ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Antiviral drugs for influenza: Pharmacology and resistance

Antiviral drugs for influenza: Pharmacology and resistance
Literature review current through: Jan 2024.
This topic last updated: Jan 26, 2024.

INTRODUCTION — Seasonal influenza is an acute respiratory illness caused by influenza A or B viruses. Influenza occurs in outbreaks and epidemics worldwide, mainly during the winter season.

Issues related to pharmacology and resistance of antiviral drugs against influenza will be reviewed here. The approach to use antiviral agents for treatment and prevention of influenza, including the approach to treatment of suspected or known infection due to drug-resistant influenza virus, is discussed separately:

(See "Seasonal influenza in nonpregnant adults: Treatment".)

(See "Seasonal influenza and pregnancy".)

(See "Seasonal influenza in adults: Role of antiviral prophylaxis for prevention".)

(See "Seasonal influenza in children: Management".)

(See "Seasonal influenza in children: Prevention with antiviral drugs".)

Updated information about influenza activity and antiviral resistance can be found on the United States Centers for Disease Control and Prevention website and the World Health Organization website.

DRUG CLASSES — Classes of antiviral drugs for treatment of influenza include (table 1) [1-3]:

Neuraminidase inhibitors − Neuraminidase inhibitors include oseltamivir, zanamivir, peramivir, and laninamivir (approved for use in Japan but remains investigational elsewhere); they are active against influenza A and B. Neuraminidase inhibitors interfere with release of progeny influenza virus from infected cells, thereby preventing new rounds of infection.

Endonuclease inhibitorBaloxavir is a selective inhibitor of influenza cap-dependent endonuclease mediated by the viral polymerase acidic protein; it is active against influenza A and B.

Adamantanes − Adamantanes include amantadine and rimantadine; they are active against influenza A. These drugs target the M2 protein of influenza A, which forms a proton channel in the viral membrane that is essential for viral replication.

Due to emergence of high rates of adamantane resistance among influenza A viruses, the United States Centers for Disease Control and Prevention (CDC) recommends that adamantanes not be used for the treatment of influenza in the United States [1].

Neuraminidase inhibitors — Oseltamivir, zanamivir, peramivir, and, laninamivir (approved for use in Japan but remains investigational elsewhere) are structurally related drugs with activity against seasonal influenza as well as avian influenza A strains [4,5]. (See "Avian influenza: Epidemiology and transmission".)

General principles

Mechanism of action — Influenza hemagglutinin is a viral surface glycoprotein that binds to sialic acid residues on respiratory epithelial cell surface glycoproteins for initiation of infection. After viral replication, progeny virions are also bound to the host cell via sialic acid residues on cell surface glycoproteins. Neuraminidase-mediated removal of these sialic acid moieties permits release of progeny virions.

Neuraminidase inhibitors are sialic acid analogs that competitively inhibit neuraminidase; these drugs interfere with the release of progeny influenza virus from infected cells, thereby preventing new rounds of infection [6].

Pharmacokinetics

Oseltamivir − Oseltamivir phosphate is administered orally. It is available as a capsule or powder for liquid suspension that is rapidly metabolized to the active form, oseltamivir carboxylate [7]. A single 100 mg dose yields a peak plasma concentration of 250 mcg/L, with an elimination half-life of approximately eight hours [8]. Food does not affect peak concentration or overall systemic exposure. Elimination is primarily renal, and dose reduction is recommended for patients with reduced creatinine clearance.

Zanamivir − Zanamivir is administered as an inhaled powder and has poor oral bioavailability. Approximately 15 percent of inhaled zanamivir is deposited in the bronchi and lungs, with the remainder staying in the oropharynx [9]. It is highly concentrated in the respiratory tract [6]. Excretion is primarily renal, but, given limited systemic bioavailability, there is no need to modify the dose in patients with renal insufficiency. The pulmonary half-life is 2.8 hours [10]. Zanamivir inhalation powder should not be reconstituted in any liquid formulation and is not recommended for use in nebulizers or mechanical ventilators [11].

An intravenous (IV) formulation of zanamivir is available in the European Union and the United Kingdom but not in the United States. The usual recommended dose in adults is 600 mg every 12 hours for 5 to 10 days. The drug is renally cleared with a half-life of approximately two to three hours in healthy individuals. Dose adjustment is necessary in the setting of renal insufficiency [12].

Peramivir − Peramivir is administered as a single IV dose because it has a strong and prolonged affinity for influenza virus neuraminidase [13-15]. The recommended dose of peramivir is 600 mg IV and the elimination half-life in healthy adults is approximately 20 hours. Elimination is primarily renal, so dosing must be adjusted in the setting of renal insufficiency.

Adverse effects — Issues related to adverse effects are discussed separately. (See "Seasonal influenza in nonpregnant adults: Treatment", section on 'Antiviral efficacy and adverse effects'.)

Drug resistance — Updated information about resistance patterns among circulating influenza viruses can be found at the CDC website. A comprehensive list of neuraminidase inhibitor resistance mutations can be found at the World Health Organization (WHO) website.

Most neuraminidase inhibitor resistance affects oseltamivir and peramivir but not zanamivir. Resistance to the neuraminidase inhibitors appears to arise less readily than adamantane resistance.

Oseltamivir

Epidemiology

General principles – In general, oseltamivir resistance (and peramivir cross-resistance) is rare; since 2009, 99 percent of influenza virus isolates tested in the United States have been susceptible to neuraminidase inhibitors [1,16].

