Scolaris Content Display Scolaris Content Display

نقش واکسن‌ها در پیشگیری از ابتلا به آنفلوآنزا در بزرگسالان سالم

Contraer todo Desplegar todo

Referencias

aa Barrett 2011 {published data only}

Barrett PN, Berezuk G, Fritsch S, Aichinger G, Hart MK, El‐Amin W, et al. Efficacy, safety, and immunogenicity of a Vero‐cell‐culture‐derived trivalent influenza vaccine: a multicentre, double‐blind, randomised, placebo‐controlled trial. Lancet 2011;377(9767):751‐9. CENTRAL
Ehrlich HJ, Berezuk G, Fritsch S, Aichinger G, Singer J, Portsmouth D, et al. Clinical development of a Vero cell culture‐derived seasonal influenza vaccine. Vaccine 2011;30(29):4377‐86. CENTRAL
Ehrlich HJ, Singer J, Berezuk G, Fritsch S, Aichinger G, Hart MK, et al. A cell culture‐derived influenza vaccine provides consistent protection against infection and reduces the duration and severity of disease in infected individuals. Clinical Infectious Diseases 2012;54(7):946‐54. CENTRAL

aa Beran 2009a {published data only}

Beran J, Wertzova V, Honegr K, Kaliskova E, Havlickova M, Havlik J, et al. Challenge of conducting a placebo‐controlled randomized efficacy study for influenza vaccine in a season with low attack rate and a mismatched vaccine B strain: a concrete example. BMC Infectious Diseases 2009;9:2. [DOI: 10.1186/1471‐2334‐9‐2]CENTRAL

aa Beran 2009b {published data only}

Beran J, Vesikari T, Wertzova V, Karvonen A, Honegr K, Lindblad N, et al. Efficacy of inactivated split‐virus influenza vaccine against culture‐confirmed influenza in healthy adults: a prospective, randomized, placebo‐controlled trial. Journal of Infectious Diseases 2009;200:1861‐9. CENTRAL

aa Bridges 2000a {published data only}

Buxton Bridges C, Thompson VV, Meltzer MI, Reeve GR, Talamonti VJ, Cox NJ, et al. Effectiveness and cost benefit of influenza vaccination of healthy working adults, a randomized controlled trial. JAMA 2000;284(13):1655‐63. CENTRAL

aa Bridges 2000b {published data only}

Buxton Bridges C, Thompson VV, Meltzer MI, Reeve GR, Talamonti VJ, Cox NJ. Effectiveness and cost benefit of influenza vaccination of healthy working adults, a randomized controlled trial. JAMA 2000;284(13):1655‐63. CENTRAL

aa Eddy 1970 {published data only}

Eddy TS, Davies NA. The effect of vaccine on a closed epidemic of Hong Kong influenza. South African Medical Journal 1970;44(8):214‐6. CENTRAL

aa Edwards 1994a {published data only}

Edwards KM, Dupont WD, Westrich MK, Plummer WD, Palmer PS, Wright PF. A randomized controlled trial of cold adapted and inactivated vaccines for the prevention of influenza A disease. Journal of Infectious Diseases 1994;169(1):68‐76. CENTRAL

aa Edwards 1994b {published data only}

Edwards KM, Dupont WD, Westrich MK, Plummer WD, Palmer PS, Wright PF. A randomized controlled trial of cold adapted and inactivated vaccines for the prevention of influenza A disease. Journal of Infectious Diseases 1994;169(1):68‐76. CENTRAL

aa Edwards 1994c {published data only}

Edwards KM, Dupont WD, Westrich MK, Plummer WD, Palmer PS, Wright PF. A randomized controlled trial of cold adapted and inactivated vaccines for the prevention of influenza A disease. Journal of Infectious Diseases 1994;169(1):68‐76. CENTRAL

aa Edwards 1994d {published data only}

Edwards KM, Dupont WD, Westrich MK, Plummer WD, Palmer PS, Wright PF. A randomized controlled trial of cold adapted and inactivated vaccines for the prevention of influenza A disease. Journal of Infectious Diseases 1994;169(1):68‐76. CENTRAL

aa Frey 2010 {published data only}

Frey S, Vesikari T, Szymczakiewicz‐Multanowska A, Lattanzi M, Izu A, Groth N, et al. Clinical efficacy of cell culture‐derived and egg‐derived inactivated subunit influenza vaccines in healthy adults. Clinical Infectious Diseases 2010;51(9):997‐1004. CENTRAL

