contrary to influenza viruses A/H3N2, A/H1N2, and B, 140 A/H1N1 isolates showed very low NA activity when using 141 standard infection medium (Table 1). Addition of 5 mg/ml 142 BSA to the medium on time of virus harvest was shown to 143 enhance and stabilize NA N1 activity. Such a technical mod- 144 ification was implemented for the 2003 2004 isolates. Due 145 to the lack of NA activity A/H1N1 isolates from the 2002 to 146 2003 season, 88 isolates could not be introduced in the NA 147 inhibition test against NA inhibitors. In the perspective of a 148 continuous monitoring the sensitivity of influenza isolates to 149 anti-neuraminidase drugs, an interesting approach would be 150 to use MDCK SIAT1 cells for isolating viruses from clinical 151 swabs. Indeed, Matrosovich et al. (2003) who established this 152 modified MDCK cell line showed that over-expression of Sia 153

O. Ferraris et al. / Antiviral Research xxx (2005) xxx xxx 3 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 Table 2a Influenza A and B IC 50 upper limit Subtype A/H3N2 A/H1N2 A/H1N1 B (N = 128) (N = 50) (N =9) (N = 80) Zanamivir (IC 50 (nm)) Mean 2.28 3.09 0.92 4.19 Max 5.97 6.70 1.09 11.90 Upper limit 5.6 6.4 1.2 13.4 Oseltamivir carboxylate (IC 50 (nm)) Mean 0.67 0.9 1.34 12.99 Max 4.46 2.38 1.62 30.20 Upper limit 2.1 2.2 1.7 33.5 Mean values, maximum data, upper limit value (mean + 2.5 S.D.) for each subtype and for both zanamivir and oseltamivir carboxylate. 2 6 virus receptors was responsible for an increased binding of clinical isolates when compared with MDCK cells. From these results it is possible to expect a better replication of influenza isolates and, as a consequence, a higher NA activity, and an increase in the number of isolates entering in the enzymatic inhibition test with anti-neuraminidase drugs. The neuraminidase activity inhibition assay was performed using a standardized dose of virus (10 nmol/h/ml). This virus dose was incubated with 10-fold dilutions of neuraminidase inhibitors prepared in 32.5 mm MES ph 5.8, 4 mm CaCl 2 buffer (from 30,000 to 0.003 nm) and incubated for 15 min at 37 C. The reaction was started by adding MUN substrate. Plates were incubated for 1 h at 37 C. The reaction was stopped by adding 150 l of 50 mm glycine buffer ph 10.4 per well. Fluorescence was immediately quantified by using a DYNEX, MRX fluorometer. IC 50 values of antiviral drugs were calculated according to the concentration required for reducing NA activity by 50%. Baseline sensitivity of N1, N2 and B viruses were calculated from the mean IC 50 values of non-outliers strains (Table 2a). IC 50 values showed differences according to subtypes and NAI (Fig. 1). Viruses with N2 NA were shown to be more sensitive to oseltamivir carboxylate than to zanamivir inversely to N1 NA viruses and B viruses. These results are in agreement with those reported 177 previously (McKimm-Breschkin et al., 2003; Wetherall et al., 178 2003; Hurt et al., 2004). 179 A total of 80 influenza B isolates and 9 A/H1N1 isolates 180 were introduced in the neuraminidase inhibition test. All of 181 them were found sensitive to zanamivir and oseltamivir car- 182 boxylate as well. Out of 178 N2 carrying viruses, only 10 183 plotted at the upper limit (>mean + 2.5 S.D. values) and con- 184 cerned either zanamivir or oseltamivir carboxylate IC 50 val- 185 ues (Tables 2a and 2b). Two A/H1N2 isolates and six A/H3N2 186 isolates showed zanamivir IC 50 values in the upper limit, cor- 187 responding to an average 1.5-fold reduction of sensitivity, 188 while one A/H1N2 isolate and two A/H3N2 isolates showed 189 oseltamivir carboxylate IC 50 values in the upper limit, corre- 190 sponding to a three-fold and five-fold reduction in sensitivity, 191 respectively. 