Prior to 2007, oseltamivir resistance was rare in clinical settings; in clinical trials, it emerged in 1 to 5 percent of cases [17-19].

During the 2007-2008 influenza season, oseltamivir-resistant isolates emerged in Europe [19-22]. None of the patients in the initial reports of oseltamivir-resistant influenza had been taking oseltamivir, suggesting transmission of resistant virus between individuals [20,23]. The severity of illness due to oseltamivir-resistant influenza was similar to oseltamivir-susceptible influenza [24,25].

During the 2008-2009 influenza season, high rates of oseltamivir resistance (>90 percent) were observed in the United States, Australia, and the Philippines [26,27].

During the 2009 H1N1 influenza A pandemic, the predominantly oseltamivir-susceptible pandemic strain replaced the oseltamivir-resistant seasonal H1N1 strain worldwide [28]. A small minority of pandemic H1N1 influenza A virus isolates with oseltamivir resistance were detected from patients in several countries, including Japan, the United States, China, Hong Kong, Singapore, Vietnam, Denmark, and Australia [29-38]. Among 37 cases in the United States, 76 percent occurred among immunocompromised patients and 89 percent occurred among patients who had received oseltamivir; thus, these appear to be actual risk factors for oseltamivir resistance [35].

However, in one community cluster of oseltamivir-resistant pandemic H1N1 influenza A infection in Vietnam, none of the individuals had received oseltamivir prophylaxis [33]. In addition, sustained community transmission of oseltamivir-resistant pandemic H1N1 influenza A was identified in 29 individuals in New South Wales, Australia [37,38]. Hemagglutinin and neuraminidase sequence analysis indicated that the resistant strains were closely related, suggesting spread of a single variant. Only one patient had received oseltamivir prior to collection of a respiratory specimen for resistance testing.

The neuraminidase mutation H275Y, which caused oseltamivir resistance among seasonal H1N1 influenza A isolates beginning in 2007, was also detected in isolates from patients with oseltamivir-resistant pandemic H1N1 influenza A infection [32,39]. Furthermore, community clusters of oseltamivir resistance in untreated individuals may be explained by the presence of permissive neuraminidase mutations that restore the viral fitness of H275Y [38,40].

Risk factors – Factors associated with the emergence of antiviral drug resistance include the administration of postexposure prophylaxis with a neuraminidase inhibitor (particularly if underdosed), immunosuppression, and prolonged antiviral treatment [41,42].

The increased risk among immunocompromised patients may be attributable to prolonged viral shedding despite antiviral therapy [43-49]. In one report of three immunocompromised patients, resistant influenza variants developed during treatment [46]. One patient received oseltamivir prophylaxis when a close contact was diagnosed with influenza; she subsequently developed respiratory symptoms and was started on full treatment doses of oseltamivir. Despite this, she died of influenza B infection; a virus subpopulation was recovered with neuraminidase and hemagglutinin mutations conferring reduced susceptibility to oseltamivir and zanamivir. Two other patients developed resistant influenza A virus with mutations in genes encoding the M2, neuraminidase, and hemagglutinin proteins; both viruses were resistant to oseltamivir but susceptible to zanamivir.

A similar pattern of oseltamivir resistance among immunocompromised patients was observed during the 2009 H1N1 influenza A pandemic. Two nosocomial clusters of oseltamivir-resistant pandemic H1N1 influenza A infection were identified in Wales and the United States [50,51]. In one study, 8 of 10 patients had oseltamivir-resistant virus and 4 of 8 were infected by direct transmission of resistant virus [50].

Mechanisms

Influenza A − Mechanisms of resistance observed among influenza A viruses include [52,53]:

Neuraminidase mutations − Mutations resulting in amino acid substitutions in the neuraminidase conserved active site (either part of catalytic residues or so-called framework amino acids surrounding the active site) usually result in drug-specific resistance.

-H275Y mutation − The most common neuraminidase mutation occurring in H1N1 viruses is the H275Y mutation (histidine to tyrosine substitution at amino acid 275); it is the mutation responsible for the oseltamivir resistance that emerged and spread among seasonal H1N1 influenza A viruses between 2007 and 2009 (but rarely among 2009 pandemic and postpandemic seasonal H1N1 influenza A viruses) [19,54].

The H275Y mutation reduces susceptibility of H1N1 influenza virus to oseltamivir (by more than 400-fold) and also reduces susceptibility to peramivir [55] but does not cause resistance to zanamivir in vitro [23,56-58]. This mutation is considered by WHO as clinically relevant due to its frequency and the availability of clinical data showing reduced treatment efficacy [59].

A nosocomial cluster of infection with an H1N1 influenza A virus harboring the H275Y mutation suggested that the mutated virus was transmissible and retained pathogenicity [60], possibly due to the presence of permissive or compensatory mutations, increasing the cell surface expression of the neuraminidase [61].

The H1N1 virus responsible for the 2009 pandemic, which has continued to circulate, has generally retained oseltamivir susceptibility, with approximately 1 percent of viruses exhibiting resistance, most frequently in immunocompromised patients [42].

The epidemiology of influenza viruses possessing the H275Y mutation is presented above. (See 'Epidemiology' above.)

