gms | German Medical Science

GMS Infectious Diseases

Paul-Ehrlich-Gesellschaft für Infektionstherapie e.V. (PEG) (PEG)

ISSN 2195-8831

Influenza viruses – antiviral therapy and resistance

Review Article

Suche in Medline nach

  • corresponding author Susanne Duwe - Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany

GMS Infect Dis 2017;5:Doc04

doi: 10.3205/id000030, urn:nbn:de:0183-id0000307

Veröffentlicht: 25. April 2017

© 2017 Duwe.
Dieser Artikel ist ein Open-Access-Artikel und steht unter den Lizenzbedingungen der Creative Commons Attribution 4.0 License (Namensnennung). Lizenz-Angaben siehe http://creativecommons.org/licenses/by/4.0/.


Abstract

Influenza is a serious and frequently underestimated, but vaccine preventable disease. The adamantane derivates rimantadine and amantadine and the neuraminidase inhibitors zanamivir and oseltamivir are the only antiviral drugs currently approved in Europe for therapy and prophylaxis of influenza infections. Resistance to these drugs occurs due to mutations within the therapeutic target proteins M2 ion channel protein and viral neuraminidase. An unexpected occurrence of oseltamivir-resistant seasonal A(H1N1) viruses was detected in winter 2007/2008. The prevalence of these viruses increased rapidly and nearby all viruses circulating during the following seasons were resistant to oseltamivir. The A(H1N1)pdm09 viruses replaced the former seasonal A(H1N1) subtype during the 2009–2010 influenza season. Fortunately, resistance to neuraminidase inhibitors was detected in A(H1N1)pdm09, A(H3N2) and influenza B viruses only sporadically and was treatment related mostly. Comprehensive analyses of circulating viruses showed a high prevalence of A(H3N2) influenza viruses that are resistant to adamantane derivates since 2004/2005 and a progressive trend in the prevalence of resistant viruses up to 100% in following seasons. The M2 ion channel protein of A(H1N1)pdm09 viruses is associated with the Eurasian avian-like swine lineage and thus show “natural” resistance to adamantane derivates. Therefore, only neuraminidase inhibitors are recommended for influenza treatment today.

This manuscript summarizes the occurrence and spread of antiviral resistant influenza viruses and highlights the importance for developing and/or approving new antiviral compounds.

Keywords: infection, antiviral resistance, neuraminidase, surveillance, monitoring


1 Fundamental knowledge

Influenza viruses belong to the family Orthomyxoviridae that is characterized by the ability to attach on glycoproteins of host cell surfaces and a segmented genome composed of single stranded, negatively orientated ribonucleic acid (–ssRNA). Based on their molecular features and serological characteristics of their nucleoproteins and matrix proteins influenza viruses are divided into three genera: Influenzavirus A, Influenzavirus B and Influenzavirus C [1]. Whereas infections with influenza C viruses are often symptomless in humans, influenza A and B viruses cause annual epidemics known as seasonal flu, and influenza A viruses also cause pandemics at random intervals [2].

Influenza A viruses are zoonotic pathogens that can infect a broad range of species including birds, pigs and humans. According to the antigenic properties of their surface glycoproteins hemagglutinin (HA) and neuraminidase (NA) influenza A viruses are further divided into 18 HA and 11 NA subtypes (H1–H16 and N1–N9 in wild waterfowl, H17, H18 and N10, N11 in bats) [3]. In comparison to influenza A viruses influenza B viruses are less variable. They are antigenically related to either B/Victoria/2/87 or B/Yamagata/16/88 and are distinguished into two lineages which are referred to as the Yamagata and the Victoria lineage [4].

The life cycle of the influenza virus begins with binding of the virus particles to the surface of the host cells. Binding is mediated by the interaction of viral hemagglutinin (HA) with sialyloligosaccharides on proteins and lipids of the cell membranes. Due to receptor-mediated endocytosis the virus is internalized into the host cell enclosed by an endosome. Triggered by low pH in late endosomes and mediated by M2 ion channel, a conformational change of HA induces the fusion of the viral and the endosomal membrane. This triggers the release of uncoated viral ribonucleoprotein (vRNP) complexes into the cytosol of the host cell cytoplasm. After transport of vRNP complexes into the nucleus, replication and transcription follows the amplification of vRNA and synthesis of mRNAs for viral protein synthesis. Newly assembled vRNPs are exported to the cytoplasm and assembled with viral proteins at budding sites within the host cell membrane, followed by the budding and, after cleavage by neuraminidase, release of influenza virions [2].

Minor changes in viral proteins (antigenic-drift) are caused by high genomic variability of influenza viruses. Due to the lack of a proof-reading activity of the viral polymerase point mutations in the vRNA occur that might result in amino acid exchanges of the antigenic epitopes of the HA and NA proteins. This worsens the recognition and neutralization (abolishment of infectivity) of influenza viruses by the host’s immune system antibodies and thus may lead to immune evasive virus variants. Antigenic-drift is the cause for influenza epidemics by weakening the original bond between antigen and antibodies produced by vaccination, or infection induced immunity.

Antigenic shift results from a reassortment between at least two influenza subtypes resulting in a new influenza subtype. This may generate a novel virus able to infect the majority of the population due to their lack of immunity towards the unknown pathogen leading to high infection rates and pandemic spreads [3], [5]. Both mechanisms, point mutations as well as reassortment, can affect the efficacy of anti-influenza drugs.

Influenza occurs globally with an estimated annual attack rate of 5–10% in adults and 20–30% in children. Both influenza A and influenza B viruses cause seasonal epidemics and out-of-season sporadic cases and outbreaks [6]. A typical influenza is marked by sudden onset of fever (≥38.5°C), dry cough, sore throat, body aches and pains, and headaches. Other symptoms may include general fatigue, sweating, rhinorrhea, but also nausea, vomiting, and diarrhea. In more severe cases, infection can lead to pneumonia, bacterial superinfection and even death. Nevertheless, not all patients fall ill with typical symptomatology. The duration of the disease is usually 5–7 days, which can be considerably longer depending on complications and risk factors [7].

Influenza is a vaccine preventable disease and influenza vaccines have been available for use in Europe since the 1960s. Currently most influenza vaccines contain three different influenza strains (trivalent): two influenza A strains (A(H1N1) and A(H3N2) subtypes) and one influenza B strain (Victoria or Yamagata lineage). Starting from the influenza season 2014–2015 new quadrivalent combination vaccines containing four different influenza strains are increasingly becoming available in the European Union/European Economic Area (EU/EEA). These vaccines contain two influenza A strains (A(H1N1) and A(H3N2) subtypes) and two influenza B strains (Victoria and Yamagata lineages) [8]. Annual vaccinations with the current antigen combination are recommended by WHO even when the antigen composition of the vaccine is unchanged compared with the previous season [6]. In Germany, the Standing Committee on Vaccination (STIKO) recommends annual vaccination in autumn as a standard vaccination for all persons aged 60 years and older, and where indicated in specific groups of persons e.g. children, adolescents and adults with an increased health risk resulting from an underlying disease, all pregnant women, persons at increased risk, e. g., medical personnel, persons in establishments dealing extensively with the public, as well as persons who may be possible sources of infection by caring for individuals at particular risk [9].


