Publikationsserver der Universitätsbibliothek Marburg

Titel:Untersuchungen zur Optimierung der genetischen Konstellation von Influenzaimpfstoff-Saatviren unter Verwendung einer attenuierten Mutante des Virusisolats A/FPV/Rostock/1934 (H7N1)
Autor:Alex, Nina
Weitere Beteiligte: Bölker, Michael (Prof. Dr.)
Veröffentlicht:2013
URI:https://archiv.ub.uni-marburg.de/diss/z2014/0224
DOI: https://doi.org/10.17192/z2014.0224
URN: urn:nbn:de:hebis:04-z2014-02241
DDC: Biowissenschaften, Biologie
Titel (trans.):Studies to optimize the genetic constellation of influenza vaccine seed viruses using an attenuated mutant of the virus isolate A/FPV/Rostock/1934 (H7N1)
Publikationsdatum:2014-10-21
Lizenz:https://rightsstatements.org/vocab/InC-NC/1.0/

Dokument

Schlagwörter:
Reassortanten, Antigen, Impfstoffsaatviren, reassortants, Influenza-A-Viren, Influenza-A-Viren, vaccine-seed, Antigen, Impfstoffsaatviren, Influenza-A-virus, antigen, Reassortanten

Zusammenfassung:
Influenza-A-Viren lösen beim Menschen hochansteckende Infektionen der Atemwege aus, die in Abhängigkeit des Immunstatus der Person und der Pathogenität des Virus unterschiedlich schwere Krankheitsverläufe nehmen können. Eine Impfung bietet hierbei den effektivsten Schutz vor einer Infektion und den damit einhergehenden gesundheitlichen Komplikationen. Zum jetzigen Zeitpunkt erfolgt die Herstellung der Impfstoffe überwiegend in embryonierten Hühnereiern, jedoch zunehmend auch in zellkulturbasierten Vermehrungssystemen (MDCK- und Vero-Zellen). Einer der wichtigsten limitierenden Faktoren für die ausreichende Impfstoffversorgung der Bevölkerung stellt dabei die erzeugbare Antigenausbeute dar, die abhängig von dem eingesetzten Saatvirus stark variabel sein kann. Insbesondere bei neu auftretenden pandemischen Viren, wie z.B. denen der H1N1-Pandemie 2009 sowie den H5N1-Isolaten, sind die erzielbaren Antigenmengen häufig sehr niedrig. In dieser Arbeit wurde mit dem Fokus höhere Antigenausbeuten zu erzielen, die Eignung der apathogenen, attenuierten Variante FPVmut des hochpathogenen aviären Influenzastammes A/Huhn/Rostock/1934 (H7N1) als neuer, alternativer Akzeptorstamm für die Generierung der Impfstoffsaatviren im Vergleich zu dem gegenwärtig durch die WHO empfohlenen humanen H1N1-Stamm A/Puerto Rico/8/1934 (PR8) analysiert. Zusätzlich wurde der Einfluss der Inkorporation des zu den Oberflächenantigenen Hämagglutinin und Neuraminidase (HA und NA) homologen M-Gens in die Saatviren untersucht. Hierzu wurden mittels des Reversen-Genetik-Systems Doppel-Reassortanten (HA+NA) sowie Triple-Reassortanten (HA+NA+M) in den beiden zu vergleichenden Akzeptorstämmen hergestellt. Bei dieser Reassortierungsmethode kann die genetische Zusammensetzung der zu erzeugenden rekombinanten Viren vorab gezielt festgelegt werden, da die Virusgenerierung ausschließlich ausgehend von klonierter cDNA erfolgt. Hierzu werden acht für die Gensegmente kodierenden Plasmide in Zellen transfiziert. Als Donorviren wurden sowohl saisonale Viren der Subtypen H1N1 und H3N2 als auch (potenziell) pandemische Isolate der Subtypen H1N1pdm09, H5N1 und H9N2 eingesetzt. FPVmut zeichnet sich bei Vermehrung in embryonierten Hühnereiern sowie MDCK- und Vero-Zellen durch identische Ausbeuten im Vergleich mit dem aufgrund seiner guten Wachstumseigenschaften für die Erzeugung von Influenzasaatviren standardmäßig eingesetzten PR8-Isolat aus. Zusätzlich erwiesen sich beide Hintergrundstämme grundsätzlich als gleichermaßen geeignet für die Erzeugung rekombinanter Viren, da mit ihnen im selben Zeitrahmen ein identischer Satz an Reassortanten erzeugt werden konnte. Bei der Untersuchung der Replikationseigenschaften zeigte sich, dass bei den Viren mit dem FPVmut-Hintergrund eine produktive MDCK-Zellinfektion zu früheren Zeitpunkten einsetzte und ergänzend bei den aviären Donor-Isolaten eine gesteigerte Infektionsausbreitung beobachtet werden konnte. Kein wesentlicher Unterschied war in der NA-Ausbeute oder der Virusmorphologie in Abhängigkeit von dem Hintergrundstamm oder der genetischen Zusammensetzung festzustellen. Bei Verwendung des aviären H5N1 konnte eine deutliche Steigerung der Antigenausbeute im Hühnerei- und noch signifikanter im Vero-Anzuchtsystem erzielt werden, wodurch sich für dieses potenziell pandemische Isolat das FPVmut als ein leistungsfähigerer Akzeptorstamm im Vergleich zu dem PR8-Standardsystem herausstellte. Hierbei wurden sowohl höhere gemittelte als auch maximal erreichbare HA-Titer sowie höhere absolute HA-Antigenmengen in der Allantoisflüssigkeit infizierter Hühnereier gemessen. Bei der Vermehrung in Vero-Zellen wurden in den Doppel-Reassortanten nach einer 72-stündigen Inkubation 3fach höhere HA-Antigenmengen im FPVmut Akzeptorstamm erhalten. Im Falle des H9N2-Isolates bewirkte die Inkorporation des homologen M-Gensegmentes im FPVmut-Hintergrund eine Steigerung der Antigenausbeute um den Faktor 1,5. Auch bei dem H1N1pdm09-Isolat wurden die höchsten Einzelwerte der HA-Ausbeuten nach Inkorporation des M-Gensegmentes im PR8-Hintergrund gemessen. Somit konnte gezeigt werden, dass sich isolatabhängig Triple-Reassortanten als sinnvolle Variation der genetischen Zusammensetzung der Saatviren eignen können und die Verwendung eines aviären Akzeptorstammes zu einer deutlichen Steigerung der erzielbaren HA-Antigenausbeute in unterschiedlichen Vermehrungssystemen führen kann.