-Other mutations – Less frequent mutations among H1N1 viruses include alterations at residues I223 and S247N (table 2) [62]. These are associated with low levels of oseltamivir resistance (usually less than a 10-fold increase in 50 percent inhibitory concentration [IC50] values) but can potentiate resistance conferred by the H275Y mutation.

Among H3N2 viruses, the most frequent mutations conferring oseltamivir resistance are E119V and R292K; R292K is also associated with reduced zanamivir inhibition [63]. Mutants with deletion of a few amino acids in the neuraminidase that exhibit resistance to oseltamivir have been observed in patients [43,44].

Hemagglutinin mutations − Mutations in the hemagglutinin at or near the site that binds to sialic acid residues, reducing the efficiency of viral binding; this leads to diminished dependence on neuraminidase for viral replication. Such mutations confer broad cross-resistance in vitro to oseltamivir, zanamivir, and some investigational neuraminidase inhibitors; however, these mutations are of uncertain clinical significance.

Influenza BOseltamivir resistance has been observed rarely among influenza B viruses; neuraminidase mutations conferring oseltamivir resistance are summarized in the table (table 2) [46,62,64-70].

During the 2010-2011 influenza season, a novel neuraminidase substitution, I221V, associated with reduced susceptibility to oseltamivir (two- to sixfold increase in IC50) was detected in North Carolina [67]. Among 209 clinical isolates of influenza B, the I221V mutation was detected in 22 percent of cases in North Carolina; it was observed in 10 percent of cases in South Carolina and 0.3 percent of cases from 45 other states. This mutation was not detected during the subsequent season.

Zanamivir — Very little zanamivir resistance has been observed, and the majority of cases of oseltamivir resistance have not resulted in cross-resistance to zanamivir. Few studies have directly compared the prevalence of oseltamivir and zanamivir resistance, given the rarity of zanamivir resistance and the less frequent use of zanamivir.

A few cases of zanamivir resistance associated with changes at residue 119 have been described among immunocompromised individuals (table 2). The E119I H3N2 variant, detected in a 2-year-old boy with lymphoproliferative disorder, exhibited reduced susceptibility to both zanamivir and peramivir and highly reduced susceptibility to oseltamivir [71].

Other substitutions were identified in patients infected with the 2009 H1N1 influenza A pandemic virus. In one report, a stem cell transplant recipient developed H1N1 influenza with both H275Y and E119D neuraminidase mutations; he had received both oseltamivir and zanamivir [72]. The H275Y mutation conferred resistance to oseltamivir and peramivir, whereas the E119D mutation conferred resistance to multiple neuraminidase inhibitors (zanamivir, oseltamivir, peramivir, and laninamivir). The combination of the two mutations increased the magnitude of neuraminidase inhibitor resistance further.

Another report describes an immunocompromised infant who developed influenza infection caused by an H1N1 influenza strain possessing H275Y and E119G (with glycine replacing glutamic acid at position 119) neuraminidase mutations following prolonged treatment with oseltamivir and zanamivir [73]. These mutations conferred resistance to multiple neuraminidase inhibitors.

Peramivir — For H1N1 viruses, there is generally cross-resistance between oseltamivir and peramivir; the H275Y mutation in the neuraminidase of H1N1 viruses confers high levels of resistance to both drugs [32].

Multidrug resistance

Multineuraminidase resistance − The E119D/G mutations have been associated with resistance to multiple neuraminidase inhibitors in pandemic H1N1 isolates [72,73]. In addition, changes at residue 223 (I223 R/V) in the 2009 H1N1 influenza A pandemic virus confer various levels of resistance to many neuraminidase inhibitors [74,75], especially when combined with the H275Y mutation [62].

Dual oseltamivir-adamantane resistance – Dual resistance to oseltamivir and adamantanes correlates with the extent of oseltamivir resistance. Among 1457 seasonal H1N1 influenza A isolates collected worldwide between 2008 and 2010, 28 viruses (1.9 percent) with dual resistance were detected from five countries [76]. Twenty-one of the dually resistant viruses were collected from China, four from the United States, and one each from Canada, Kenya, and Vietnam. The resistant viruses belonged to four genetic backgrounds and resulted from several mechanisms, including exchange of M and neuraminidase genes between clade 2B and clade 2C variants, emergence of mutations conferring adamantane resistance in oseltamivir-resistant viruses during antiviral therapy, transmission from others, and possibly spontaneously [76,77].

Baloxavir

General principles — Baloxavir is a selective inhibitor of influenza cap-dependent endonuclease; it blocks influenza proliferation by inhibiting the initiation of mRNA synthesis [78]. Baloxavir has activity against influenza A viruses, including H7N9 and H5N1 viruses, and influenza B viruses [78]. It has activity against influenza viruses that are resistant to oseltamivir and it appears to have synergistic activity with neuraminidase inhibitors.

Following administration, baloxavir marboxil undergoes hydrolysis to its active form, baloxavir, which is metabolized by UGT1A3 and CYP3A [78]. Baloxavir is primarily eliminated by biliary excretion. Its mean elimination half-life is 96 hours.

Coadministration of baloxavir with dairy products, calcium-fortified beverages, polyvalent cation-containing laxatives, antacids, or oral supplements (eg, calcium, iron, magnesium, selenium, zinc) should be avoided.

In a phase III trial, adverse events that were considered to be related to baloxavir were uncommon (eg, diarrhea in 1.8 percent) [79]. Hypersensitivity reactions (eg, anaphylaxis, urticaria, angioedema, erythema multiforme) have been reported in postmarketing surveillance [80].