192 NAs of isolates exhibiting IC 50 values in the upper limit 193 range were subsequently sequenced. RNA was extracted 194 from cell culture supernatants by using QIAamp viral RNA 195 kit (QIAgen, Valencia, CA). Amplified products with spe- 196 cific neuraminidase gene primers (forward primer N2Fp1 197 5 gtgaagatgaatccaaatcaa 3, N2Fp15 5 tacagaaattg- 198 gtcaaagccg 3 and reverse primer N2Rp2 5 gcgaaagct- 199 tatataggc 3, N2Rp4 5 acaaacgcattccgactcctg 3 200 (Eurogentec, Liege, Belgium)) were purified from dntps 201 and primers in excess using the DNA Clean & Concentra- 202 tor 5 kit (Zymo Research Inc., USA) and the GFX TM PCR 203 DNA and Gel band purification Kit (Amersham Biosciences 204 Europe Gmbh, Orsay, France). Sequencing was performed in 205 both directions with 120 pmol purified template and 10 pmol 206 of the primers used in the PCR. Cycle sequencing was car- 207 ried out with fluorescent-labeled dideoxy-chain terminators 208 and the reagents in the BigDye sequencing kit (Applied 209 Biosystems, Foster City, CA). All 10 N2 neuraminidase out- 210 liers exhibited several mutations apart from antigenic sites 211 and apart from phylogenetically informative regions (PIRs) 212 (Fanning et al., 2000). None of these N2 neuraminidases 213 Fig. 1. Box plots of zanamivir and oseltamivir carboxylate IC 50 values for A/H3N2, A/H1N2, A/H1N1 and B isolates obtained in the fluorometric neuraminidase inhibition test. The horizontal mark on either side of the box represent the 5% and 95% confidence limits. The box stretches from the lower hinge (25th percentile) to the upper hinge (75th percentile). The mean is shown as a line across the box.

Table 2b Influenza A and B IC 50 upper limit Virus strains Subtype NA activity (nmol/h/ml) Zanamivir IC 50 (nm) a Zanamivir isolate/sydney b Zanamivir isolate/mean IC 50 c Oseltamivir carboxylate IC50 (nm) a Oseltamivir carboxylate isolate/sydney b Oseltamivir carboxylate isolate/mean IC 50 c A/H3N2 A/H1N2 A/H3N2 A/H1N2 A/Lyon/523/03 H3N2 321 5.72 1.4 2.5 4.09 5.2 6.1 A/Lyon-CHU/8698/03 H3N2 193 2.3 0.6 1 4.46 5.7 6.7 A/Lyon-PAH/1014/03 H3N2 857 5.91 1.5 2.6 0.42 0.5 0.6 A/Lyon-PAH/1070/03 H3N2 377 5.75 1.4 2.5 0.33 0.4 0.5 A/Lyon/771/03 H3N2 159 5.75 1.4 2.5 0.76 1 1.1 A/Poitiers/1194/03 H3N2 686 5.97 1.5 2.6 1 1.3 1.5 A/Lyon/1300/03 H3N2 639 5.59 1.4 2.5 1.43 1.8 2.7 A/Lyon-CHU/8633/03 H1N2 927 6.51 1.6 2.1 0.66 0.8 0.7 A/Poitiers/1355/03 H1N2 1293 3.02 0.8 1 2.38 3 2.6 A/Lyon/1281/03 H1N2 383 6.7 1.7 2.2 0.88 1.1 1 A/Sydney/05/97 H3N2 221.9 3.95 1 1.73 1.28 0.79 1 1.18 0.9 R292K H3N2 52.5 14.07 3.6 6.2 4.55 13 16916 19946 14849 B/Beijing/1/87 wt B 437 8.39 11.64 B/Beijing/1/87 R B 44.2 14000 277 a Selecting of isolates as outliers when IC 50 values were near or above the upper limit value with at least one inhibitor. IC 50 values are the average of at least three determinations for each virus-antiviral combination. b Fold reduction of isolates sensitivity to NA inhibitors: isolates/a/sydney/05/97 IC 50 ratio. c Fold reduction of isolates sensitivity to NA inhibitors compared to the mean IC 50 for the same subtype: isolates/mean