Bibliographie / References

  1. Salk JE, Pearson HE, . Immunization against influenza with observations during an epidemic of influenza A one year after vaccination. Am J Hyg 1945 Nov;42:307-22.
  2. Keitel WA, Atmar RL, Cate TR, et al. Safety of high doses of influenza vaccine and effect on antibody responses in elderly persons. Arch Intern Med 2006 May 22;166(10):1121-7.
  3. Elleman CJ, Barclay WS. The M1 matrix protein controls the filamentous phenotype of influenza A virus. Virology 2004 Mar 30;321(1):144-53.
  4. Neumann G, Kawaoka Y. Host range restriction and pathogenicity in the context of influenza pandemic. Emerg Infect Dis 2006 Jun;12(6):881-6.
  5. Olsen B, Munster VJ, Wallensten A, Waldenstrom J, Osterhaus AD, Fouchier RA. Global patterns of influenza a virus in wild birds. Science 2006 Apr 21;312(5772):384- 8.
  6. Kilbourne ED, MURPHY JS. Genetic studies of influenza viruses. I. Viral morphology and growth capacity as exchangeable genetic traits. Rapid in ovo adaptation of early passage Asian strain isolates by combination with PR8. J Exp Med 1960 Mar 1;111:387-406.
  7. Suguitan AL, Jr., McAuliffe J, Mills KL, et al. Live, attenuated influenza A H5N1 candidate vaccines provide broad cross-protection in mice and ferrets. PLoS Med 2006 Sep;3(9):e360.
  8. Fulvini AA, Ramanunninair M, Le J, et al. Gene constellation of influenza A virus reassortants with high growth phenotype prepared as seed candidates for vaccine production. PLoS One 2011;6(6):e20823.
  9. Fouchier RA, Munster V, Wallensten A, et al. Characterization of a novel influenza A virus hemagglutinin subtype (H16) obtained from black-headed gulls. J Virol 2005 Mar;79(5):2814-22.
  10. Claas EC, Osterhaus AD, van BR, et al. Human influenza A H5N1 virus related to a highly pathogenic avian influenza virus. Lancet 1998 Feb 14;351(9101):472-7.
  11. Subbarao K, Chen H, Swayne D, et al. Evaluation of a genetically modified reassortant H5N1 influenza A virus vaccine candidate generated by plasmid-based reverse genetics. Virology 2003 Jan 5;305(1):192-200.
  12. Dormitzer PR. Cell culture-derived Influenza vaccines. In: Rappuoli R, Del Giudice G, editors. Influenza vaccines for the future. 2nd ed. Basel, Springer, 2011: p. 293-312.
  13. Oxford J, Gilbert A, Lambkin-Williams R. Influenza vaccines have a short but illustrious history of dedicated science enabling the rapid global production of A/swine (H1N1) vaccine in the current pandemic. In: Rappuoli R, Del Giudice G, editors. Influenza vaccines for the future. 2nd ed. Basel, Springer, 2011: p. 115-47.
  14. Neumann G, Kawaoka Y. Reverse genetics systems for the generation of segmented negative-sense RNA viruses entirely from cloned cDNA. Curr Top Microbiol Immunol 2004;283:43-60.
  15. Modrow S, Falke D, Truyen U, Schätzl H. Orthomyxoviren. Molekulare Virologie. 3th ed. 2010: p. 355-84.
  16. Cate TR. Clinical manifestations and consequences of influenza. Am J Med 1987 Jun 19;82(6A):15-9.
  17. Huang RT, Rott R, Klenk HD. Influenza viruses cause hemolysis and fusion of cells. Virology 1981 Apr 15;110(1):243-7.
  18. Webster RG, Kawaoka Y, Bean WJ. What is the potential of avirulent influenza viruses to complement a cleavable hemagglutinin and generate virulent strains? Virology 1989 Aug;171(2):484-92.
  19. Garcia-Sastre A, Palese P. The cytoplasmic tail of the neuraminidase protein of influenza A virus does not play an important role in the packaging of this protein into viral envelopes. Virus Res 1995 Jun;37(1):37-47.
  20. Collin N, de R, X. Vaccine production capacity for seasonal and pandemic (H1N1) 2009 influenza. Vaccine 2009 Aug 20;27(38):5184-6.
  21. Partridge J, Kieny MP. Global production of seasonal and pandemic (H1N1) influenza vaccines in 2009-2010 and comparison with previous estimates and global action plan targets. Vaccine 2010 Jul 5;28(30):4709-12.
  22. Schmitt AP, Lamb RA. Influenza virus assembly and budding at the viral budozone. Adv Virus Res 2005;64:383-416.
  23. Halperin SA, Smith B, Mabrouk T, et al. Safety and immunogenicity of a trivalent, inactivated, mammalian cell culture-derived influenza vaccine in healthy adults, seniors, and children. Vaccine 2002 Jan 15;20(7-8):1240-7.
  24. Shortridge KF. Poultry and the influenza H5N1 outbreak in Hong Kong, 1997: abridged chronology and virus isolation. Vaccine 1999 Jul 30;17 Suppl 1:S26-S29.
  25. Simonsen L, Taylor RJ, Viboud C, Miller MA, Jackson LA. Mortality benefits of influenza vaccination in elderly people: an ongoing controversy. Lancet Infect Dis 2007 Oct;7(10):658-66.