Drug resistance — Emergence of polymerase acidic (PA) protein variants with I38T/M/F or E23K substitutions, which confer reduced susceptibility to baloxavir, has been described with variable frequency following a single dose of baloxavir (in up to 10 percent in trials of adolescents and adults [79,81] and in up to 24 percent in children [82-84]). In one study, these variants were associated with transient rises in viral titers, prolonged viral detection, and slower initial improvement of symptoms [81]. The emergence of resistance after a single dose raises concerns about the long-term utility of this drug as monotherapy, particularly if it is used widely.

The I38T substitution resulted in severely impaired replication in one in vitro study [85]; however, subsequent animal studies have suggested that viruses with the I38T substitution have similar replicative fitness, pathogenicity, and transmissibility as wild-type viruses [86-88]. Human transmission of this resistant variant has been documented [89,90]. The I38T substitution is considered by WHO as clinically relevant due to its frequency of occurrence and the availability of clinical data suggesting reduced treatment efficacy [91]. Other less frequent baloxavir resistance substitutions, such as I38F/M and E23K/G in the PA protein, have been reported [86,92]. For a comprehensive list of baloxavir resistance mutations, please refer to the WHO website.

Adamantanes — Amantadine and rimantadine are closely related adamantanes (also called M2 inhibitors) with comparable efficacy. Widespread emergence of adamantane-resistant influenza A strains has limited the clinical utility of these drugs; they should not be used routinely for influenza treatment or prophylaxis.

Mechanism of action — Resistance to amantadine and rimantadine is mediated by single nucleotide changes involving the transmembrane portion of the M2 molecule [93,94]. Such mutations confer cross-resistance between the adamantanes [94].

Drug resistance

General principles − The adamantanes target the M2 protein of influenza A viruses only. This protein forms an ion channel in the viral membrane that is essential for efficient viral replication [95-97].

Resistance to amantadine and rimantadine is mediated by single amino acid change (usually S31N) that has no effect on virus replication [77,98]. Such mutations confer cross-resistance between the adamantanes [94]. The rate of spontaneous mutations resulting in drug resistance in tissue culture is quite high, between 1:1000 and 1:10,000 [94]. Adamantane resistance can occur spontaneously or emerge as soon as two to three days following initiation of adamantane treatment [94]. Resistant viruses are genetically stable, virulent, and transmissible [99,100].

Mechanisms − Single-point mutations in the codons for amino acids at positions 26, 27, 30, 31, or 34 of the M2 protein affect the transmembrane portion of this protein, the M2 ion channel, and confer cross-resistance to both amantadine and rimantadine [93,94,101]. The mutation at position 31 is most common.

Epidemiology − Widespread dissemination of adamantane-resistant influenza was first noted in 2003 to 2004 among H3N2 viruses, primarily in Asia [101]. Substantial rates of adamantane resistance are now present worldwide in both H3N2 and H1N1 strains.

In the United States, the incidence of resistance was less than 2 percent until the 2004-2005 influenza season, when 15 percent of isolates demonstrated resistance [101,102]. During the 2005-2006 season, 92 percent of H3N2 influenza A viruses isolated from patients in 26 states contained an amino acid substitution at position 31 of the M2 protein, which confers resistance to both amantadine and rimantadine [103,104]. During the 2009-2010 influenza season in the United States, all H3N2 influenza A and 2009 pandemic H1N1 isolates tested were resistant to the adamantanes [105].

CLINICAL APPROACH TO DRUG RESISTANCE — Issues related to management of patients with suspected or known infection due to drug-resistant influenza virus are discussed separately. (See "Seasonal influenza in nonpregnant adults: Treatment", section on 'Antiviral resistance'.)

SUMMARY

Drug classes − Classes of antiviral drugs for treatment of influenza include neuraminidase inhibitors (oseltamivir, zanamivir, peramivir, and laninamivir), endonuclease inhibitor (baloxavir), and adamantanes (amantadine and rimantadine) (table 1). (See 'Drug classes' above.)

Clinical approach to drug resistance − Issues related to management of patients with suspected or known infection due to drug-resistant influenza virus are discussed separately. (See "Seasonal influenza in nonpregnant adults: Treatment", section on 'Antiviral resistance'.)

Neuraminidase inhibitor resistance

Epidemiology – Antiviral resistance is rare (approximately 1 percent); risk factors include administration of postexposure prophylaxis, prolonged antiviral treatment, and immunosuppression. (See 'Epidemiology' above.)

Mechanisms − Mechanisms of neuraminidase inhibitor resistance include mutations in neuraminidase (most commonly H275Y) and hemagglutinin (table 2). (See 'Mechanisms' above.)

Baloxavir resistance − The emergence of resistance after a single dose of baloxavir raises concerns about the long-term utility of this drug as monotherapy. The most common resistance mutation is I38T in the polymerase acidic protein. (See 'Baloxavir' above.).

Adamantane resistance − Widespread emergence of adamantane-resistant influenza A strains has limited the clinical utility of these drugs; they should not be used routinely for influenza treatment or prophylaxis. (See 'Adamantanes' above.)