2 Currently available influenza medications

In Germany prescription medicines from two classes of active substances are approved for prevention and therapy of influenza infection (Table 1 [Tab. 1]). The M2 ion channel inhibitor amantadine belongs to the group of adamantanes and blocks the release of viral RNA into the cytoplasm of the host cell. This effect is achieved with therapeutic dosage of the active substance only with influenza A not with influenza B viruses because of different structure of the ion channel in both influenza species [10]. Due to the current resistance patterns of circulating viruses, the clinical use of adamantanes is not recommended presently (refer section 6). Thus, amantadine is currently not used for the treatment of influenza infections.

The compounds oseltamivir (Tamiflu™) and zanamivir (Relenza™) belong to the group of neuraminidase inhibitors and were approved and authorized by the European Medicines Agency (EMA) for prevention and treatment of influenza in the European Union/European Economic Area (EU/EEA) in 2002 and 1999 [11], [12]. They inhibit selectively the neuraminidase of influenza A and B viruses. Thus, the release of new viruses from infected cells is prevented.

Oseltamivir is admitted by EMA for adults and children including full-term newborns. It represents a prodrug and is available as oseltamivir phosphate in the form of capsules and as a powder for oral suspension. The compound is biotransformed by esterases in the intestinal tract and in the liver into the active metabolite oseltamivir carboxylate. The most common possible side effects include nausea, vomiting and headache. Antiviral therapy in neonates and young infants (<3 months) should be considered after careful risk assessment in severe clinical disease and should follow the recommendations of the German Society of Pediatric Infectious Diseases (DGPI) [11], [13].

Zanamivir showed unsatisfactory oral bioavailability and was therefore mixed with lactose and inhaled as a dry powder. Undesirable side effects are headache, diarrhea, nausea, and vomiting. Zanamivir is approved by EMA for treatment and prevention of influenza infections in patients from five years old. EMA issued in 2011 a summary on compassionate use for intravenous (i.v.) zanamivir in a specific targeted population and should be considered only to treat i.v. critically ill adults and children having a life-threatening condition [12], [14], [15].

In 2014 the neuraminidase inhibitors oseltamivir and zanamivir have been subject for critically discussion concerning their effectiveness and safety, as well as the appropriateness of stockpiling these drugs for use in future influenza pandemics [16], [17], [18], [19], [20]. An expert opinion from the European Center for Disease Prevention and Control (ECDC) and national authorities as well as national professional virological, medical and therapeutical associations (GfV e.V., DVV e.V, PEG e.V) confirmed earlier assessments that there is no significant new evidence from randomized control trials to support any changes to the approved indication and recommended use of neuraminidase inhibitors in EU/EEA Member States. This position was consistent with guidance from the World Health Organization (WHO) and other national public health organizations in the United States and Australia [21], [22], [23], [24].

Peramivir (USA: Rapivab™, Japan: Rapiacta™, South Korea: PeramiFlu™) is a cyclopentane neuraminidase inhibitor of influenza type A and type B viruses, which inhibits also various antiviral resistant influenza viruses. Due to its poor oral bioavailability peramivir was developed as intramuscular (i.m.) or intravenous (i.v.) applicable active substance. The most common adverse reaction is diarrhea [25], [26]. During the pandemic in 2009 peramivir was admitted in the USA based on an emergency use authorization (EUA) by the U. S. Food and Drug Administration (FDA) until June 23, 2010. On December 19, 2014 the FDA approved also peramivir for treatment of acute uncomplicated influenza in patients 18 years or older who have been symptomatic for more than two days. It is administered as a single i.v. dose [27]. In Japan and South Korea peramivir injection is approved since 2010 for the treatment of adults, children and infants with influenza [28].

Laninamivir octanoate (Japan: Inavir®) the octanoyl ester prodrug of laninamivir represents a zanamivir analogue. It is a long acting neuraminidase inhibitor (LANI) with therapeutic efficacy after one single nasal inhaled administration. Following inhalation laninamivir octanoate is absorbed by the epithelial cells lining the respiratory tract where it is rapidly hydrolyzed to the active laninamivir [29], [30], [31]. Laninamivir was approved for influenza treatment in 2010 and for prevention of influenza both in adults and children in 2013 in Japan [32].

T-705 (Favipiravir, Avigan™) represents a prodrug for oral administration. It selectively inhibits a broad-sprectrum of RNA-dependent RNA polymerases and thus inhibits the viral gene replication of influenza viruses and nine other RNA virus families [33], [34]. The active metabolite, T-705 ribofuranosyltriphosphate (T-705RTP) is generated intracellularly via phosphorylation by various cellular kinases [35]. The incorporation of T-705RTP into viral RNA during replication leads to a high mutation rate that generates a nonviable viral phenotype (lethal mutagenesis) [36]. Since March 2014 favipiravir is only approved for influenza pandemic preparedness in Japan [37].

All currently approved drugs for influenza prophylaxis and therapy (ion channel blocker, neuraminidase and polymerase inhibitors) are so-called direct acting drugs (DAD). In contrast, DAS181 (Fludase™), administered as an inhalable dry powder, is a broad spectrum host directed investigational influenza antiviral. It is being developed as a medication to prevent and treat infections due to common respiratory viruses like influenza, parainfluenza, and other viruses using cell surface sialic acids as receptors during attachment. Generated as a recombinant fusion protein composed of a sialidase catalytic domain derived from Actinomyces viscosus fused with a cell surface-anchoring sequence, it cleaves the linkages of sialic acid on host cells thereby removing the receptors for viral attachment [38]. This renders the cells inaccessible to infection by virus. It has also demonstrated activity in individual cases of parainfluenza in immunosuppressed patients [39]. The affect on viral load of influenza viruses and the safety and tolerability of the drug is currently being evaluated in phase 2 clinical trials in the USA [40].


3 Influenza antiviral resistance testing

Resistance to antiviral drugs is caused by subtype and inhibitor specific point mutations in the viral genes of the therapeutic target proteins. To date, no resistance was detected for the polymerase inhibitor T-705 until now. In contrast, ion channel blocker- and neuraminidase inhibitor-resistant influenza viruses with point mutations in gene segments coding for the M2 ion channel and neuraminidase, respectively, are known. These mutations can be detected by sequence-based technologies (classical sequencing, pyrosequencing) or, by the use of e.g. RT-PCR or melting point analysis of specific PCR amplification products (genotypic resistance analysis) [41], [42], [43], [44], [45]. These genotypic assays are advantageous as they can be performed with high sensitivity directly on the clinical specimen without the need for virus isolation. But, estimation of in vitro resistance to inhibitors on the basis of genotypic profiles is only reliable when the correlation between the presence of a specific point mutation and drug susceptibility of influenza viruses was proven. Such a mutation represents a molecular marker of resistance. In case of M2 ion channel blockers five molecular markers of resistance are known [46]. In contrast, in most cases estimation of in vitro resistance to neuraminidase inhibitors on the basis of genotypic profiles is unreliable, because currently insufficient amount of data are present to correlate between the presence of molecular marker of resistance and an actual in vitro resistance marked by an increase of the 50%-inhibitory concentration (IC50). Mutations causing the NA H275Y substitution (N1 numbering, subtypes A(H1N1)pdm09 and A(H5N1)) are the only ones consistently associated with increased levels of IC50-values to oseltamivir detected in NA inhibition assays (phenotypic resistance).