  26. Lalor PA, Webby RJ, Morrow J, et al. Plasmid DNA-based vaccines protect mice and ferrets against lethal challenge with A/Vietnam/1203/04 (H5N1) influenza virus. J Infect Dis 2008 Jun 15;197(12):1643-52.
  27. Frey S, Vesikari T, Szymczakiewicz-Multanowska A, et al. Clinical efficacy of cell culture-derived and egg-derived inactivated subunit influenza vaccines in healthy adults. Clin Infect Dis 2010 Nov 1;51(9):997-1004.
  28. Wright PF, Cherry JD, Foy HM, et al. Antigenicity and reactogenicity of influenza A/USSR/77 virus vaccine in children--a multicentered evaluation of dosage and safety. Rev Infect Dis 1983 Jul;5(4):758-64.
  29. Wright PF, Thompson J, Vaughn WK, Folland DS, Sell SH, Karzon DT. Trials of influenza A/New Jersey/76 virus vaccine in normal children: an overview of age- related antigenicity and reactogenicity. J Infect Dis 1977 Dec;136 Suppl:S731-S741.
  30. Baez M, Palese P, Kilbourne ED. Gene composition of high-yielding influenza vaccine strains obtained by recombination. J Infect Dis 1980 Mar;141(3):362-5.
  31. Wise HM, Barbezange C, Jagger BW, et al. Overlapping signals for translational regulation and packaging of influenza A virus segment 2. Nucleic Acids Res 2011 Sep 1;39(17):7775-90.
  32. Schild GC, Henry-Aymard M, Pereira HG. A quantitative, single-radial-diffusion test for immunological studies with influenza virus. J Gen Virol 1972 Aug;16(2):231-6.
  33. McCauley JW, Mahy BW, Inglis SC. Nucleotide sequence of fowl plague virus RNA segment 7. J Gen Virol 1982 Jan;58 Pt 1:211-5.
  34. Yewdell JW, Ince WL. Virology. Frameshifting to PA-X influenza. Science 2012 Jul 13;337(6091):164-5.
  35. Garten W, Klenk H-D. Cleavage activation of the influenza virus hemagglutinin and its role in pathogenesis. In: Klenk HD, Matrosovich.M.N., Stech J, editors. Avian Influenza. Vol 27 ed. Basel, Karger, 2008: p. 156-67.
  36. Ito T, Kawaoka Y, Ohira M, et al. Replacement of internal protein genes, with the exception of the matrix, in equine 1 viruses by equine 2 influenza virus genes during evolution in nature. J Vet Med Sci 1999 Aug;61(8):987-9.
  37. Bergeron C, Valette M, Lina B, Ottmann M. Genetic content of Influenza H3N2 vaccine seeds. PLoS Curr 2010;2:RRN1165.
  38. Epstein SL, Price GE. Cross-protective immunity to influenza A viruses. Expert Rev Vaccines 2010 Nov;9(11):1325-41.
  39. Belser JA, Bridges CB, Katz JM, Tumpey TM. Past, present, and possible future human infection with influenza virus A subtype H7. Emerg Infect Dis 2009
  40. Yedidia TB. The 3rd international conference on Influenza Vaccines for the World (IVW 2009). Hum Vaccin 2009 Aug;5(8):508-9.
  41. Wise HM, Hutchinson EC, Jagger BW, et al. Identification of a novel splice variant form of the influenza a virus m2 ion channel with an antigenically distinct ectodomain. PLoS Pathog 2012 Nov;8(11):e1002998.
  42. Abt M, de JJ, Laue M, Wolff T. Improvement of H5N1 influenza vaccine viruses: influence of internal gene segments of avian and human origin on production and hemagglutinin content. Vaccine 2011 Jul 18;29(32):5153-62.
  43. Hoffmann E, Stech J, Guan Y, Webster RG, Perez DR. Universal primer set for the full-length amplification of all influenza A viruses. Arch Virol 2001 Dec;146(12):2275- 89.
  44. Horimoto T, Takada A, Fujii K, et al. The development and characterization of H5 influenza virus vaccines derived from a 2003 human isolate. Vaccine 2006 Apr 24;24(17):3669-76.
  45. Isken B, Genzel Y, Reichl U. Productivity, apoptosis, and infection dynamics of influenza A/PR/8 strains and A/PR/8-based reassortants. Vaccine 2012 Jul 27;30(35):5253-61.
  46. Schulze-Horsel J, Schulze M, Agalaridis G, Genzel Y, Reichl U. Infection dynamics and virus-induced apoptosis in cell culture-based influenza vaccine production-Flow cytometry and mathematical modeling. Vaccine 2009 May 5;27(20):2712-22.
  47. Neirynck S, Deroo T, Saelens X, Vanlandschoot P, Jou WM, Fiers W. A universal influenza A vaccine based on the extracellular domain of the M2 protein. Nat Med 1999 Oct;5(10):1157-63.
  48. Fiers W, De FM, Birkett A, Neirynck S, Min JW. A "universal" human influenza A vaccine. Virus Res 2004 Jul;103(1-2):173-6.
  49. De FM, Fiers W, Martens W, et al. Improved design and intranasal delivery of an M2e-based human influenza A vaccine. Vaccine 2006 Nov 10;24(44-46):6597-601.
  50. Fiers W, De FM, El BK, et al. M2e-based universal influenza A vaccine. Vaccine 2009 Oct 23;27(45):6280-3.
  51. Lambert LC, Fauci AS. Influenza vaccines for the future. N Engl J Med 2010 Nov 18;363(21):2036-44.
  52. Stephenson I, Gust I, Pervikov Y, Kieny MP. Development of vaccines against influenza H5. Lancet Infect Dis 2006 Aug;6(8):458-60.
  53. Calcagnile S, Zuccotti GV. The virosomal adjuvanted influenza vaccine. Expert Opin Biol Ther 2010 Feb;10(2):191-200.
  54. Arbeitsgemeinschaft Influenza. Bericht zur Epidemiologie der Influenza in Deutschland Saison 2011/12; URL:http://influenza.rki.de/Saisonberichte/2011.pdf [Zugriff 05.02.2013]. Berlin; 2012.