  1. Centers for Disease Control and Prevention. Influenza antiviral medications: Summary for clinicians. https://www.cdc.gov/flu/professionals/antivirals/summary-clinicians.htm (Accessed on October 22, 2020).
  2. Uyeki TM, Bernstein HH, Bradley JS, et al. Clinical Practice Guidelines by the Infectious Diseases Society of America: 2018 Update on Diagnosis, Treatment, Chemoprophylaxis, and Institutional Outbreak Management of Seasonal Influenzaa. Clin Infect Dis 2019; 68:e1.
  3. Uyeki TM. Influenza. Ann Intern Med 2021; 174:ITC161.
  4. Leneva IA, Goloubeva O, Fenton RJ, et al. Efficacy of zanamivir against avian influenza A viruses that possess genes encoding H5N1 internal proteins and are pathogenic in mammals. Antimicrob Agents Chemother 2001; 45:1216.
  5. Leneva IA, Roberts N, Govorkova EA, et al. The neuraminidase inhibitor GS4104 (oseltamivir phosphate) is efficacious against A/Hong Kong/156/97 (H5N1) and A/Hong Kong/1074/99 (H9N2) influenza viruses. Antiviral Res 2000; 48:101.
  6. Moscona A. Neuraminidase inhibitors for influenza. N Engl J Med 2005; 353:1363.
  7. Davies BE. Pharmacokinetics of oseltamivir: an oral antiviral for the treatment and prophylaxis of influenza in diverse populations. J Antimicrob Chemother 2010; 65 Suppl 2:ii5.
  8. Bardsley-Elliot A, Noble S. Oseltamivir. Drugs 1999; 58:851.
  9. Cheer SM, Wagstaff AJ. Zanamivir: an update of its use in influenza. Drugs 2002; 62:71.
  10. Peng AW, Milleri S, Stein DS. Direct measurement of the anti-influenza agent zanamivir in the respiratory tract following inhalation. Antimicrob Agents Chemother 2000; 44:1974.
  11. US Food and Drug Administration. MedWatch Safety Alert. Relenza (zanamivir) inhalation powder. www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm186081.htm (Accessed on October 09, 2009).
  12. European Medicines Agency. Dectova. https://www.ema.europa.eu/en/documents/product-information/dectova-epar-product-information_en.pdf (Accessed on November 24, 2020).
  13. Kohno S, Kida H, Mizuguchi M, et al. Efficacy and safety of intravenous peramivir for treatment of seasonal influenza virus infection. Antimicrob Agents Chemother 2010; 54:4568.
  14. Rapivab (peramivir injection), for intravenous use, prescribing information. http://www.accessdata.fda.gov/drugsatfda_docs/label/2016/206426s003lbl.pdf (Accessed on November 20, 2020).
  15. Scott LJ. Peramivir: A Review in Uncomplicated Influenza. Drugs 2018; 78:1363.
  16. Centers for Disease Control and Prevention. Past Flu Seasons. https://www.cdc.gov/flu/season/past-flu-seasons.htm (Accessed on October 14, 2021).
  17. Hayden FG, Pavia AT. Antiviral management of seasonal and pandemic influenza. J Infect Dis 2006; 194 Suppl 2:S119.
  18. Kiso M, Mitamura K, Sakai-Tagawa Y, et al. Resistant influenza A viruses in children treated with oseltamivir: descriptive study. Lancet 2004; 364:759.
  19. World Health Organization. Influenza A(H1N1) virus resistance to oseltamivir http://www.who.int/influenza/patient_care/antivirals/oseltamivir_summary/en/ (Accessed on October 18, 2011).
  20. Lackenby A, Hungnes O, Dudman SG, et al. Emergence of resistance to oseltamivir among influenza A(H1N1) viruses in Europe. Euro Surveill 2008; 13.
  21. Meijer A, Lackenby A, Hungnes O, et al. Oseltamivir-resistant influenza virus A (H1N1), Europe, 2007-08 season. Emerg Infect Dis 2009; 15:552.
  22. Hauge SH, Dudman S, Borgen K, et al. Oseltamivir-resistant influenza viruses A (H1N1), Norway, 2007-08. Emerg Infect Dis 2009; 15:155.
  23. World Health Organization. WHO/ECDC frequently asked questions for Oseltamivir Resistance http://www.who.int/influenza/patient_care/antivirals/oseltamivir_faqs/en/ (Accessed on October 18, 2011).
  24. Dharan NJ, Gubareva LV, Meyer JJ, et al. Infections with oseltamivir-resistant influenza A(H1N1) virus in the United States. JAMA 2009; 301:1034.
  25. Weinstock DM, Zuccotti G. The evolution of influenza resistance and treatment. JAMA 2009; 301:1066.
  26. Centers for Disease Control and Prevention. Flu View: A weekly influenza surveillance report prepared by the influenza division. 2009-2009 influenza season week ending January 31, 2009 http://www.cdc.gov/flu/weekly/ (Accessed on October 18, 2011).
  27. World Health Organization. Influeza A (H1N1) virus resistance to oseltamivir. 2008 Southern hemisphere influenza season http://www.bepast.org/disease_information/Pandemic%20Influenza/Pandemic%20Influenza%20Information/2008_08_20H1N1_WHO_oseltamivir_resistance.pdf (Accessed on October 18, 2011).
  28. Whitley RJ, Boucher CA, Lina B, et al. Global assessment of resistance to neuraminidase inhibitors, 2008-2011: the Influenza Resistance Information Study (IRIS). Clin Infect Dis 2013; 56:1197.