IC 50 ratio. Table 3 NA mutations of outlier isolates Virus strains Subtype Amino acids mutations A18S L23F C42F E119G R143V R152K E199K H274Y R292K S332F K431N A/Lyon/523/03 H3N2 A18S L23F C42F R143V A/Lyon-CHU/8698/03 H3N2 A18S L23F C42F R143V A/Lyon-PAH/1014/03 H3N2 S332F A/Lyon-PAH/1070/03 H3N2 R143V A/Lyon/771/03 H3N2 E199K K431N A/Poitiers/1194/03 H3N2 S332F A/Lyon/1300/03 H3N2 R143V E199K A/Lyon-CHU/8633/03 H1N2 E199K K431N A/Poitiers/1355/03 H1N2 E199K K431N A/Lyon/1281/03 H1N2 E199K K431N A/Sydney/05/97 H3N2 R292K H3N2 R292K B/Beijing/1/87 wt B B/Beijing/1/87 R B E119G Amino acids mutations in bold correspond to reference in vitro NA mutations. 4 O. Ferraris et al. / Antiviral Research xxx (2005) xxx xxx

O. Ferraris et al. / Antiviral Research xxx (2005) xxx xxx 5 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 with reduced zanamivir or oseltamivir carboxylate sensitivity, revealed variations on amino acids residues previously correlated with a consistent pattern of altered sensitivity to these inhibitors (Table 3). Nevertheless, while no variation was shared by all 10 NAs, some of them (E199K, K431N and R143V) were associated with four to five viruses in the outlier range. In addition, three mutations, A18S, L23F, and C42F were found in two viruses exhibiting a five-fold reduction in NA sensitivity to oseltamivir carboxylate. The reduction in NA sensitivity of these isolates to anti-neuraminidase drugs was very low when compared with the in vivo (Kiso et al., 2004) or in vitro resistant strains. The use of the fluorometric NA test allowed us to establish the baseline sensitivity of N1, N2, and B influenza virus neuraminidases to neuraminidase inhibitors. Such a baseline was found dependent on both NA type and subtype and NA inhibitors. The reduction in NA sensitivity of the N2 isolates we ranged as outlier viruses, was similar to values previously reported by McSharry et al. (2004) and Hurt et al. (2004). These N2 outliers could not be considered as resistant viruses when considering, first, the limited reduction in NA sensitivity to the inhibitors and, second, the existence of mutations on NA which did not belong to those currently associated with influenza resistance to zanamivir and oseltamivir inhibitors (E119G, R152K, H274Y, and R292K). Acknowledgments We are grateful to J. Jolly, S. Paquier, G. Cartet, and G. Burfin for technical assistance. This work is performed in part with the support of the 6 framework Program in the NoE VIRGIL. References Abed, Y., Bourgault, A.M., Fenton, R.J., Morley, P.J., Gower, D., Owens, I.J., Tisdale, M., Boivin, G., 2002. Characterization of 2 influenza A(H3N2) clinical isolates with reduced susceptibility to neuraminidase inhibitors due to mutations in the hemagglutinin gene. J. Infect. Dis. 186, 1074 1080. Barnett, J.M., Cadman, A., Gor, D., Dempsey, M., Walters, M., Candlin, A., Tisdale, M., Morley, P.J., Owens, I.J., Fenton, R.J., Lewis, A.P., Claas, E.C., Rimmelzwaan, G.F., De Groot, R., Osterhaus, A.D., 2000. Zanamivir susceptibility monitoring and characterization of influenza virus clinical isolates obtained during phase II clinical efficacy studies. Antimicrob. Agents Chemother. 