  55. Ben-Yedidia T. Multimeric-001: BiondVax's universal flu vaccine. Interview by Duc Le. Expert Rev Vaccines 2010 Mar;9(3):241-2.
  56. DiNapoli JM, Nayak B, Yang L, et al. Newcastle disease virus-vectored vaccines expressing the hemagglutinin or neuraminidase protein of H5N1 highly pathogenic avian influenza virus protect against virus challenge in monkeys. J Virol 2010 Feb;84(3):1489-503.
  57. Corti D, Suguitan AL, Jr., Pinna D, et al. Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine. J Clin Invest 2010 May;120(5):1663-73.
  58. Mubareka S, Palese P. Influenza virus: The Biology of a changing virus. In: Rappuoli R, Del Giudice G, editors. Influenza vaccines for the future. 2nd ed. Basel, Springer, 2011: p. 3-26.
  59. Shortridge KF, Stuart-Harris CH. An influenza epicentre? Lancet 1982 Oct 9;2(8302):812-3.
  60. Chen W, Calvo PA, Malide D, et al. A novel influenza A virus mitochondrial protein that induces cell death. Nat Med 2001 Dec;7(12):1306-12.
  61. Leroux-Roels I, Borkowski A, Vanwolleghem T, et al. Antigen sparing and cross- reactive immunity with an adjuvanted rH5N1 prototype pandemic influenza vaccine: a randomised controlled trial. Lancet 2007 Aug 18;370(9587):580-9.
  62. Nayak DP, Hui EK, Barman S. Assembly and budding of influenza virus. Virus Res 2004 Dec;106(2):147-65.
  63. Murphy BR, Kasel JA, Chanock RM. Association of serum anti-neuraminidase antibody with resistance to influenza in man. N Engl J Med 1972 Jun 22;286(25):1329-32.
  64. Shinya K, Ebina M, Yamada S, Ono M, Kasai N, Kawaoka Y. Avian flu: influenza virus receptors in the human airway. Nature 2006 Mar 23;440(7083):435-6.
  65. Subbarao K, Klimov A, Katz J, et al. Characterization of an avian influenza A (H5N1) virus isolated from a child with a fatal respiratory illness. Science 1998 Jan 16;279(5349):393-6.
  66. Palese P, Tobita K, Ueda M, Compans RW. Characterization of temperature sensitive influenza virus mutants defective in neuraminidase. Virology 1974 Oct;61(2):397-410.
  67. Murphy BR, Buckler-White AJ, London WT, Snyder MH. Characterization of the M protein and nucleoprotein genes of an avian influenza A virus which are involved in host range restriction in monkeys. Vaccine 1989 Dec;7(6):557-61.
  68. Committee for Proprietary medicinal products. Note for guidance on harmonisation of requirements for influenza vaccines CHMP/BWP/214/96. 1997 Mar 12.
  69. Nobusawa E, Aoyama T, Kato H, Suzuki Y, Tateno Y, Nakajima K. Comparison of complete amino acid sequences and receptor-binding properties among 13 serotypes of hemagglutinins of influenza A viruses. Virology 1991 Jun;182(2):475-85.
  70. Scholtissek C, Rott R, Orlich M, Harms E, Rohde W. Correlation of pathogenicity and gene constellation of an influenza A virus (fowl plague). I. Exchange of a single gene. Virology 1977 Aug;81(1):74-80.
  71. O´Hagan DT, Tsai T, Reed S. Emulsion-based adjuvants for improved influenza vaccines. In: Rappuoli R, Del Giudice G, editors. Influenza vaccines for the future. 2nd ed. Basel, Springer, 2011: p. 327-57.
  72. Lazarowitz SG, Choppin PW. Enhancement of the infectivity of influenza A and B viruses by proteolytic cleavage of the hemagglutinin polypeptide. Virology 1975 Dec;68(2):440-54.
  73. Robert Koch Institut. Epidemiologisches Bulletin Aktuelle Daten und Informationen zu Infektionskrankheiten und Public Health. Epidemiologisches Bulletin 2012 Jul 30;30:283-310.
  74. Webster RG, Bean WJ, Gorman OT, Chambers TM, Kawaoka Y. Evolution and ecology of influenza A viruses. Microbiol Rev 1992 Mar;56(1):152-79.
  75. Xu KM, Li KS, Smith GJ, et al. Evolution and molecular epidemiology of H9N2 influenza A viruses from quail in southern China, 2000 to 2005. J Virol 2007 Mar;81(6):2635-45.
  76. Wood JM, Robertson JS. From lethal virus to life-saving vaccine: developing inactivated vaccines for pandemic influenza. Nat Rev Microbiol 2004 Oct;2(10):842-7.
  77. Kageyama T, Fujisaki S, Takashita E, et al. Genetic analysis of novel avian A(H7N9) influenza viruses isolated from patients in China, February to April 2013. Euro Surveill 2013;18(15).
  78. Stevens J, Blixt O, Paulson JC, Wilson IA. Glycan microarray technologies: tools to survey host specificity of influenza viruses. Nat Rev Microbiol 2006 Nov;4(11):857-64.
  79. Fiore AE, Bridges CB, Katz JM, Cox NJ. Inactivated influenza vaccines. In: Plotkin SA, Orenstein WA, Offit PA, editors. Vaccines. 6th ed. Elsevier Saunders, 2013: p. 257-93.
  80. Mischler R, Metcalfe IC. Inflexal V a trivalent virosome subunit influenza vaccine: production. Vaccine 2002 Dec 20;20 Suppl 5:B17-B23.
  81. Garcia-Sastre A, Egorov A, Matassov D, et al. Influenza A virus lacking the NS1 gene replicates in interferon-deficient systems. Virology 1998 Dec 20;252(2):324-30.