aa Hammond 1978 {published data only}

Hammond ML, Ferris AA, Faine S. Effective protection against influenza after vaccination with subunit vaccine. Medical Journal of Australia 1978;1(6):301‐3. CENTRAL

aa Jackson 2010a {published data only}

Jackson LA, Gaglani MJ, Keyserling HL, Balser J, Bouveret N, Fries L, et al. Safety, efficacy, and immunogenicity of an inactivated influenza vaccine in healthy adults: a randomised, placebo‐controlled trial over two influenza seasons. BMC Infectious Diseases 2010;10:71. CENTRAL

aa Jackson 2010b {published data only}

Jackson LA, Gaglani MJ, Keyserling HL, Balser J, Bouveret N, Fries L, et al. Safety, efficacy, and immunogenicity of an inactivated influenza vaccine in healthy adults: a randomised, placebo‐controlled trial over two influenza seasons. BMC Infectious Diseases 2010;10:71. CENTRAL

aa Keitel 1988a {published data only}

Keitel WA, Cate TR, Couch RB. Efficacy of sequential annual vaccination with inactivated influenza virus vaccine. American Journal of Epidemiology 1988;127(2):353‐64. CENTRAL

aa Keitel 1988b {published data only}

Keitel WA, Cate TR, Couch RB. Efficacy of sequential annual vaccination with inactivated influenza virus vaccine. American Journal of Epidemiology 1988;127(2):353‐64. CENTRAL

aa Keitel 1997a {published data only}

Keitel WA, Cate TR, Couch RB, Huggins LL, Hess KR. Efficacy of repeated annual immunization with inactivated influenza virus vaccines over a five year period. Vaccine 1997;15(10):1114‐22. CENTRAL

aa Keitel 1997b {published data only}

Keitel WA, Cate TR, Couch RB, Huggins LL, Hess KR. Efficacy of repeated annual immunization with inactivated influenza virus vaccines over a five year period. Vaccine 1997;15(10):1114‐22. CENTRAL

aa Keitel 1997c {published data only}

Keitel WA, Cate TR, Couch RB, Huggins LL, Hess KR. Efficacy of repeated annual immunization with inactivated influenza virus vaccines over a five year period. Vaccine 1997;15(10):1114‐22. CENTRAL

aa Langley 2011 {published data only}

Langley JM, Aoki F, Ward BJ, McGeer A, Angel JB, Stiver G, et al. A nasally administered trivalent inactivated influenza vaccine is well tolerated, stimulates both mucosal and systemic immunity, and potentially protects against influenza illness. Vaccine 2011;29(10):1921‐8. CENTRAL

aa Leibovitz 1971 {published data only}

Leibovitz A, Coultrip RL, Kilbourne ED, Legters LJ, Smith CD, Chin J, et al. Correlated studies of a recombinant influenza‐virus vaccine. IV. Protection against naturally occurring influenza in military trainees. Journal of Infectious Diseases 1971;124(5):481‐7. CENTRAL

aa Mcbride 2016a {published data only}

Mcbride WJ, Abhayaratna WP, Barr I, Booy R, Carapetis J, Carson S, et al. Efficacy of a trivalent influenza vaccine against seasonal strains and against 2009 pandemic H1N1: a randomized, placebo‐controlled trial. Vaccine 2016;34(41):4991‐7. CENTRAL

aa Mcbride 2016b {published data only}

Mcbride WJ, Abhayaratna WP, Barr I, Booy R, Carapetis J, Carson S, et al. Efficacy of a trivalent influenza vaccine against seasonal strains and against 2009 pandemic H1N1: a randomized, placebo‐controlled trial. Vaccine 2016;34(41):4991‐7. CENTRAL

aa Mesa Duque 2001 {published data only}

Mesa Duque SS, Moreno AP, Hurtado G, Arbelàaz Montoya MP. Effectiveness of an influenza vaccine in a working population in Colombia [Effectividad de una vacuna anti gripal en una poblaciòn laboral colombiana]. Pan American Journal of Public Health 2001;10(4):232‐9. CENTRAL

aa Mixéu 2002 {published data only}

Mixéu MA, Vespa GN, Forleo‐Neto E, Toniolo‐Neto J, Alves PM. Impact of influenza vaccination on civilian aircrew illness and absenteeism. Aviation, Space, and Environmental Medicine 2002;73(9):876‐80. CENTRAL