The genotypic resistance analysis is considered the gold standard for M2 ion channel blockers, but phenotypic resistance analysis represents the gold standard for NAI (Table 2 [Tab. 2]) [47]. It measures the inhibition of viral neuraminidase activity and can be carried out by colorimetric, fluorometric or luminometric neuraminidase inhibition assays [48], [49]. Cell-based assays measuring the inhibition of viral replication are not recommended for NA inhibitor susceptibility testing, because they have been shown to lack sensitivity and reliability, and can give falsely high or low IC50 values [47], [50], [51]. All phenotypic methods require cultured virus but allow the determination of 50% inhibitory concentration, i.e. that concentration of active compound required to inhibit enzyme activity by 50%. The WHO expert working group on surveillance of influenza antiviral susceptibility (WHO antiviral working group, AVWG) has established a set of criteria to define the antiviral susceptibility of viruses based on the fold-change of their IC50 value compared to reference IC50 values. For influenza A viruses, use of normal (<10-fold increase), reduced (10–100-fold increase) and highly reduced (>100-fold increase) inhibition, and for influenza B viruses the same criteria but using <5-fold, 5–50-fold and >50-fold increases compared to the median for viruses from the same type/subtype/lineage showing ‘normal inhibition’ (NI), is recommended. A reduced susceptibility to NA inhibitors measured in phenotypic assays should be verified by the detection of the associated molecular resistance marker [52]. Generally, the clinical relevance of the resistance detected using the NA inhibition assay in combination with genotypic analysis needs to be further evaluated. The IC50 values should not be used to draw a direct correlation with the drug concentrations needed to inhibit virus replication in the infected human host, as clinical data to support such inferences are inadequate. Although, assessment of influenza virus susceptibility to NA inhibitors provides a reliable and reasonably comprehensive approach to the identification of NA inhibitor-resistant isolates for surveillance purposes, there is a pressing need to establish a clinically relevant IC50 cutoff value which could be used to differentiate statistical outliers from truly resistant viruses [52], [53].


4 Quality assurances of testing and monitoring antiviral susceptibility of influenza viruses

The implementation of genotypic and/or phenotypic tests for antiviral susceptibility is complex and the laboratory expertise required for validation and troubleshooting should be considered. Especially for neuraminidase inhibition assays, although some are commercially available (NA-Fluor, NA-Star, NA-XTD; Life Technologies), additional training is likely to be necessary [47], [49]. The use of genotypic methods also requires a good working knowledge of the encoded NA amino acid substitutions, known to confer reduced susceptibility to antivirals in specific influenza virus types and subtypes [47]. The standard operation protocols for in house NA inhibition assays provided from WHO addressed mainly the National Influenza Centres (NICs) but are also available for clinical laboratories interested in influenza antiviral resistance testing [54]. However, there is a strongly recommendation, that individual results of antiviral susceptibility testing in clinical laboratories should be collected nationally at the NIC [55].

The external quality assessment (EQA) exercise held for European influenza reference laboratories in 2011 has identified the need for harmonization antiviral susceptibility testing and data interpretation. Although interpretation of the data generally matched the consensus, only 85% of participants identified oseltamivir reduced susceptibility/resistance in a mixture of A(H1N1)pdm09 oseltamivir-sensitive/-resistant viruses and 23% considered oseltamivir-sensitive influenza B virus reduced susceptible/resistant [56]. The external quality assessment for influenza antiviral susceptibility for the European Reference Laboratory Network for Human Influenza (ERLI-Net) held two years later in 2013 showed that compared to 2010 the score improved, although in total only 85% of the oseltamivir results and for zanamivir 73% of results were correct [57].

When deciding whether implementation of antiviral susceptibility testing within a laboratory is practical together with the long-term costs and the requirements for testing, a training of laboratory staff in implementation and validation, troubleshooting, data analysis and interpretation should be considered. This is often best delivered through one-to-one training by a partner laboratory (referred to as “twinning”) [47], [56].


5 Monitoring of antiviral susceptibility of influenza viruses in Germany

Worldwide, except Europe, antiviral surveillance data were mainly reported via the WHO Collaboration Centres (WHOCC). In Europe antiviral resistance is monitored by the European Centre for Disease Prevention and Control (ECDC) and the WHO/Europe Influenza Surveillance (EuroFlu) based on the reports sent by influenza reference laboratories (NIC) to The ECDC Surveillance System (TESSy). In Germany, circulating influenza viruses were tested in the national influenza centre (NRZ Influenza; http://www.rki.de/DE/Content/Infekt/NRZ/Influenza/arbeitsbereiche/abeitsbereiche_node.html) for their antiviral susceptibility on a large scale by using genotypic and phenotypic methods, thus emergence and spread of resistant influenza viruses were monitored. The data were reported weekly to TESSy and additionally published via the weekly and seasonally reports of the Working Group Influenza (AGI; https://www.influenza.rki.de). The examination of a representative number of influenza A and influenza B viruses circulating in Germany between October 1998 and May 2016 yielded a resistance situation comparable with other countries worldwide.


6 Resistance of influenza viruses to adamantane derivates

Resistance to the adamantane derivates amantadine and rimantadine can emerge rapidly during treatment. A prevalence of <1% was observed in circulating viruses until 2004. During the influenza season 2004–2005 a high prevalence and a strong spreading of community A(H3N2) isolates resistant to adamantanes were detected in Asia, Europe, Australia and the USA [58]. In the following winter 2005–2006 the prevalence of these viruses increased to nearly 100% [59]. These findings and the first reports on the detection of resistant seasonal A(H1N1) viruses rendered this class of drugs mostly ineffective [60]. In response, the CDC recommended against the use of adamantane derivates for treatment and prevention of influenza infections [61]. In Germany, the prevalence of primary resistance of A(H3N2) viruses to adamantane derivates was 12% during the 2004–2005 season. Resistance increased to 81% during the following winter 2005–2006. Very high frequency of resistant A(H3N2) viruses continued to be detected, with 100% in the 2007–2008 influenza season [62]. The resistance-associated S31N substitution (serine-to-asparagine) of the M2 ion channel protein (AGT→AAT) was the most commonly observed mutation [63]. In Germany, all tested A(H1N1) viruses remained sensitive to amantadin and rimantadin [62]. In the northern hemisphere increased prevalence of resistance among A(H1N1) viruses also has been observed, although the overall proportion of resistant A(H1N1) viruses was not as high as that for A(H3N2) viruses until 2009 [59]. The appearance of influenza viruses of subtype A(H1N1)pdm09 led to a pandemic in 2009, followed by complete A(H1N1) subtype replacement. Due to reassortment (antigenic shift) the A(H1N1)pdm09 viruses carry the M2 ion channel protein originated from the Eurasian avian-like swine lineage [64]. Viruses that are associated with this lineage possess the M2-S31N substitution as a polymorphism, and show thus “natural” resistance to adamantane derivates [65], [66].