  29. Centers for Disease Control and Prevention (CDC). Update: influenza activity - United States, August 30-October 31, 2009. MMWR Morb Mortal Wkly Rep 2009; 58:1236.
  30. Centers for Disease Control and Prevention (CDC). Oseltamivir-resistant novel influenza A (H1N1) virus infection in two immunosuppressed patients - Seattle, Washington, 2009. MMWR Morb Mortal Wkly Rep 2009; 58:893.
  31. Centers for Disease Control and Prevention (CDC). Oseltamivir-resistant 2009 pandemic influenza A (H1N1) virus infection in two summer campers receiving prophylaxis--North Carolina, 2009. MMWR Morb Mortal Wkly Rep 2009; 58:969.
  32. Baz M, Abed Y, Papenburg J, et al. Emergence of oseltamivir-resistant pandemic H1N1 virus during prophylaxis. N Engl J Med 2009; 361:2296.
  33. Le QM, Wertheim HF, Tran ND, et al. A community cluster of oseltamivir-resistant cases of 2009 H1N1 influenza. N Engl J Med 2010; 362:86.
  34. Inoue M, Barkham T, Leo YS, et al. Emergence of oseltamivir-resistant pandemic (H1N1) 2009 virus within 48 hours. Emerg Infect Dis 2010; 16:1633.
  35. Graitcer SB, Gubareva L, Kamimoto L, et al. Characteristics of patients with oseltamivir-resistant pandemic (H1N1) 2009, United States. Emerg Infect Dis 2011; 17:255.
  36. Morlighem JÉ, Aoki S, Kishima M, et al. Mutation analysis of 2009 pandemic influenza A(H1N1) viruses collected in Japan during the peak phase of the pandemic. PLoS One 2011; 6:e18956.
  37. Hurt AC, Hardie K, Wilson NJ, et al. Community transmission of oseltamivir-resistant A(H1N1)pdm09 influenza. N Engl J Med 2011; 365:2541.
  38. Hurt AC, Hardie K, Wilson NJ, et al. Characteristics of a widespread community cluster of H275Y oseltamivir-resistant A(H1N1)pdm09 influenza in Australia. J Infect Dis 2012; 206:148.
  39. European Center for Disease Prevention and Control. Oseltamivir-resistant pandemic (H1N1) 2009 influenza virus, October 2009 http://www.ecdc.europa.eu/en/activities/sciadvice/Lists/ECDC%20Reviews/ECDC_DispForm.aspx?List=512ff74f-77d4-4ad8-b6d6-bf0f23083f30&ID=683 (Accessed on November 19, 2009).
  40. Abed Y, Pizzorno A, Bouhy X, et al. Impact of potential permissive neuraminidase mutations on viral fitness of the H275Y oseltamivir-resistant influenza A(H1N1)pdm09 virus in vitro, in mice and in ferrets. J Virol 2014; 88:1652.
  41. Boivin G. Detection and management of antiviral resistance for influenza viruses. Influenza Other Respir Viruses 2013; 7 Suppl 3:18.
  42. Hurt AC, Chotpitayasunondh T, Cox NJ, et al. Antiviral resistance during the 2009 influenza A H1N1 pandemic: public health, laboratory, and clinical perspectives. Lancet Infect Dis 2012; 12:240.
  43. Abed Y, Baz M, Boivin G. A novel neuraminidase deletion mutation conferring resistance to oseltamivir in clinical influenza A/H3N2 virus. J Infect Dis 2009; 199:180.
  44. Memoli MJ, Hrabal RJ, Hassantoufighi A, et al. Rapid selection of a transmissible multidrug-resistant influenza A/H3N2 virus in an immunocompromised host. J Infect Dis 2010; 201:1397.
  45. Memoli MJ, Hrabal RJ, Hassantoufighi A, et al. Rapid selection of oseltamivir- and peramivir-resistant pandemic H1N1 virus during therapy in 2 immunocompromised hosts. Clin Infect Dis 2010; 50:1252.
  46. Ison MG, Gubareva LV, Atmar RL, et al. Recovery of drug-resistant influenza virus from immunocompromised patients: a case series. J Infect Dis 2006; 193:760.
  47. Calatayud L, Lackenby A, Reynolds A, et al. Oseltamivir-resistant pandemic (H1N1) 2009 virus infection in England and Scotland, 2009-2010. Emerg Infect Dis 2011; 17:1807.
  48. Tramontana AR, George B, Hurt AC, et al. Oseltamivir resistance in adult oncology and hematology patients infected with pandemic (H1N1) 2009 virus, Australia. Emerg Infect Dis 2010; 16:1068.
  49. Ghedin E, Holmes EC, DePasse JV, et al. Presence of oseltamivir-resistant pandemic A/H1N1 minor variants before drug therapy with subsequent selection and transmission. J Infect Dis 2012; 206:1504.
  50. Moore C, Galiano M, Lackenby A, et al. Evidence of person-to-person transmission of oseltamivir-resistant pandemic influenza A(H1N1) 2009 virus in a hematology unit. J Infect Dis 2011; 203:18.
  51. Chen LF, Dailey NJ, Rao AK, et al. Cluster of oseltamivir-resistant 2009 pandemic influenza A (H1N1) virus infections on a hospital ward among immunocompromised patients--North Carolina, 2009. J Infect Dis 2011; 203:838.
  52. Gubareva LV, Kaiser L, Hayden FG. Influenza virus neuraminidase inhibitors. Lancet 2000; 355:827.
  53. Gubareva LV. Molecular mechanisms of influenza virus resistance to neuraminidase inhibitors. Virus Res 2004; 103:199.
  54. Moscona A. Global transmission of oseltamivir-resistant influenza. N Engl J Med 2009; 360:953.