aa Mogabgab 1970a {published data only}

Mogabgab WJ, Leiderman E. Immunogenicity of 1967 polyvalent and 1968 Hong Kong influenza vaccines. JAMA 1970;211(10):1672‐6. CENTRAL

aa Mogabgab 1970b {published data only}

Mogabgab WJ, Leiderman E. Immunogenicity of 1967 polyvalent and 1968 Hong Kong influenza vaccines. JAMA 1970;211(10):1672‐6. CENTRAL

aa Monto 1982 {published data only}

Monto AS, DeWolfe Miller F, Maassab HF. Evaluation of an attenuated, cold recombinant influenza B virus vaccine. Journal of Infectious Diseases 1982;145(1):57‐64. CENTRAL

aa Monto 2009 {published data only}

Monto AS, Ohmit SE, Petrie JG, Johnson E, Truscon R, Teich E, et al. Comparative efficacy of inactivated and live attenuated influenza vaccines. New England Journal of Medicine 2009;361(13):1260‐7. CENTRAL

aa Nichol 1995 {published data only}

Nichol KL, Lind A, Margolis KL, Murdoch M, McFadden R, Hauge M. The effectiveness of vaccination against influenza in healthy, working adults. New England Journal of Medicine 1995;333(14):889‐93. CENTRAL

aa Nichol 1999a {published data only}

Nichol KL, Mendelman PM, Mallon KP, Jackson LA, Gorse GJ, Belshe RB, et al. Effectiveness of live attenuated intranasal influenza virus vaccine in healthy working adults, a randomized controlled trial. JAMA 1999;282(2):137‐44. CENTRAL

aa Ohmit 2006 {published data only (unpublished sought but not used)}

Ohmit SE, Victor JC, Rotthoff JR, Teich ER, Truscon RK, Baum LL, et al. Prevention of antigenically drifted influenza by inactivated and live attenuated vaccines. New England Journal of Medicine 2006;355(24):2513‐22. CENTRAL

aa Ohmit 2008 {published data only}

Ohmit SE, Victor JC, Teich ER, Truscon RK, Rotthoff JR, Newton DW, et al. Prevention of symptomatic seasonal influenza in 2005‐2006 by inactivated and live attenuated vaccines. Journal of Infectious Diseases 2008;198(3):312‐7. CENTRAL

aa Powers 1995a {published data only}

Powers DC, Smith GE, Anderson EL, Kennedy DJ, Hackett CS, Wilkinson BE, et al. Influenza A virus vaccines containing purified recombinant H3 hemagglutinin are well tolerated and induce protective immune responses in healthy adults. Journal of Infectious Diseases 1995;171(6):1595‐9. CENTRAL

aa Powers 1995b {published data only}

Powers DC, Smith GE, Anderson EL, Kennedy DJ, Hackett CS, Wilkinson BE, et al. Influenza A virus vaccines containing purified recombinant H3 hemagglutinin are well tolerated and induce protective immune responses in healthy adults. Journal of Infectious Diseases 1995;171(6):1595‐9. CENTRAL

aa Powers 1995c {published data only}

Powers DC, Smith GE, Anderson EL, Kennedy DJ, Hackett CS, Wilkinson BE, et al. Influenza A virus vaccines containing purified recombinant H3 hemagglutinin are well tolerated and induce protective immune responses in healthy adults. Journal of Infectious Diseases 1995;171:1595‐9. CENTRAL

aa Rytel 1977 {published data only}

Rytel MW, Jackson LJ, Niebojewski RA, Haagensen JL, Rosenkranz MA. Field trial of live attenuated influenza A/B ("Alice"/R‐75) vaccine. American Journal of Epidemiology 1977;105(1):49‐55. CENTRAL

aa Sumarokow 1971 {published data only}

Sumarokow AA, Popov VF, Nefedova LA, Salmin LV, Lazorenko NF. A study of live influenza vaccines in a controlled trial. Zhumal Mikrobiologii Epidemiologii Immunobiologii 1971;48(2):46‐52. CENTRAL

aa Tannock 1984 {published data only}

Tannock GA, Bryce DA, Hensley MJ, Saunders NA, Gillett RS, Kennedy WS. Responses to one or two doses of a deoxycholate subunit influenza vaccine in a primed population. Vaccine 1984;2(1):100‐5. CENTRAL

aa Treanor 2011 {published data only}

Treanor JJ, El Sahly H, King J, Graham I, Izikson R, Kohberger R, et al. Protective efficacy of a trivalent recombinant hemagglutinin protein vaccine (FluBlok) against influenza in healthy adults: a randomised, placebo‐controlled trial. Vaccine 2011;29(44):7733‐9. CENTRAL