7 Resistance of influenza viruses to neuraminidase inhibitors

Resistance to the neuraminidase inhibitors oseltamivir and zanamivir following EMA and FDA approval occurred only sporadically and mostly on clinical trials in immunocompromised patients. The incidence of oseltamivir resistant influenza A(H3N2) and A(H1N1) viruses seen in clinical trial samples of immunocompetent patients until July 2004 was 0.33% (4/1228) in adults (≥13 years) and 4% (17/421) in children (≤12 years), resulting in an overall incidence of 1.26% [67], [68]. An high degree of A(H1N1) and A(H3N2) oseltamivir-resistant viruses has been found within two clinical trials [67], [69]. During oseltamivir treatment about 16.3% (7/43) resistant A(H1N1) and 18% (9/59) resistant A(H3N2) viruses occurred, which was may be connected to an insufficient dosage regime.

In contrast to these clinical findings, the occurrence and spread of neuraminidase inhibitor-resistant strains within seasonal influenza viruses was a rare event until winter 2007. In comparison to sensitive viruses the resistant viruses showed a diminished viral fitness, marked by worse replication rates, and lower transmissibility, leading to the conclusion that resistance-associated mutations are unlikely to be of clinical consequence [46], [70], [71]. Therefore, the detection and rapid spread of oseltamivir-resistant A/Brisbane/59/2007-like A(H1N1) viruses in November 2007 within the seasonally circulating influenza viruses was unpredicted and unexpected [72]. All resistant viruses carried the H275Y (histidine-to-tyrosine) substitution in their neuraminidase and showed highly reduced susceptibility to oseltamivir. During the following season 2007–2008 these viruses spread rapidly over the northern hemisphere. The resistance to oseltamivir could not be explained with reassortment and was not the consequence of widespread therapeutically usage of oseltamivir [73], [74]. The infected patients were not linked epidemiologically and showed symptoms that were similar to that of infections with sensitive viruses [75], [76]. The oseltamivir-resistant viruses were shed by adults up to eight days and were transmissible by household contacts without any change in their resistance profile [76]. They did not show obvious attenuation relative to earlier viruses carrying the H275Y substitution [77]. During the summer 2008 the prevalence of oseltamivir-resistant A(H1N1) viruses increased up to 80% within the southern hemisphere. In the following season 2008–2009 almost all circulating A(H1N1) viruses were oseltamivir-resistant [78], [79]. Bloom et al. showed that two permissive secondary mutations (V234M, R222Q) occurred in seasonal A(H1N1) shortly before the widespread appearance of H275Y. These mutations restore the detrimental influence of the NA-H275Y mutation and thus enable the evolution of influenza oseltamivir resistance [80].

In 2009, a triple reassortant swine virus reassorted with an Eurasian avian-like swine virus resulting in the A(H1N1)pdm09 virus, that has spread internationally with unprecedented speed and caused the first pandemic of the 21st century [64], [78], [81]. The A(H1N1)pdm09 neuraminidase originated from the Eurasian avian-like swine A(H1N1) virus and is characterized by neuraminidase inhibitor sensitivity [64]. The former oseltamivir-resistant seasonal A(H1N1) virus become to extinct soon after the emergence of the A(H1N1)pdm09 virus [78].

In Germany, overall 3,720 influenza viruses (66% A(H1N1)pdm09, 19% A(H3N2) and 15% influenza B) circulating between April 2009 and May 2016 were analyzed phenotypically and/or genotypically [82]. Only twelve out of 2,430 A(H1N1)pdm09 viruses showed highly reduced susceptibility to the neuraminidase inhibitor oseltamivir indicated by an up to 1,000-fold increase of the IC50 for oseltamivir in phenotypic assays compared to the sensitive wild-type virus. All these resistant viruses carried the substitution H275Y in their neuraminidase and remained sensitive to zanamivir. Almost all viruses were collected from patients during or after oseltamivir treatment. Only, two pandemic viruses showed resistance to oseltamivir despite the lack of any evidence that they arose during antiviral treatment. In the first case, the virus was isolated from a respiratory sample of a 21-year-old man who developed an influenza-like illness while staying abroad for vacation. The second case was a 9-year-old otherwise healthy boy. The oseltamivir-resistance was confirmed by phenotypic analysis of the virus isolates. Pyrosequencing analysis performed on the patients’ respiratory specimens showed the molecular resistance marker NA-H275Y. In both cases the viruses remained sensitive to zanamivir in phenotypic assays with IC50<2nM and further molecular markers of resistance in viral NAs were not detected [42], [62], [83]. A(H3N2) viruses with altered susceptibility to neuraminidase inhibitors were detected in two cases. In 2012 treatment-selected A(H3N2) viruses showing reduced susceptibility to neuraminidase inhibitors due to the NA-R292K substitution were isolated from two children at day five of oseltamivir treatment [84]. Influenza B viruses showing reduced susceptibility to neuraminidase inhibitors were not detected.

Globally, about 99% of the tested influenza viruses were susceptible against the four neuraminidase inhibitors. The prevalence of viruses with reduced or highly reduced sensitivity to neuraminidase inhibitors was highest with 1.9% in the 2013–2014 influenza season, compared to the 2012–2013 and 2014–2015 seasons where the frequencies were 0.6% and 0.5%, respectively [85]. The increase of resistant viruses in 2013–2014 is associated with the detection of several clusters of A(H1N1)pdm09 viruses carrying the H275Y substitution in China, Japan and the United States [86]. In A(H3N2) and influenza B viruses, mutations, known to confer reduced susceptibility to neuraminidase inhibitors or phenotypically reduced sensitivity are being detected at a substantially lower frequency than in A(H1N1)pdm09 viruses [78].


8 Conclusions

Influenza virus infection is a serious and frequently underestimated disease. Due to the development of adamantane resistance in A(H3N2) and the emergence of adamantane-resistant A(H1N1)pdm09 viruses in 2009, the neuraminidase inhibitors zanamivir and oseltamivir are the only effective antiviral drugs, available commercially in Europe. They act against all of the currently circulating human influenza viruses. Although reduced susceptibility to these antivirals remains rare, the appearance and spread of resistant viruses in the past underlines the urgent need to develop and/or approve new active compounds and to closely monitor the resistance profiles of circulating influenza viruses in the international context.


Notes

Acknowledgement

This review was supported by the Scientific Advisory Board for Antiviral Therapy of the German Association for the Control of Virus Diseases (DVV) and the Society of Virology (GfV) as well as the Section for Antiviral Therapy of the Paul-Ehrlich-Gesellschaft (PEG). The author's grateful thanks are given to PD Dr. Michaela Schmidtke for critically reviewing and discussing the manuscript.

Competing interests

The author declares that she has no competing interests.