44, 78 87. Blick, T.J., Sahasrabudhe, A., McDonald, M., Owens, I.J., Morley, P.J., Fenton, R.J., McKimm-Breschkin, J.L., 1998. The interaction of neuraminidase and hemagglutinin mutations in influenza virus in resistance to 4-guanidino-Neu5Ac2en. Virology 246, 95 103. Burmeister, W.P., Daniels, R.S., Dayan, S., Gagnon, J., Cusack, S., Ruigrok, R.W., 1991. Sequence and crystallization of influenza virus B/Beijing/1/87 neuraminidase. Virology 180, 266 272. Carr, J., Ives, J., Kelly, L., Lambkin, R., Oxford, J., Mendel, D., Tai, L., Roberts, N., 2002. Influenza virus carrying neuraminidase with reduced sensitivity to oseltamivir carboxylate has altered properties in vitro and is compromised for infectivity and replicative ability in vivo. Antiviral Res. 54, 79 88. Fanning, T.G., Reid, A.H., Taubenberger, J.K., 2000. Influenza A 268 virus neuraminidase: regions of the protein potentially involved in 269 virus host interactions. Virology 276, 417 423. 270 Griffin, J.A., Basak, S., Compans, R.W., 1983. Effects of hexose starvation 271 and the role of sialic acid in influenza virus release. Virology 125, 272 324 334. 273 Gubareva, L.V., 2004. Molecular mechanisms of influenza virus resistance 274 to neuraminidase inhibitors. Virus Res. 103, 199 203. 275 Gubareva, L.V., Webster, R.G., Hayden, F.G., 2001. Comparison of the 276 activities of zanamivir, oseltamivir, and RWJ-270201 against clinical 277 isolates of influenza virus and neuraminidase inhibitor-resistant vari- 278 ants. Antimicrob. Agents Chemother. 45, 3403 3408. 279 Gubareva, L.V., Matrosovich, M.N., Brenner, M.K., Bethell, R.C., Web- 280 ster, R.G., 1998. Evidence for zanamivir resistance in an immunocom- 281 promised child infected with influenza B virus. J. Infect. Dis. 178, 282 1257 1262. 283 Gubareva, L.V., Robinson, M.J., Bethell, R.C., Webster, R.G., 1997. 284 Catalytic and framework mutations in the neuraminidase active site 285 of influenza viruses that are resistant to 4-guanidino-Neu5Ac2en. J. 286 Virol. 71, 3385 3390. 287 Gubareva, L.V., Bethell, R., Hart, G.J., Murti, K.G., Penn, C.R., Webster, 288 R.G., 1996. Characterization of mutants of influenza A virus selected 289 with the neuraminidase inhibitor 4-guanidino-Neu5Ac2en. J. Virol. 290 70, 1818 1827. 291 Hurt, A.C., Barr, I.G., Hartel, G., Hampson, A.W., 2004. Susceptibility 292 of human influenza viruses from Australasia and South East Asia to 293 the neuraminidase inhibitors zanamivir and oseltamivir. Antiviral Res. 294 62, 37 45. 295 Kiso, M., Mitamura, K., Sakai-Tagawa, Y., Shiraishi, K., Kawakami, C., 296 Kimura, K., Hayden, F.G., Sugaya, N., Kawaoka, Y., 2004. Resistant 297 influenza A viruses in children treated with oseltamivir: descriptive 298 study. Lancet 364, 759 765. 299 Li, W., Escarpe, P.A., Eisenberg, E.J., Cundy, K.C., Sweet, C., Jake- 300 man, K.J., Merson, J., Lew, W., Williams, M., Zhang, L., Kim, C.U., 301 Bischofberger, N., Chen, M.S., Mendel, D.B., 1998. Identification of 302 GS 4104 as an orally bioavailable prodrug of the influenza virus neu- 303 raminidase inhibitor GS 4071. Antimicrob. Agents Chemother. 42, 304 647 653. 305 Lina, B., Valette, M., Foray, S., Luciani, J., Stagnara, J., See, D.M., 306 Aymard, M., 1995. Surveillance of community-acquired viral infec- 307 tions due to respiratory viruses in Rhone-Alpes (France) during winter 308 1994 to 1995. J. Clin. Microbiol. 34, 3007 3011. 309 Liu, C., Eichelberger, M.C., Compans, R.W., Air, G.M., 1995. Influenza 310 type A virus neuraminidase does not play a role in viral entry, repli- 311 cation, assembly, or budding. J. Virol. 69, 1099 1106. 