  82. Luke CJ, Lakdawala SS, Subbarao K. Influenza vaccine -live. In: Plotkin SA, Orenstein WA, Offit PA, editors. Vaccines. 6th ed. Elsevier Saunders, 2013: p. 294- 311.
  83. Pushko P, Tumpey TM, Bu F, Knell J, Robinson R, Smith G. Influenza virus-like particles comprised of the HA, NA, and M1 proteins of H9N2 influenza virus induce protective immune responses in BALB/c mice. Vaccine 2005 Dec 30;23(50):5751-9.
  84. Pinto LH, Holsinger LJ, Lamb RA. Influenza virus M2 protein has ion channel activity. Cell 1992 May 1;69(3):517-28.
  85. Ruigrok RW, Barge A, Durrer P, Brunner J, Ma K, Whittaker GR. Membrane interaction of influenza virus M1 protein. Virology 2000 Feb 15;267(2):289-98.
  86. Subbarao K, Shaw MW. Molecular aspects of avian influenza (H5N1) viruses isolated from humans. Rev Med Virol 2000 Sep;10(5):337-48.
  87. Plant EP, Liu TM, Xie H, Ye Z. Mutations to A/Puerto Rico/8/34 PB1 gene improves seasonal reassortant influenza A virus growth kinetics. Vaccine 2012 Dec 17;31(1):207-12.
  88. Palese P, Shaw ML. Orthomyxoviridae: the viruses and their replication. In: Knipe D, Howley P, editors. Fields virology. 5th ed. Philadelphia, Lippincott Williams and Wilkins, 2007: p. 1647-89.
  89. Scholtissek C. Pigs as Mixing Vessels for the Creation of New Pandemic Influenza A Viruses. Medical Principles and Practice 1990 Jan;2(2):65-71.
  90. Kreijtz JH, Suezer Y, de MG, et al. Preclinical evaluation of a modified vaccinia virus Ankara (MVA)-based vaccine against influenza A/H5N1 viruses. Vaccine 2009 Oct 23;27(45):6296-9.
  91. Wood JM, Major D, Newman RW, et al. Preparation of vaccines against H5N1 influenza. Vaccine 2002 May 15;20 Suppl 2:S84-S87.
  92. Murphy BR, Coelingh K. Principles underlying the development and use of live attenuated cold-adapted influenza A and B virus vaccines. Viral Immunol 2002;15(2):295-323.
  93. Wang H, Feng Z, Shu Y, et al. Probable limited person-to-person transmission of highly pathogenic avian influenza A (H5N1) virus in China. Lancet 2008 Apr 26;371(9622):1427-34.
  94. Garten W, Bosch FX, Linder D, Rott R, Klenk HD. Proteolytic activation of the influenza virus hemagglutinin: The structure of the cleavage site and the enzymes involved in cleavage. Virology 1981 Dec;115(2):361-74.
  95. Bosch FX, Garten W, Klenk HD, Rott R. Proteolytic cleavage of influenza virus hemagglutinins: primary structure of the connecting peptide between HA1 and HA2 determines proteolytic cleavability and pathogenicity of Avian influenza viruses. Virology 1981 Sep;113(2):725-35.
  96. Rogers GN, Paulson JC. Receptor determinants of human and animal influenza virus isolates: differences in receptor specificity of the H3 hemagglutinin based on species of origin. Virology 1983 Jun;127(2):361-73.
  97. Connor RJ, Kawaoka Y, Webster RG, Paulson JC. Receptor specificity in human, avian, and equine H2 and H3 influenza virus isolates. Virology 1994 Nov 15;205(1):17-23.
  98. Bourmakina SV, Garcia-Sastre A. Reverse genetics studies on the filamentous morphology of influenza A virus. J Gen Virol 2003 Mar;84(Pt 3):517-27.
  99. Steinhauer DA. Role of hemagglutinin cleavage for the pathogenicity of influenza virus. Virology 1999 May 25;258(1):1-20.
  100. Lingwood D, McTamney PM, Yassine HM, et al. Structural and genetic basis for development of broadly neutralizing influenza antibodies. Nature 2012 Sep 27;489(7417):566-70.
  101. Xu R, Ekiert DC, Krause JC, Hai R, Crowe JE, Jr., Wilson IA. Structural basis of preexisting immunity to the 2009 H1N1 pandemic influenza virus. Science 2010 Apr 16;328(5976):357-60.
  102. Bullough PA, Hughson FM, Skehel JJ, Wiley DC. Structure of influenza haemagglutinin at the pH of membrane fusion. Nature 1994 Sep 1;371(6492):37-43.
  103. Stevens J, Corper AL, Basler CF, Taubenberger JK, Palese P, Wilson IA. Structure of the uncleaved human H1 hemagglutinin from the extinct 1918 influenza virus. Science 2004 Mar 19;303(5665):1866-70.
  104. Knobler SL, Mack A, Mahmoud A, Lemon SM. Summary and Assessment. In: Institue of medicine of the national academies, editor. The threat of pandemic influenza -are we ready?Washington DC, The national academies press, 2005.
  105. Feldmann A, Schafer MK, Garten W, Klenk HD. Targeted infection of endothelial cells by avian influenza virus A/FPV/Rostock/34 (H7N1) in chicken embryos. J Virol 2000 Sep;74(17):8018-27.
  106. Desselberger U, Racaniello VR, Zazra JJ, Palese P. The 3' and 5'-terminal sequences of influenza A, B and C virus RNA segments are highly conserved and show partial inverted complementarity. Gene 1980 Feb;8(3):315-28.
  107. Subbarao EK, Perkins M, Treanor JJ, Murphy BR. The attenuation phenotype conferred by the M gene of the influenza A/Ann Arbor/6/60 cold-adapted virus (H2N2) on the A/Korea/82 (H3N2) reassortant virus results from a gene constellation effect. Virus Res 1992 Sep 1;25(1-2):37-50.