aa Waldman 1969a {published data only}

Waldman RH, Bond JO, Levitt LP, Hartwig EC, Prather EC, Baratta RL, et al. An evaluation of influenza immunization. Bulletin of the World Health Organization 1969;41(3):543‐8. CENTRAL

aa Waldman 1969b {published data only}

Waldman RH, Bond JO, Levitt LP, Hartwig EC, Prather EC, Baratta RL, et al. An evaluation of influenza immunization. Bulletin of the World Health Organization 1969;41(3):543‐8. CENTRAL

aa Waldman 1969c {published data only}

Waldman RH, Bond JO, Levitt LP, Hartwig EC, Prather EC, Baratta RL, et al. An evaluation of influenza immunization. Bulletin of the World Health Organization 1969;41(3):543‐8. CENTRAL

aa Waldman 1969d {published data only}

Waldman RH, Bond JO, Levitt LP, Hartwig EC, Prather EC, Baratta RL, et al. An evaluation of influenza immunization. Bulletin of the World Health Organization 1969;41(3):543‐8. CENTRAL

aa Waldman 1972a {published data only}

Waldman RH, Coggins WJ. Influenza immunization: field trial on a university campus. Journal of Infectious Diseases 1972;126(3):242‐8. CENTRAL

aa Waldman 1972b {published data only}

Waldman RH, Coggins WJ. Influenza immunization: field trial on a university campus. Journal of Infectious Diseases 1972;126(3):242‐8. CENTRAL

aa Waldman 1972c {published data only}

Waldman RH, Coggins WJ. Influenza immunization: field trial on a university campus. Journal of Infectious Diseases 1972;126(3):242‐8. CENTRAL

aa Waldman 1972d {published data only}

Waldman RH, Coggins WJ. Influenza immunization: field trial on a university campus. Journal of Infectious Diseases 1972;126(3):242‐8. CENTRAL

aa Weingarten 1988 {published data only}

Weingarten S, Staniloff H, Ault M, Miles P, Bamberger M, Meyer RD. Do hospital employees benefit from the influenza vaccine?. Journal of General Internal Medicine 1988;3(1):32‐7. CENTRAL

aa Zhilova 1986a {published data only}

Zhilova GP, Ignat'eva GS, Orlov VA, Malikova EV, Maksakova VL. Results of a study of the effectiveness of simultaneous immunization against influenza with live and inactivated vaccines (1980 ‐ 1983). Voprosy Virusologii 1986;31(1):40‐4. CENTRAL

aa Zhilova 1986b {published data only}

Zhilova GP, Ignat'eva GS, Orlov VA, Malikova EV, Maksakova VL. Results of a study of the effectiveness of simultaneous immunization against influenza with live and inactivated vaccines (1980 ‐ 1983). Voprosy Virusologii 1986;31(1):40‐4. CENTRAL

ab Atmar 1990 {published data only}

Atmar RL, Bloom K, Keitel W, Couch RB, Greenberg SB. Effect of live attenuated, cold recombinant (CR) influenza virus vaccines on pulmonary function in healthy and asthmatic adults. Vaccine 1990;8(3):217‐24. CENTRAL

ab Betts 1977a {published data only}

Betts RF, Douglas RG, Roth FK, Little JW. Efficacy of live attenuated influenza A/Scotland/74 (H3N2) virus vaccine against challenge with influenza A/Victoria/3/75 (H3N2) virus. Journal of Infectious Diseases 1977;136(6):746‐53. CENTRAL

ab Boyce 2000 {published data only}

Boyce TG, Hsu HH, Sannella EC, Coleman‐Dockery SD, Baylis E, Zhu Y, et al. Safety and immunogenicity of adjuvanted and unadjuvanted subunit influenza vaccines administered intranasally to healthy adults. Vaccine 2000;19(2‐3):217‐26. CENTRAL

ab Caplan 1977 {published data only}

Caplan ES, Hughes TP, O'Donnel S, Levine MM, Hornick RB. Reactogenicity and immunogenicity of parenteral monovalent influenza A/Victoria/3/75 (H3N2) virus vaccine in healthy adults. Journal of Infectious Diseases 1977;136(Suppl):484‐90. CENTRAL