References

1.
Fauquet CM, Mayo MA, Maniloff J, Desselberger U, Ball LA, editors. Virus taxonomy. Classification and nomenclature of viruses. Eighth report of the International Committee on Taxonomy of Viruses. Amsterdam, Boston: Elsevier Academic Press; 2005.
2.
Kawaoka Y, Neumann G. Influenza Viruses: An Introduction. In: Kawaoka Y, Neumann G, editors. Influenza Virus: Methods and Protocols. New York et al.: Humana, Springer; 2012. p. 1-9. (Methods in Molecular Biology; 865). DOI: 10.1007/978-1-61779-621-0_1 Externer Link
3.
Shi Y, Wu Y, Zhang W, Qi J, Gao GF. Enabling the 'host jump': structural determinants of receptor-binding specificity in influenza A viruses. Nat Rev Microbiol. 2014 Dec;12(12):822-31. DOI: 10.1038/nrmicro3362 Externer Link
4.
Arvia R, Corcioli F, Pierucci F, Azzi A. Molecular markers of influenza B lineages and clades. Viruses. 2014 Nov;6(11):4437-46. DOI: 10.3390/v6114437 Externer Link
5.
Earn DJD, Dushoff J, Levin SA. Ecology and evolution of the flu. Trends Ecol Evol. 2002 Jul;17(7):334-40. DOI: 10.1016/S0169-5347(02)02502-8 Externer Link
6.
World Health Organization (WHO). Vaccines against influenza. WHO position paper – November 2012. Wkly Epidemiol Rec. 2012 Nov;87(47):461-76. Available from: http://www.who.int/wer/2012/wer8747 Externer Link
7.
Robert Koch Institut (RKI). Influenza (Teil 1): Erkrankungen durch saisonale Influenzaviren. [updated 2016 Feb 12; cited 2016 Jul 26]. (RKI-Ratgeber für Ärzte). Available from: http://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Influenza_saisonal.html Externer Link
8.
European Centre for Disease Prevention and Control (ECDC). Influenza vaccination. [cited 2016 Aug 03]. Available from: http://ecdc.europa.eu/en/healthtopics/seasonal_influenza/vaccines/Pages/influenza_vaccination.aspx Externer Link
9.
Robert Koch Institut (RKI) / German Standing Committee on Vaccination. Recommendations of the Standing Committee on Vaccination (STIKO) at the Robert Koch Institute/Effective: August 2015. Epidemiol Bull. 2015 Aug 24;(34):327-62. DOI: 10.17886/EpiBull-2015-001.3 Externer Link
10.
Wang J, Pielak RM, McClintock MA, Chou JJ. Solution structure and functional analysis of the influenza B proton channel. Nat Struct Mol Biol. 2009 Dec;16(12):1267-71. DOI: 10.1038/nsmb.1707 Externer Link
11.
European Medicines Agency (EMA). Tamiflu oseltamivir. European public assessment report (EPAR) – Summary. [updated 2015 Jul 31; cited 2016 Jul 28]. Available from: http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000402/human_med_001075.jsp&mid=WC0b01ac058001d124 Externer Link
12.
European Medicines Agency (EMA). Summary on compassionate use for iv zanamivir. EMA/484396/2011 Rev. 1. London: European Medicines Agency; 2011 [cited 2016 Jul 28]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Other/2011/03/WC500104028.pdf Externer Link
13.
Berner R; Deutsche Gesellschaft für pädiatrische Infektiologie (DGPI). Aktualisierte Empfehlung der DGPI zur Diagnostik, Therapie und Prophylaxe der Infektion mit dem Neuen Influenza A/H1N1v-Virus bei Kindern und Jugendlichen. Stand 09.11.2009. Deutsche Gesellschaft für pädiatrische Infektiologie; 2009 [cited 2016 Jul 28]. Available from: http://dgpi.de/go/wp-content/uploads/2012/12/InflA-_H1N1v_DGPI-Empfehlungen_09Nov2009.pdf Externer Link
14.
European Medicines Agency (EMA). Conditions of use, conditions for distribution and patients targeted and conditions for safety monitoring addressed to member states for IV Zanamivir available for compassionate use. EMA/671175/2015 Rev. 3. London: European Medicines Agency; 2015 [cited 2016 Jul 28]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Other/2010/02/WC500074124.pdf Externer Link
15.
European Medicines Agency (EMA). European Medicines Agency decision P/0094/2015 of 8 May 2015 on the acceptance of a modification of an agreed paediatric investigation plan for zanamivir (Relenza), (EMEA-001318-PIP01-12-M01). London: European Medicines Agency; 2015 [cited 2016 Jul 28]. Available from: http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/pips/EMEA-001318-PIP01-12-M01/pip_001026.jsp&mid=WC0b01ac058001d129 Externer Link
16.
Jefferson T, Jones M, Doshi P, Del Mar C. Neuraminidase inhibitors for preventing and treating influenza in healthy adults: systematic review and meta-analysis. BMJ. 2009 Dec;339:b5106. DOI: 10.1136/bmj.b5106 Externer Link
17.
Muthuri SG, Myles PR, Venkatesan S, Leonardi-Bee J, Nguyen-Van-Tam JS. Impact of neuraminidase inhibitor treatment on outcomes of public health importance during the 2009-2010 influenza A(H1N1) pandemic: a systematic review and meta-analysis in hospitalized patients. J Infect Dis. 2013 Feb;207(4):553-63. DOI: 10.1093/infdis/jis726 Externer Link
18.
Heneghan CJ, Onakpoya I, Jones MA, Doshi P, Del Mar CB, Hama R, Thompson MJ, Spencer EA, Mahtani KR, Nunan D, Howick J, Jefferson T. Neuraminidase inhibitors for influenza: a systematic review and meta-analysis of regulatory and mortality data. Health Technol Assess. 2016 May;20(42):1-242. DOI: 10.3310/hta20420 Externer Link
19.
Heneghan CJ, Onakpoya I, Thompson M, Spencer EA, Jones M, Jefferson T. Zanamivir for influenza in adults and children: systematic review of clinical study reports and summary of regulatory comments. BMJ. 2014 Apr;348:g2547. DOI: 10.1136/bmj.g2547 Externer Link
20.
Hsu J, Santesso N, Mustafa R, Brozek J, Chen YL, Hopkins JP, Cheung A, Hovhannisyan G, Ivanova L, Flottorp SA, Saeterdal I, Wong AD, Tian J, Uyeki TM, Akl EA, Alonso-Coello P, Smaill F, Schünemann HJ. Antivirals for treatment of influenza: a systematic review and meta-analysis of observational studies. Ann Intern Med. 2012 Apr;156(7):512-24. DOI: 10.7326/0003-4819-156-7-201204030-00411 Externer Link
21.
Schmidtke M, Mertens T; Gesellschaft für Virologie (GfV); Deutsche Vereinigung zur Bekämpfung der Viruskrankheiten (DVV); Paul-Ehrlich-Gesellschaft für Chemotherapie (PEG). Brauchen wir die Grippemittel Tamiflu und Relenza? GMS Infect Dis. 2014 Aug 25;2:Doc06. DOI: 10.3205/id000014 Externer Link