  108. Karlsson Hedestam GB, Fouchier RA, Phogat S, Burton DR, Sodroski J, Wyatt RT. The challenges of eliciting neutralizing antibodies to HIV-1 and to influenza virus. Nat Rev Microbiol 2008 Feb;6(2):143-55.
  109. The European Directorate for the Quality of Medicines. European Pharmacopoeia. 7 ed. 2012.
  110. Lamb RA, Choppin PW. The gene structure and replication of influenza virus. Annu Rev Biochem 1983;52:467-506.
  111. Roberts PC, Lamb RA, Compans RW. The M1 and M2 proteins of influenza A virus are important determinants in filamentous particle formation. Virology 1998 Jan 5;240(1):127-37.
  112. Belshe RB. The origins of pandemic influenza--lessons from the 1918 virus. N Engl J Med 2005 Nov 24;353(21):2209-11.
  113. Wiley DC, Skehel JJ. The structure and function of the hemagglutinin membrane glycoprotein of influenza virus. Annu Rev Biochem 1987;56:365-94.
  114. Barman S, Ali A, Hui EK, Adhikary L, Nayak DP. Transport of viral proteins to the apical membranes and interaction of matrix protein with glycoproteins in the assembly of influenza viruses. Virus Res 2001 Sep;77(1):61-9.
  115. Galarza JM, Latham T, Cupo A. Virus-like particle vaccine conferred complete protection against a lethal influenza virus challenge. Viral Immunol 2005;18(2):365- 72.
  116. Pringle CR. Virus taxonomy 1. Arch Virol 1996;141(11):2251-6.
  117. Palese P, Compans RW. Inhibition of influenza virus replication in tissue culture by 2- deoxy-2,3-dehydro-N-trifluoroacetylneuraminic acid (FANA): mechanism of action. J Gen Virol 1976 Oct;33(1):159-63.
  118. OIE. On avian influenza: Manual of Diagnostic tests ans vaccines; URL:http://web.oie.int/eng/normes/mmanual/2008/pdf/2.03.04_AI.pdf [Zugriff 17.01.2013]. 2009. 17-1-2013.
  119. World Health Organisation. WHO guidance on development of influenza vaccine reference viruses by reverse genetics; URL:http://whqlibdoc.who.int/hq/2005/WHO_CDS_CSR_GIP_2005.6.pdf [Zugriff 24.07.2012]. 2012. 24-7-2012.
  120. World Health Organisation. Global pandemic influenza action plan to increase vaccine supply; URL:http://whqlibdoc.who.int/hq/2006/WHO_IVB_06.13_eng.pdf [Zugriff: 11.03.2013]. WHO/IVB/06.13;WHO/CDS/EPR/GIP/2006.1 . 2013. 11-3-2013.
  121. Skehel JJ, Wiley DC. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu Rev Biochem 2000;69:531-69.
  122. Duggan ST, Plosker GL. Intanza 15 microg intradermal seasonal influenza vaccine: in older adults (aged >or=60 years). Drugs Aging 2010 Jul 1;27(7):597-605. [130] Prophylactic vaccinations; URL:http://www.en.influenza.pl/ida/prophylactic_vaccinations.html [Zugriff 08.02.2013]. 2013. 8-2-2013.
  123. Krauss U, Eggert T. Ligation Calculator; URL:http://www.insilico.uni-duesseldorf.de/ [Zugriff 16.04.2013]. 2013. 16-4-2013.
  124. Selman M, Dankar SK, Forbes NE, Jia JJ, Brown EG. Adaptive mutation in influenza A virus non-structural gene is linked to host switching and induces a novel protein by alternative splicing. Emerg Microbes Infect 2012 Nov 21;1:e42.
  125. Tashiro M, Ciborowski P, Klenk HD, Pulverer G, Rott R. Role of Staphylococcus protease in the development of influenza pneumonia. Nature 1987 Feb 5;325(6104):536-7.
  126. Wagner R, Herwig A, Azzouz N, Klenk HD. Acylation-mediated membrane anchoring of avian influenza virus hemagglutinin is essential for fusion pore formation and virus infectivity. J Virol 2005 May;79(10):6449-58.
  127. Ito T, Couceiro JN, Kelm S, et al. Molecular basis for the generation in pigs of influenza A viruses with pandemic potential. J Virol 1998 Sep;72(9):7367-73.
  128. Gomez-Puertas P, Albo C, Perez-Pastrana E, Vivo A, Portela A. Influenza virus matrix protein is the major driving force in virus budding. J Virol 2000
  129. Fodor E, Devenish L, Engelhardt OG, Palese P, Brownlee GG, Garcia-Sastre A. Rescue of influenza A virus from recombinant DNA. J Virol 1999 Nov;73(11):9679-82.
  130. Latham T, Galarza JM. Formation of wild-type and chimeric influenza virus-like particles following simultaneous expression of only four structural proteins. J Virol 2001 Jul;75(13):6154-65.
  131. Ali A, Avalos RT, Ponimaskin E, Nayak DP. Influenza virus assembly: effect of influenza virus glycoproteins on the membrane association of M1 protein. J Virol 2000 Sep;74(18):8709-19.
  132. Reid AH, Fanning TG, Janczewski TA, McCall S, Taubenberger JK. Characterization of the 1918 "Spanish" influenza virus matrix gene segment. J Virol 2002 Nov;76(21):10717-23.
  133. McCown MF, Pekosz A. Distinct domains of the influenza a virus M2 protein cytoplasmic tail mediate binding to the M1 protein and facilitate infectious virus production. J Virol 2006 Aug;80(16):8178-89.
  134. Neumann G, Watanabe T, Ito H, et al. Generation of influenza A viruses entirely from cloned cDNAs. Proc Natl Acad Sci U S A 1999 Aug 3;96(16):9345-50.
  135. Hoffmann E, Neumann G, Kawaoka Y, Hobom G, Webster RG. A DNA transfection system for generation of influenza A virus from eight plasmids. Proc Natl Acad Sci U S A 2000 May 23;97(11):6108-13.