312 Matrosovich, M., Matrosovich, T., Carr, J., Roberts, N.A., Klenk, H.D., 313 2003. Overexpression of the alpha-2, 6-sialyltransferase in MDCK 314 cells increases influenza virus sensitivity to neuraminidase inhibitors. 315 J. Virol. 77, 8418 8425. 316 McKimm-Breschkin, J.L., Trivedi, T., Hampson, A., Hay, A., Klimov, 317 A., Tashiro, M., Hayden, F., Zambon, M., 2003. Neuraminidase 318 sequence analysis and susceptibilities of influenza virus clinical iso- 319 lates to zanamivir and oseltamivir. Antimicrob. Agents Chemother. 320 47, 2264 2272. 321 McKimm-Breschkin, J.L., Sahasrabudhe, A., Blick, T.J., McDonald, M., 322 Colman, P.M., Hart, G.J., Bethell, R.C., Varghese, J.N., 1998. Muta- 323 tions in a conserved residue in the influenza virus neuraminidase active 324 site decreases sensitivity to Neu5Ac2en-derived inhibitors. J. Virol. 72, 325 2456 2462. 326 McKimm-Breschkin, J.L., Blick, T.J., Sahasrabudhe, A., Tiong, T., Mar- 327 shall, D., Hart, G.J., Bethell, R.C., Penn, C.R., 1996. Generation and 328 characterization of variants of NWS/G70C influenza virus after in 329 vitro passage in 4-amino-Neu5Ac2en and 4-guanidino-Neu5Ac2en. 330 Antimicrob. Agents Chemother. 40, 40 46. 331 McSharry, J.J., McDonough, A.C., Olson, B.A., Drusano, G.L., 2004. 332 Phenotypic drug susceptibility assay for influenza virus neuraminidase 333 inhibitors. Clin. Diagn. Lab. Immunol. 11, 21 28. 334

6 O. Ferraris et al. / Antiviral Research xxx (2005) xxx xxx 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 Potier, M., Mameli, L., Belisle, M., Dallaire, L., Melancon, S.B., 1979. Fluorometric assay of neuraminidase with a sodium (4-methylumbelliferyl-alpha-d-N-acetylneuraminate) substrate. Anal. Biochem. 94, 287 296. Staschke, K.A., Colacino, J.M., Baxter, A.J., Air, G.M., Bansal, A., Hornback, W.J., Munroe, J.E., Laver, W.G., 1995. Molecular basis for the resistance of influenza viruses to 4-guanidino-Neu5Ac2en. Virology 214, 642 646. Tai, C.Y., Escarpe, P.A., Sidwell, R.W., Williams, M.A., Lew, W., Wu, H., Kim, C.U., Mendel, D.B., 1998. Characterization of human influenza virus variants selected in vitro in the presence of the neuraminidase inhibitor GS 4071. Antimicrob. Agents Chemother. 42, 3234 3241. Taylor, N.R., von Itzstein, M., 1994. Molecular modeling studies on ligand binding to sialidase from influenza virus and the mechanism of catalysis. J. Med. Chem. 37, 616 624. Tisdale, M., 2000. Monitoring of viral susceptibility: new challenges with the development of influenza NA inhibitors. Rev. Med. Virol. 10, 45 55. von Itzstein, M., Wu, W.Y., Kok, G.B., Pegg, M.S., Dyason, J.C., Jin, B., 352 Van Phan, T., Smythe, M.L., White, H.F., Oliver, S.W., Colman, P.M., 353 Varghese, J.N., Ryan, D.M., Woods, J.M., Bethell, R.C., Hotham, V.J., 354 Cameron, J.M., Penn, C.R., 1993. Rational design of potent sialidase- 355 based inhibitors of influenza virus replication. Nature 363, 418 356 423. 357 Wetherall, N.T., Trivedi, T., Zeller, J., Hodges-Savola, C., McKimm- 358 Breschkin, J.L., Zambon, M., Hayden, F.G., 2003. Evalu- 359 ation of neuraminidase enzyme assays using different sub- 360 strates to measure susceptibility of influenza virus clinical iso- 361 lates to neuraminidase inhibitors: report of the neuraminidase 362 inhibitor susceptibility network. J. Clin. Microbiol. 41, 742 363 750. 364 Wiley, D.C., Skehel, J.J., 1987. The structure and function of the 365 hemagglutinin membrane glycoprotein of influenza virus. Annu. Rev. 366 Biochem. 56, 365 394. 367