22.
European Center for Disease Prevention and Control (ECDC). ECDC preliminary scientific advice. Expert Opinion on neuraminidase inhibitors for prevention and treatment of influenza. Review of recent systematic reviews and meta-analyses. European Center for Disease Prevention and Control; 2016 [cited 2016 July 28]. Available from: http://ecdc.europa.eu/en/publications/Publications/neuraminidase-inhibitors-flu-consultation.pdf Externer Link
23.
Bundesregierung. Antwort auf die Kleine Anfrage der Abgeordneten Kordula Schulz-Asche, Dr. Harald Terpe, Maria Klein-Schmeink, weiterer Abgeordneter und der Fraktion BÜNDNIS 90/DIE GRÜNEN – Drucksache 18/1227 – Zweifel an der Wirksamkeit der Neuraminidasehemmer Tamiflu und Relenza. Drucksache 18/1384. Bundestag; 2014 [cited 2016 Jul 28]. Available from: http://dipbt.bundestag.de/dip21/btd/18/013/1801384.pdf Externer Link
24.
Hurt AC, Kelly H. Debate Regarding Oseltamivir Use for Seasonal and Pandemic Influenza. Emerging Infect Dis. 2016 Jun;22(6):949-55. DOI: 10.3201/eid2206.151037 Externer Link
25.
BioCryst. Ravipab: Peramivir injection. [cited 2016 August 03]. Available from: http://www.rapivab.com Externer Link
26.
Alame MM, Massaad E, Zaraket H. Peramivir: A Novel Intravenous Neuraminidase Inhibitor for Treatment of Acute Influenza Infections. Front Microbiol. 2016;7:450. DOI: 10.3389/fmicb.2016.00450 Externer Link
27.
U.S. Food and Drug Administration (FDA). FDA approves Rapivab to treat flu infection. FDA News Release. 2014 Dec 22 [cited 2016 Aug 03, archived 2016 Oct 22]. Available from: https://wayback.archive-it.org/7993/20161022101232/http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm427755.htm Externer Link
28.
Hata A, Akashi-Ueda R, Takamatsu K, Matsumura T. Safety and efficacy of peramivir for influenza treatment. Drug Des Devel Ther. 2014;8:2017-38. DOI: 10.2147/DDDT.S46654 Externer Link
29.
Watanabe A, Chang SC, Kim MJ, Chu DW, Ohashi Y; MARVEL Study Group. Long-acting neuraminidase inhibitor laninamivir octanoate versus oseltamivir for treatment of influenza: A double-blind, randomized, noninferiority clinical trial. Clin Infect Dis. 2010 Nov;51(10):1167-75. DOI: 10.1086/656802 Externer Link
30.
Sugaya N, Ohashi Y. Long-acting neuraminidase inhibitor laninamivir octanoate (CS-8958) versus oseltamivir as treatment for children with influenza virus infection. Antimicrob Agents Chemother. 2010 Jun;54(6):2575-82. DOI: 10.1128/AAC.01755-09 Externer Link
31.
Ison MG. Antivirals and resistance: influenza virus. Curr Opin Virol. 2011 Dec;1(6):563-73. DOI: 10.1016/j.coviro.2011.09.002 Externer Link
32.
Daiichy-Sankyo. Daiichi Sankyo Receives Approval for the Use of Inavir® to Prevent Influenza. Press Release. 2013 Dec 20. [cited 2016 Aug 03]. Available from: http://www.daiichisankyo.com/media_investors/media_relations/press_releases/detail/006052.html Externer Link
33.
Furuta Y, Gowen BB, Takahashi K, Shiraki K, Smee DF, Barnard DL. Favipiravir (T-705), a novel viral RNA polymerase inhibitor. Antiviral Res. 2013 Nov;100(2):446-54. DOI: 10.1016/j.antiviral.2013.09.015 Externer Link
34.
MediVector I. Favipiravir. [cited 2016 August 03]. Available from: http://www.medivector.com/case-studies/anti-viral-case-study/favipiravir-one-drug-many-bugs/ Externer Link
35.
Furuta Y, Takahashi K, Kuno-Maekawa M, Sangawa H, Uehara S, Kozaki K, Nomura N, Egawa H, Shiraki K. Mechanism of action of T-705 against influenza virus. Antimicrob Agents Chemother. 2005 Mar;49(3):981-6. DOI: 10.1128/AAC.49.3.981-986.2005 Externer Link
36.
Baranovich T, Wong SS, Armstrong J, Marjuki H, Webby RJ, Webster RG, Govorkova EA. T-705 (favipiravir) induces lethal mutagenesis in influenza A H1N1 viruses in vitro. J Virol. 2013 Apr;87(7):3741-51. DOI: 10.1128/JVI.02346-12 Externer Link
37.
Toyama Chemical. The New Drug Application Approval of “AVIGAN® Tablet 200mg” in Japan for the anti-influenza virus drug. News Release. 2014 Mar 24. [cited 2016 Aug 03]. Available from: http://www.toyama-chemical.co.jp/eng/news/news140324e.html Externer Link
38.
Malakhov MP, Aschenbrenner LM, Smee DF, Wandersee MK, Sidwell RW, Gubareva LV, Mishin VP, Hayden FG, Kim DH, Ing A, Campbell ER, Yu M, Fang F. Sialidase fusion protein as a novel broad-spectrum inhibitor of influenza virus infection. Antimicrob Agents Chemother. 2006 Apr;50(4):1470-9. DOI: 10.1128/AAC.50.4.1470-1479.2006 Externer Link
39.
Moss RB, Hansen C, Sanders RL, Hawley S, Li T, Steigbigel RT. A phase II study of DAS181, a novel host directed antiviral for the treatment of influenza infection. J Infect Dis. 2012 Dec;206(12):1844-51. DOI: 10.1093/infdis/jis622 Externer Link
40.
Ansun Biopharma. Phase 2B Study on Safety and Therapeutic Efficacy of DAS181 in Adult Subjects With Naturally Acquired Influenza. In: ClinicalTrials.gov. 2012 [updated 2013 Jul 11; cited 2016 Aug 03]. Available from: https://clinicaltrials.gov/ct2/show/record/NCT01740063 Externer Link
41.
Duwe S, Schweiger B. A new and rapid genotypic assay for the detection of neuraminidase inhibitor resistant influenza A viruses of subtype H1N1, H3N2, and H5N1. J Virol Methods. 2008 Nov;153(2):134-41. DOI: 10.1016/j.jviromet.2008.07.017 Externer Link
42.
Duwe SC, Wedde M, Birkner P, Schweiger B. Genotypic and phenotypic resistance of pandemic A/H1N1 influenza viruses circulating in Germany. Antiviral Res. 2011 Jan;89(1):115-8. DOI: 10.1016/j.antiviral.2010.11.001 Externer Link
43.
Redlberger-Fritz M, Aberle SW, Strassl R, Popow-Kraupp T. Rapid identification of neuraminidase inhibitor resistance mutations in seasonal influenza virus A(H1N1), A(H1N1)2009, and A(H3N2) subtypes by melting point analysis. Eur J Clin Microbiol Infect Dis. 2012 Jul;31(7):1593-601. DOI: 10.1007/s10096-011-1482-9 Externer Link
44.
Deyde VM, Sheu TG, Trujillo AA, Okomo-Adhiambo M, Garten R, Klimov AI, Gubareva LV. Detection of molecular markers of drug resistance in 2009 pandemic influenza A (H1N1) viruses by pyrosequencing. Antimicrob Agents Chemother. 2010 Mar;54(3):1102-10. DOI: 10.1128/AAC.01417-09 Externer Link