  136. Bui M, Whittaker G, Helenius A. Effect of M1 protein and low pH on nuclear transport of influenza virus ribonucleoproteins. J Virol 1996 Dec;70(12):8391-401.
  137. Chen BJ, Leser GP, Morita E, Lamb RA. Influenza virus hemagglutinin and neuraminidase, but not the matrix protein, are required for assembly and budding of plasmid-derived virus-like particles. J Virol 2007 Jul;81(13):7111-23.
  138. Cheung CL, Vijaykrishna D, Smith GJ, et al. Establishment of influenza A virus (H6N1) in minor poultry species in southern China. J Virol 2007 Oct;81(19):10402-12.
  139. Horimoto T, Kawaoka Y. Reverse genetics provides direct evidence for a correlation of hemagglutinin cleavability and virulence of an avian influenza A virus. J Virol 1994 May;68(5):3120-8.
  140. A revision of the system of nomenclature for influenza viruses: a WHO memorandum. Bull World Health Organ 1980;58(4):585-91.
  141. Simpson DA, Lamb RA. Alterations to influenza virus hemagglutinin cytoplasmic tail modulate virus infectivity. J Virol 1992 Feb;66(2):790-803.
  142. Kilbourne ED. Future influenza vaccines and the use of genetic recombinants. Bull World Health Organ 1969;41(3):643-5.
  143. Tian SF, Buckler-White AJ, London WT, Reck LJ, Chanock RM, Murphy BR. Nucleoprotein and membrane protein genes are associated with restriction of replication of influenza A/Mallard/NY/78 virus and its reassortants in squirrel monkey respiratory tract. J Virol 1985 Mar;53(3):771-5.
  144. Chen BJ, Leser GP, Jackson D, Lamb RA. The influenza virus M2 protein cytoplasmic tail interacts with the M1 protein and influences virus assembly at the site of virus budding. J Virol 2008 Oct;82(20):10059-70.
  145. Hai R, Martinez-Sobrido L, Fraser KA, Ayllon J, Garcia-Sastre A, Palese P. Influenza B virus NS1-truncated mutants: live-attenuated vaccine approach. J Virol 2008 Nov;82(21):10580-90.
  146. Steel J, Lowen AC, Pena L, et al. Live attenuated influenza viruses containing NS1 truncations as vaccine candidates against H5N1 highly pathogenic avian influenza. J Virol 2009 Feb;83(4):1742-53.
  147. Nugent KM, Pesanti EL. Tracheal function during influenza infections. Infect Immun 1983 Dec;42(3):1102-8.
  148. Sui J, Hwang WC, Perez S, et al. Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses. Nat Struct Mol Biol 2009 Mar;16(3):265-73.
  149. Wise HM, Foeglein A, Sun J, et al. A complicated message: Identification of a novel PB1-related protein translated from influenza A virus segment 2 mRNA. J Virol 2009
  150. Nayak DP, Balogun RA, Yamada H, Zhou ZH, Barman S. Influenza virus morphogenesis and budding. Virus Res 2009 Aug;143(2):147-61.
  151. Neumann G, Noda T, Kawaoka Y. Emergence and pandemic potential of swine-origin H1N1 influenza virus. Nature 2009 Jun 18;459(7249):931-9.
  152. Pandey A, Singh N, Sambhara S, Mittal SK. Egg-independent vaccine strategies for highly pathogenic H5N1 influenza viruses. Hum Vaccin 2010 Feb;6(2):178-88.
  153. Vemula SV, Mittal SK. Production of adenovirus vectors and their use as a delivery system for influenza vaccines. Expert Opin Biol Ther 2010 Oct;10(10):1469-87.
  154. Neumann G, Hughes MT, Kawaoka Y. Influenza A virus NS2 protein mediates vRNP nuclear export through NES-independent interaction with hCRM1. EMBO J 2000 Dec 15;19(24):6751-8.
  155. Lamb RA, Lai CJ, Choppin PW. Sequences of mRNAs derived from genome RNA segment 7 of influenza virus: colinear and interrupted mRNAs code for overlapping proteins. Proc Natl Acad Sci U S A 1981 Jul;78(7):4170-4.
  156. Tong S, Li Y, Rivailler P, et al. A distinct lineage of influenza A virus from bats. Proc Natl Acad Sci U S A 2012 Mar 13;109(11):4269-74.
  157. Fouchier RA, Schneeberger PM, Rozendaal FW, et al. Avian influenza A virus (H7N7) associated with human conjunctivitis and a fatal case of acute respiratory distress syndrome. Proc Natl Acad Sci U S A 2004 Feb 3;101(5):1356-61.
  158. Tellier R. Review of aerosol transmission of influenza A virus. Emerg Infect Dis 2006
  159. Shi M, Jagger BW, Wise HM, Digard P, Holmes EC, Taubenberger JK. Evolutionary Conservation of the PA-X Open Reading Frame in Segment 3 of Influenza A Virus. J Virol 2012 Nov;86(22):12411-3.
  160. Paterson D, Fodor E. Emerging Roles for the Influenza A Virus Nuclear Export Protein (NEP). PLoS Pathog 2012 Dec;8(12):e1003019.
  161. Jagger BW, Wise HM, Kash JC, et al. An overlapping protein-coding region in influenza A virus segment 3 modulates the host response. Science 2012 Jul 13;337(6091):199-204.
  162. Kilbourne ED, Laver WG, Schulman JL, Webster RG. Antiviral activity of antiserum specific for an influenza virus neuraminidase. J Virol 1968 Apr;2(4):281-8.
  163. Schulman JL, Khakpour M, Kilbourne ED. Protective effects of specific immunity to viral neuraminidase on influenza virus infection of mice. J Virol 1968 Aug;2(8):778-86.
  164. Licensure of a high-dose inactivated influenza vaccine for persons aged >or=65 years (Fluzone High-Dose) and guidance for use -United States, 2010. MMWR Morb Mortal Wkly Rep 2010 Apr 30;59(16):485-6.