  55. Pizzorno A, Bouhy X, Abed Y, Boivin G. Generation and characterization of recombinant pandemic influenza A(H1N1) viruses resistant to neuraminidase inhibitors. J Infect Dis 2011; 203:25.
  56. Ong AK, Hayden FG. John F. Enders lecture 2006: antivirals for influenza. J Infect Dis 2007; 196:181.
  57. Mishin VP, Hayden FG, Gubareva LV. Susceptibilities of antiviral-resistant influenza viruses to novel neuraminidase inhibitors. Antimicrob Agents Chemother 2005; 49:4515.
  58. Sheu TG, Deyde VM, Okomo-Adhiambo M, et al. Surveillance for neuraminidase inhibitor resistance among human influenza A and B viruses circulating worldwide from 2004 to 2008. Antimicrob Agents Chemother 2008; 52:3284.
  59. World Health Organization (WHO). Global Influenza Programme. Laboratory methodologies for testing the antiviral susceptibility of influenza viruses: Neuraminidase inhibitor (NAI). https://www.who.int/teams/global-influenza-programme/laboratory-network/quality-assurance/antiviral-susceptibility-influenza/neuraminidase-inhibitor (Accessed on January 10, 2024).
  60. Gooskens J, Jonges M, Claas EC, et al. Morbidity and mortality associated with nosocomial transmission of oseltamivir-resistant influenza A(H1N1) virus. JAMA 2009; 301:1042.
  61. Bloom JD, Gong LI, Baltimore D. Permissive secondary mutations enable the evolution of influenza oseltamivir resistance. Science 2010; 328:1272.
  62. Samson M, Pizzorno A, Abed Y, Boivin G. Influenza virus resistance to neuraminidase inhibitors. Antiviral Res 2013; 98:174.
  63. Pizzorno A, Abed Y, Boivin G. Influenza drug resistance. Semin Respir Crit Care Med 2011; 32:409.
  64. Hatakeyama S, Sugaya N, Ito M, et al. Emergence of influenza B viruses with reduced sensitivity to neuraminidase inhibitors. JAMA 2007; 297:1435.
  65. Carr S, Ilyushina NA, Franks J, et al. Oseltamivir-resistant influenza A and B viruses pre- and postantiviral therapy in children and young adults with cancer. Pediatr Infect Dis J 2011; 30:284.
  66. Escuret V, Frobert E, Bouscambert-Duchamp M, et al. Detection of human influenza A (H1N1) and B strains with reduced sensitivity to neuraminidase inhibitors. J Clin Virol 2008; 41:25.
  67. Garg S, Moore Z, Lee N, et al. A cluster of patients infected with I221V influenza b virus variants with reduced oseltamivir susceptibility--North Carolina and South Carolina, 2010-2011. J Infect Dis 2013; 207:966.
  68. Escuret V, Collins PJ, Casalegno JS, et al. A novel I221L substitution in neuraminidase confers high-level resistance to oseltamivir in influenza B viruses. J Infect Dis 2014; 210:1260.
  69. van der Vries E, Ip DK, Cowling BJ, et al. Outcomes and Susceptibility to Neuraminidase Inhibitors in Individuals Infected With Different Influenza B Lineages: The Influenza Resistance Information Study. J Infect Dis 2016; 213:183.
  70. Fage C, Abed Y, Checkmahomed L, et al. In Vitro Properties and Virulence of Contemporary Recombinant Influenza B Viruses Harboring Mutations of Cross-Resistance to Neuraminidase Inhibitors. Viruses 2018; 11.
  71. Okomo-Adhiambo M, Demmler-Harrison GJ, Deyde VM, et al. Detection of E119V and E119I mutations in influenza A (H3N2) viruses isolated from an immunocompromised patient: challenges in diagnosis of oseltamivir resistance. Antimicrob Agents Chemother 2010; 54:1834.
  72. L'Huillier AG, Abed Y, Petty TJ, et al. E119D Neuraminidase Mutation Conferring Pan-Resistance to Neuraminidase Inhibitors in an A(H1N1)pdm09 Isolate From a Stem-Cell Transplant Recipient. J Infect Dis 2015; 212:1726.
  73. Tamura D, DeBiasi RL, Okomo-Adhiambo M, et al. Emergence of Multidrug-Resistant Influenza A(H1N1)pdm09 Virus Variants in an Immunocompromised Child Treated With Oseltamivir and Zanamivir. J Infect Dis 2015; 212:1209.
  74. Nguyen HT, Fry AM, Loveless PA, et al. Recovery of a multidrug-resistant strain of pandemic influenza A 2009 (H1N1) virus carrying a dual H275Y/I223R mutation from a child after prolonged treatment with oseltamivir. Clin Infect Dis 2010; 51:983.
  75. van der Vries E, Stelma FF, Boucher CA. Emergence of a multidrug-resistant pandemic influenza A (H1N1) virus. N Engl J Med 2010; 363:1381.
  76. Sheu TG, Fry AM, Garten RJ, et al. Dual resistance to adamantanes and oseltamivir among seasonal influenza A(H1N1) viruses: 2008-2010. J Infect Dis 2011; 203:13.
  77. Hayden FG, de Jong MD. Emerging influenza antiviral resistance threats. J Infect Dis 2011; 203:6.
  78. Heo YA. Baloxavir: First Global Approval. Drugs 2018; 78:693.
  79. Hayden FG, Sugaya N, Hirotsu N, et al. Baloxavir Marboxil for Uncomplicated Influenza in Adults and Adolescents. N Engl J Med 2018; 379:913.