45.
Lackenby A, Democratis J, Siqueira MM, Zambon MC. Rapid quantitation of neuraminidase inhibitor drug resistance in influenza virus quasispecies. Antivir Ther (Lond). 2008;13(6):809-20.
46.
Abed Y, Goyette N, Boivin G. Generation and characterization of recombinant influenza A (H1N1) viruses harboring amantadine resistance mutations. Antimicrob Agents Chemother. 2005 Feb;49(2):556-9. DOI: 10.1128/AAC.49.2.556-559.2005 Externer Link
47.
World Health Organization (WHO), editor. Practical guidance for national influenza centres establishing or implementing neuraminidase inhibitor susceptibility surveillance. Geneva: WHO; 2016. Available from: http://www.who.int/influenza/gisrs_laboratory/antiviral_susceptibility/practical_guidance/en/ Externer Link
48.
Nguyen HT, Sheu TG, Mishin VP, Klimov AI, Gubareva LV. Assessment of pandemic and seasonal influenza A (H1N1) virus susceptibility to neuraminidase inhibitors in three enzyme activity inhibition assays. Antimicrob Agents Chemother. 2010 Sep;54(9):3671-7. DOI: 10.1128/AAC.00581-10 Externer Link
49.
Murtaugh W, Mahaman L, Healey B, Peters H, Anderson B, Tran M, Ziese M, Carlos MP. Evaluation of three influenza neuraminidase inhibition assays for use in a public health laboratory setting during the 2011-2012 influenza season. Public Health Rep. 2013 Sep-Oct;128 Suppl 2:75-87.
50.
Gubareva LV, Matrosovich MN, Brenner MK, Bethell RC, Webster RG. Evidence for zanamivir resistance in an immunocompromised child infected with influenza B virus. J Infect Dis. 1998 Nov;178(5):1257-62. DOI: 10.1086/314440 Externer Link
51.
Tisdale M. Monitoring of viral susceptibility: new challenges with the development of influenza NA inhibitors. Rev Med Virol. 2000 Jan-Feb;10(1):45-55. DOI: 10.1002/(SICI)1099-1654(200001/02)10:1<45::AID-RMV265>3.0.CO;2-R Externer Link
52.
Meetings of the WHO working group on surveillance of influenza antiviral susceptibility – Geneva, November 2011 and June 2012. Wkly Epidemiol Rec. 2012 Sep 28;87(39):369-74. Available from: http://www.who.int/wer/2012/wer8739.pdf Externer Link
53.
Okomo-Adhiambo M, Sleeman K, Ballenger K, Nguyen HT, Mishin VP, Sheu TG, Smagala J, Li Y, Klimov AI, Gubareva LV. Neuraminidase inhibitor susceptibility testing in human influenza viruses: a laboratory surveillance perspective. Viruses. 2010 Oct;2(10):2269-89. DOI: 10.3390/v2102269 Externer Link
54.
WHO Global Influenza Surveillance Network. Manual for the laboratory diagnosis and virological surveillance of influenza. Geneva: WHO; 2011. Available from: http://www.who.int/influenza/gisrs_laboratory/manual_diagnosis_surveillance_influenza Externer Link
55.
Pozo F, Lina B, Andrade HR, Enouf V, Kossyvakis A, Broberg E, Daniels R, Lackenby A, Meijer A; Community Network of Reference Laboratories for Human Influenza in Europe. Guidance for clinical and public health laboratories testing for influenza virus antiviral drug susceptibility in Europe. J Clin Virol. 2013 05;57(1):5-12. DOI: 10.1016/j.jcv.2013.01.009 Externer Link
56.
Thompson CI, Lackenby A, Daniels RS, McCauley JW, Pereyaslov D, Broberg EK, Meijer A, Zambon MC. Evaluation of influenza virus antiviral susceptibility testing in Europe: results from the first external quality assessment exercise. J Clin Virol. 2013 Mar;56(3):212-8. DOI: 10.1016/j.jcv.2012.11.005 Externer Link
57.
European Centre for Disease Prevention and Control (ECDC). External quality assessment for influenza antiviral susceptibility for the European Reference Laboratory Network for human influenza, 2013. Stockholm: ECDC; 2014. DOI: 10.2900/2979 Externer Link
58.
Bright RA, Medina MJ, Xu X, Perez-Oronoz G, Wallis TR, Davis XM, Povinelli L, Cox NJ, Klimov AI. Incidence of adamantane resistance among influenza A (H3N2) viruses isolated worldwide from 1994 to 2005: a cause for concern. Lancet. 2005 Oct 1;366(9492):1175-81. DOI: 10.1016/S0140-6736(05)67338-2 Externer Link
59.
Hayden F. Developing new antiviral agents for influenza treatment: what does the future hold? Clin Infect Dis. 2009 Jan;48 Suppl 1:S3-13. DOI: 10.1086/591851 Externer Link
60.
Deyde VM, Xu X, Bright RA, Shaw M, Smith CB, Zhang Y, Shu Y, Gubareva LV, Cox NJ, Klimov AI. Surveillance of resistance to adamantanes among influenza A(H3N2) and A(H1N1) viruses isolated worldwide. J Infect Dis. 2007 Jul 15;196(2):249-57. DOI: 10.1086/518936 Externer Link
61.
Centers for Disease Control and Prevention (CDC). CDC Recommends against the Use of Amantadine and Rimantadine for the Treatment or Prophylaxis of Influenza in the United States during the 2005–06 Influenza Season. CDC health alert. 2006 Jan 14 [cited 2016 Aug 04]. Available from: https://web.archive.org/web/20080503092840/http://www.cdc.gov/flu/han011406.htm Externer Link
62.
Duwe S, Schweiger B. Resistenzen bei Influenzaviren. In: Brem G, editor. Zoonotische Influenzaviren. Erreger zwischen Banalität und globaler Bedrohung. Symposium der Österreichischen Akademie der Wissenschaften (ÖAW) am 29. und 30. März 2012 in Wien. Wien: Verlag der Österreichischen Akademie der Wissenschaften; 2013. p. 121. (Academia Scientiarium Austriaca / Classis mathematica et historico-naturalis, Acta et Commentationes; 2).
63.
Belshe RB, Smith MH, Hall CB, Betts R, Hay AJ. Genetic basis of resistance to rimantadine emerging during treatment of influenza virus infection. J Virol. 1988 May;62(5):1508-12.
64.
Neumann G, Noda T, Kawaoka Y. Emergence and pandemic potential of swine-origin H1N1 influenza virus. Nature. 2009 Jun;459(7249):931-9. DOI: 10.1038/nature08157 Externer Link
65.
Schmidtke M, Zell R, Bauer K, Krumbholz A, Schrader C, Suess J, Wutzler P. Amantadine resistance among porcine H1N1, H1N2, and H3N2 influenza A viruses isolated in Germany between 1981 and 2001. Intervirology. 2006;49(5):286-93. DOI: 10.1159/000094244 Externer Link
66.