  165. Khan K, Arino J, Hu W, et al. Spread of a novel influenza A (H1N1) virus via global airline transportation. N Engl J Med 2009 Jul 9;361(2):212-4.
  166. Musher DM. How contagious are common respiratory tract infections? N Engl J Med 2003 Mar 27;348(13):1256-66.
  167. Robert Koch-Institut. Antworten des RKI auf häufig gestellte Fragen zur saisonalen Influenzaimpfung Für wen wird die Impfung gegen die saisonale Influenza empfohlen?; URL:http://www.rki.de/DE/Content/Infekt/Impfen/FAQ/Influenza/faq_ges.html?nn=237 5548 [Zugriff 04.02.2013]. 7-9-2012. 4-2-2013.
  168. Robert Koch-Institut. Antworten des RKI auf häufig gestellte Fragen zur saisonalen Influenzaimpfung Warum wird die Influenzaimpfung auch für Schwangere empfohlen?; URL:http://www.rki.de/SharedDocs/FAQ/Impfen/Influenza/FAQ03.html?nn=2370434 [Zugriff 04.02.2013]. 7-9-2012. 4-2-2013.
  169. Maeda T, Ohnishi S. Activation of influenza virus by acidic media causes hemolysis and fusion of erythrocytes. FEBS Lett 1980 Dec 29;122(2):283-7.
  170. Klenk HD, Rott R, Orlich M, Blodorn J. Activation of influenza A viruses by trypsin treatment. Virology 1975 Dec;68(2):426-39.
  171. Lubeck MD, Palese P, Schulman JL. Nonrandom association of parental genes in influenza A virus recombinants. Virology 1979 May;95(1):269-74.
  172. Johansson BE, Bucher DJ, Pokorny BA, Mikhail A, Kilbourne ED. Identification of PR8 M1 protein in influenza virus high-yield reassortants by M1-specific monoclonal antibodies. Virology 1989 Aug;171(2):634-6.
  173. Zhirnov OP. Solubilization of matrix protein M1/M from virions occurs at different pH for orthomyxo-and paramyxoviruses. Virology 1990 May;176(1):274-9.
  174. Godley L, Pfeifer J, Steinhauer D, et al. Introduction of intersubunit disulfide bonds in the membrane-distal region of the influenza hemagglutinin abolishes membrane fusion activity. Cell 1992 Feb 21;68(4):635-45.
  175. Klenk HD, Garten W. Host cell proteases controlling virus pathogenicity. Trends Microbiol 1994 Feb;2(2):39-43.
  176. Jin H, Lu B, Zhou H, et al. Multiple amino acid residues confer temperature sensitivity to human influenza virus vaccine strains (FluMist) derived from cold-adapted A/Ann Arbor/6/60. Virology 2003 Feb 1;306(1):18-24.
  177. Zhirnov OP, Grigoriev VB. Disassembly of influenza C viruses, distinct from that of influenza A and B viruses requires neutral-alkaline pH. Virology 1994 Apr;200(1):284- 91.
  178. Peiris M, Yuen KY, Leung CW, et al. Human infection with influenza H9N2. Lancet 1999 Sep 11;354(9182):916-7.
  179. Kreijtz JH, Fouchier RA, Rimmelzwaan GF. Immune responses to influenza virus infection. Virus Res 2011 Dec;162(1-2):19-30.
  180. Kieny MP, Costa A, Hombach J, et al. A global pandemic influenza vaccine action plan. Vaccine 2006 Sep 29;24(40-41):6367-70.
  181. Taylor DN, Treanor JJ, Sheldon EA, et al. Development of VAX128, a recombinant hemagglutinin (HA) influenza-flagellin fusion vaccine with improved safety and immune response. Vaccine 2012 Aug 24;30(39):5761-9.
  182. Claas EC, de Jong JC, van BR, Rimmelzwaan GF, Osterhaus AD. Human influenza virus A/HongKong/156/97 (H5N1) infection. Vaccine 1998 May;16(9-10):977-8.
  183. World Health Organisation. Human infection with influenza A(H7N9) virus in China; URL:http://www.who.int/csr/don/2013_04_01/en/index.html [Zugriff 14.04.2013]. 2013. 14-4-2013.
  184. World Health Organisation. Human infection with influenza A(H7N9) virus in China - update 12.04.2013; URL:http://www.who.int/csr/don/2013_04_12/en/index.html [Zugriff 14.04.2013]. 2013. 14-4-2013.
  185. World Health Organisation Global influenza programme. WHO manual on animal influenza diagnosis and surveillance; URL:http://www.who.int/csr/resources/publications/influenza/whocdscsrncs20025rev. pdf [Zugriff 31.01.2012]. WHO/CDS/CSR/NCS/2002.5 Rev.1 . 2002. 31-1-2012.
  186. World Health Organisation. A Description of the Process of Seasonal and H5N1 Influenza Vaccine Virus Selection and Development; URL:http://www.who.int/influenza/resources/documents/Fluvaccvirusselection.pdf [Zugriff 07.12.2012]. 2012. 7-12-2012.
  187. World Health Organisation. Influenza vaccine viruses and reagents; URL:http://www.who.int/influenza/vaccines/virus/en/ [Zugriff 06.02.2013]. 2013. 6-2- 2013.
  188. World Health Organisation. Global action plan for influenza vaccines (GAP); URL:http://www.who.int/influenza_vaccines_plan/en/ [Zugriff 21.02.2013]. 2013. 21- 2-2013.
  189. World Health Organisation. Global action plan for influenza vaccines: Objective 2 Increase in vaccine production capacity; URL:http://www.who.int/influenza_vaccines_plan/objectives/objective2/en/index.html [Zugriff 21.02.2013]. 2013. 21-2-2013.
  190. Production of pilot lots of inactivated influenza vaccine in response to a pandemic threat: an interim biosafety risk assessment. Wkly Epidemiol Rec 2003 Nov 21;78(47):405-8.


* Das Dokument ist im Internet frei zugänglich - Hinweise zu den Nutzungsrechten