  80. Xofluza (baloxavir marboxil) for oral use, prescribing information. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/210854s001lbl.pdf (Accessed on October 17, 2019).
  81. Uehara T, Hayden FG, Kawaguchi K, 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.
  82. Hirotsu N, Sakaguchi H, Sato C, et al. Baloxavir Marboxil in Japanese Pediatric Patients With Influenza: Safety and Clinical and Virologic Outcomes. Clin Infect Dis 2020; 71:971.
  83. Wagatsuma K, Saito R, Chon I, et al. Duration of fever and symptoms in influenza-infected children treated with baloxavir marboxil during the 2019-2020 season in Japan and detection of influenza virus with the PA E23K substitution. Antiviral Res 2022; 201:105310.
  84. Saito R, Osada H, Wagatsuma K, et al. Duration of fever and symptoms in children after treatment with baloxavir marboxil and oseltamivir during the 2018-2019 season and detection of variant influenza a viruses with polymerase acidic subunit substitutions. Antiviral Res 2020; 183:104951.
  85. Omoto S, Speranzini V, Hashimoto T, et al. Characterization of influenza virus variants induced by treatment with the endonuclease inhibitor baloxavir marboxil. Sci Rep 2018; 8:9633.
  86. Imai M, Yamashita M, Sakai-Tagawa Y, 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.
  87. Checkmahomed L, M'hamdi Z, Carbonneau J, 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 2020; 221:63.
  88. Chesnokov A, Patel MC, Mishin VP, et al. Replicative Fitness of Seasonal Influenza A Viruses With Decreased Susceptibility to Baloxavir. J Infect Dis 2020; 221:367.
  89. Takashita E, Ichikawa M, Morita H, 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.
  90. Takashita E, Fujisaki S, Morita H, et al. A community cluster of influenza A(H3N2) virus infection with reduced susceptibility to baloxavir due to a PA E199G substitution in Japan, February to March 2023. Euro Surveill 2023; 28.
  91. World Health Organization (WHO). Global Influenza Programme. Laboratory methodologies for testing the antiviral susceptibility of influenza viruses: Polymerase acidic (PA) inhibitor, Baloxavir. https://www.who.int/teams/global-influenza-programme/laboratory-network/quality-assurance/antiviral-susceptibility-influenza/polymerase-acidic-protein-inhibitor (Accessed on January 10, 2024).
  92. Takashita E, Abe T, Morita H, et al. Influenza A(H1N1)pdm09 virus exhibiting reduced susceptibility to baloxavir due to a PA E23K substitution detected from a child without baloxavir treatment. Antiviral Res 2020; 180:104828.
  93. Hayden FG, Belshe RB, Clover RD, et al. Emergence and apparent transmission of rimantadine-resistant influenza A virus in families. N Engl J Med 1989; 321:1696.
  94. Belshe RB, Smith MH, Hall CB, et al. Genetic basis of resistance to rimantadine emerging during treatment of influenza virus infection. J Virol 1988; 62:1508.
  95. Bui M, Whittaker G, Helenius A. Effect of M1 protein and low pH on nuclear transport of influenza virus ribonucleoproteins. J Virol 1996; 70:8391.
  96. Tosteson MT, Pinto LH, Holsinger LJ, Lamb RA. Reconstitution of the influenza virus M2 ion channel in lipid bilayers. J Membr Biol 1994; 142:117.
  97. Takeda M, Pekosz A, Shuck K, et al. Influenza a virus M2 ion channel activity is essential for efficient replication in tissue culture. J Virol 2002; 76:1391.
  98. Nelson MI, Simonsen L, Viboud C, et al. The origin and global emergence of adamantane resistant A/H3N2 influenza viruses. Virology 2009; 388:270.
  99. Hayden FG, Sperber SJ, Belshe RB, et al. Recovery of drug-resistant influenza A virus during therapeutic use of rimantadine. Antimicrob Agents Chemother 1991; 35:1741.
  100. Hayden FG, Hay AJ. Emergence and transmission of influenza A viruses resistant to amantadine and rimantadine. Curr Top Microbiol Immunol 1992; 176:119.
  101. Bright RA, Medina MJ, Xu X, et al. Incidence of adamantane resistance among influenza A (H3N2) viruses isolated worldwide from 1994 to 2005: a cause for concern. Lancet 2005; 366:1175.
  102. Hayden FG. Antiviral resistance in influenza viruses--implications for management and pandemic response. N Engl J Med 2006; 354:785.
  103. CDC Health Alert. 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 www.cdc.gov/flu/han011406.htm (Accessed on October 18, 2011).
  104. Bright RA, Shay DK, Shu B, et al. Adamantane resistance among influenza A viruses isolated early during the 2005-2006 influenza season in the United States. JAMA 2006; 295:891.
  105. Centers for Disease Control and Prevention (CDC). Update: influenza activity - United States, 2009-10 season. MMWR Morb Mortal Wkly Rep 2010; 59:901.
Topic 7008 Version 42.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