Krumbholz A, Schmidtke M, Bergmann S, Motzke S, Bauer K, Stech J, Dürrwald R, Wutzler P, Zell R. High prevalence of amantadine resistance among circulating European porcine influenza A viruses. J Gen Virol. 2009 Apr;90(Pt 4):900-8. DOI: 10.1099/vir.2008.007260-0 Externer Link
67.
Ward P, Small I, Smith J, Suter P, Dutkowski R. Oseltamivir (Tamiflu) and its potential for use in the event of an influenza pandemic. J Antimicrob Chemother. 2005 Feb;55 Suppl 1:i5-i21. DOI: 10.1093/jac/dki018 Externer Link
68.
Stephenson I, Democratis J, Lackenby A, McNally T, Smith J, Pareek M, Ellis J, Bermingham A, Nicholson K, Zambon M. Neuraminidase inhibitor resistance after oseltamivir treatment of acute influenza A and B in children. Clin Infect Dis. 2009 Feb;48(4):389-96. DOI: 10.1086/596311 Externer Link
69.
Kiso M, Mitamura K, Sakai-Tagawa Y, Shiraishi K, Kawakami C, Kimura K, Hayden FG, Sugaya N, Kawaoka Y. Resistant influenza A viruses in children treated with oseltamivir: descriptive study. Lancet. 2004;364(9436):759-65. DOI: 10.1016/S0140-6736(04)16934-1 Externer Link
70.
Yen HL, Herlocher LM, Hoffmann E, Matrosovich MN, Monto AS, Webster RG, Govorkova EA. Neuraminidase inhibitor-resistant influenza viruses may differ substantially in fitness and transmissibility. Antimicrob Agents Chemother. 2005 Oct;49(10):4075-84. DOI: 10.1128/AAC.49.10.4075-4084.2005 Externer Link
71.
Ives JA, Carr JA, Mendel DB, Tai CY, Lambkin R, Kelly L, Oxford JS, Hayden FG, Roberts NA. The H274Y mutation in the influenza A/H1N1 neuraminidase active site following oseltamivir phosphate treatment leave virus severely compromised both in vitro and in vivo. Antiviral Res. 2002 Aug;55(2):307-17. DOI: 10.1016/S0166-3542(02)00053-0 Externer Link
72.
Hauge SH, Dudman S, Borgen K, Lackenby A, Hungnes O. Oseltamivir-resistant influenza viruses A (H1N1), Norway, 2007-08. Emerging Infect Dis. 2009 Feb;15(2):155-62. DOI: 10.3201/eid1502.081031 Externer Link
73.
Meijer A, Lackenby A, Hungnes O, Lina B, van-der-Werf S, Schweiger B, Opp M, Paget J, van-de-Kassteele J, Hay A, Zambon M; European Influenza Surveillance Scheme. Oseltamivir-resistant influenza virus A (H1N1), Europe, 2007-08 season. Emerging Infect Dis. 2009 Apr;15(4):552-60. DOI: 10.3201/eid1504.181280 Externer Link
74.
Hurt AC, Ernest J, Deng YM, Iannello P, Besselaar TG, Birch C, Buchy P, Chittaganpitch M, Chiu SC, Dwyer D, Guigon A, Harrower B, Kei IP, Kok T, Lin C, McPhie K, Mohd A, Olveda R, Panayotou T, Rawlinson W, Scott L, Smith D, D’Souza H, Komadina N, Shaw R, Kelso A, Barr IG. Emergence and spread of oseltamivir-resistant A(H1N1) influenza viruses in Oceania, South East Asia and South Africa. Antiviral Res. 2009 Jul;83(1):90-3. DOI: 10.1016/j.antiviral.2009.03.003 Externer Link
75.
Buchholz U, Brockmann S, Duwe S, Schweiger B, an der Heiden M, Reinhardt B, Buda S. Household transmissibility and other characteristics of seasonal oseltamivir-resistant influenza A(H1N1) viruses, Germany, 2007-8. Euro Surveill. 2010 Feb;15(6). pii: 19483. Available from: http://www.eurosurveillance.org/ViewArticle.aspx?ArticleId=19483 Externer Link
76.
Duwe S, Heider A, Braun C, Schweiger B, Buchholz U. Person-to-person transmission of oseltamivir-resistant influenza A/H1N1 viruses in two households; Germany 2007/08. J Clin Virol. 2009 Nov;46(3):295-7. DOI: 10.1016/j.jcv.2009.07.022 Externer Link
77.
Rameix-Welti MA, Enouf V, Cuvelier F, Jeannin P, van der Werf S. Enzymatic properties of the neuraminidase of seasonal H1N1 influenza viruses provide insights for the emergence of natural resistance to oseltamivir. PLoS Pathog. 2008 Jul;4(7):e1000103. DOI: 10.1371/journal.ppat.1000103 Externer Link
78.
Hurt AC. The epidemiology and spread of drug resistant human influenza viruses. Curr Opin Virol. 2014 Oct;8:22-9. DOI: 10.1016/j.coviro.2014.04.009 Externer Link
79.
Moscona A. Global transmission of oseltamivir-resistant influenza. N Engl J Med. 2009 Mar;360(10):953-6. DOI: 10.1056/NEJMp0900648 Externer Link
80.
Bloom JD, Gong LI, Baltimore D. Permissive secondary mutations enable the evolution of influenza oseltamivir resistance. Science. 2010 Jun;328(5983):1272-5. DOI: 10.1126/science.1187816 Externer Link
81.
Schnitzler SU, Schnitzler P. An update on swine-origin influenza virus A/H1N1: a review. Virus Genes. 2009 Dec;39(3):279-92. DOI: 10.1007/s11262-009-0404-8 Externer Link
82.
Arbeitsgemeinschaft Influenza (AGI). Wochenberichte der AGI. [cited 2016 Aug 24]. Available from: https://influenza.rki.de/Wochenberichte.aspx Externer Link
83.
Shayegi N, Schweiger B, Duwe S, Pöhlmann C, Bornhäuser M, Ehninger G, Schetelig J. Antiviral treatment of influenza A (H1N1-09) guided by molecular resistance testing in aplasia after allo-SCT. Bone Marrow Transplant. 2011 Nov;46(11):1492-4. DOI: 10.1038/bmt.2010.331 Externer Link
84.
Rath B, Chen X, Spies V, Muehlhans S, Obermeier P, Tief F, Seeber L, Karsch K, Milde J, Skopnik H, Schweiger B, Duwe SC. Prospective surveillance of antiviral resistance in hospitalized infants less than 12 months of age with A(H3N2) influenza infection and treated with oseltamivir. Antivir Ther. 2017 Feb 16. DOI: 10.3851/IMP3141 Externer Link
85.
Hurt AC, Besselaar TG, Daniels RS, Ermetal B, Fry A, Gubareva L, Huang W, Lackenby A, Lee RT, Lo J, Maurer-Stroh S, Nguyen HT, Pereyaslov D, Rebelo-de-Andrade H, Siqueira MM, Takashita E, Tashiro M, Tilmanis D, Wang D, Zhang W, Meijer A. Global update on the susceptibility of human influenza viruses to neuraminidase inhibitors, 2014-2015. Antiviral Res. 2016 Aug;132:178-85. DOI: 10.1016/j.antiviral.2016.06.001 Externer Link
86.
Takashita E, Meijer A, Lackenby A, Gubareva L, Rebelo-de-Andrade H, Besselaar T, Fry A, Gregory V, Leang SK, Huang W, Lo J, Pereyaslov D, Siqueira MM, Wang D, Mak GC, Zhang W, Daniels RS, Hurt AC, Tashiro M. Global update on the susceptibility of human influenza viruses to neuraminidase inhibitors, 2013-2014. Antiviral Res. 2015 May;117:27-38. DOI: 10.1016/j.antiviral.2015.02.003 Externer Link