ab El'shina 1996 {published data only}

El'shina GA, Masalin IM, Shervali VI, Gorbunov MA, Lonskaia NI, Agafonova LV, et al. The trivalent polymer‐subunit influenza vaccine Grippol studied in a controlled epidemiological trial. Voenno‐Meditsinskii Zhurnal 1996;317(8):57‐60. CENTRAL

ab Evans 1976 {published data only}

Evans AE, Letley E, Ferris RD, Freestone DS. WRL 105 strain live attenuated influenza vaccine; comparison of one and two dose schedules. Journal of Hygiene 1976;77(3):327‐32. CENTRAL

ab Forsyth 1967 {published data only}

Forsyth JR. An assessment of oil adjuvant and aqueous influenza vaccines. I. Reactions to the vaccines. Journal of Hygiene 1967;65(4):485‐95. CENTRAL

ab Goodeve 1983 {published data only}

Goodeve A, Potter CW, Clark A, Jennings R, Schild GC, Yetts R. A graded‐dose study of inactivated, surface antigen influenza B vaccine in volunteers: reactogenicity, antibody response and protection to challenge virus infection. Journal of Hygiene 1983;90(1):107‐15. CENTRAL

ab Hrabar 1977 {published data only}

Hrabar A, Vodopija I, Andre FE, Mitchell JR, Maassab HF, Hennessy AV, et al. A placebo‐controlled dose‐response study of the reactogenicity and immunogenicity of a cold‐adapted recombinant A/Victoria/3/75 (H3N2) live influenza virus candidate vaccine in healthy volunteers. Developments in Biological Standardization 1977;39:53‐60. CENTRAL

ab Keitel 1993a {published data only}

Keitel WA, Couch RB, Quarles JM, Cate TR, Baxter B, Maassab HF. Trivalent attenuated cold‐adapted influenza virus vaccine: reduced viral shedding and serum antibody responses in susceptible adults. Journal of Infectious Diseases 1993;167(2):305‐11. CENTRAL

ab Keitel 1993b {published data only}

Keitel WA, Couch RB, Quarles JM, Cate TR, Baxter B, Maassab HF. Trivalent attenuated cold‐adapted influenza virus vaccine: reduced viral shedding and serum antibody responses in susceptible adults. Journal of Infectious Diseases 1993;167(2):305‐11. CENTRAL

ab Langley 2005 {published data only}

Langley JM, Halperin SA, McNeil S, Smith B, Jones T, Burt D, et al. Safety and immunogenicity of a Proteosome‐trivalent inactivated influenza vaccine, given nasally to healthy adults. Vaccine 2005;24(10):1601‐8. CENTRAL

ab Lauteria 1974 {published data only}

Lauteria SF, Kantzler GB, High PC, Lee JD, Waldman RH. An attenuated influenza virus vaccine: reactogenicity, transmissibility, immunogenicity, and protective efficacy. Journal of Infectious Diseases 1974;130(4):380‐3. CENTRAL

ab Miller 1977 {published data only}

Miller LW, Togo Y, Hornick RB. Clinical and serologic effects of live attenuated serum inhibitor‐resistant influenza B vaccine in seronegative adults. Journal of Medical Virology 1977;1(3):193‐9. CENTRAL

ab Pyrhönen 1981 {published data only}

Pyrhönen S, Suni J, Romo M. Clinical trial of a subunit influenza vaccine. Scandinavian Journal of Infectious Diseases 1981;13:95‐9. CENTRAL

ab Reeve 1982 {published data only}

Reeve P, Pibermann M, Bachmayer H, Liehl E, Moritz A, Ganzinger U, et al. Studies in man with a cold‐recombinant live influenza B virus vaccine. Journal of Medical Virology 1982;9(1):1‐9. CENTRAL

ab Rocchi 1979a {published data only}

Rocchi G, Ragona G, Piga C, Pelosio A, Volpi A, Vella S, et al. Influenza vaccination with live‐attenuated and inactivated virus‐vaccines during an outbreak of disease. Journal of Hygiene 1979;83(3):383‐90. CENTRAL

ab Rocchi 1979b {published data only}

Rocchi G, Ragona G, Piga C, Pelosio A, Volpi A, Vella S, et al. Influenza vaccination with live‐attenuated and inactivated virus‐vaccines during an outbreak of disease. Journal of Hygiene 1979;83(3):383‐90. CENTRAL

ab Saxen 1999 {published data only}

Saxen H, Virtanen M. Randomized, placebo‐controlled double blind study on the efficacy of influenza immunization on absenteeism of health care workers. Pediatric Infectious Disease Journal 1999;18(9):779‐83. CENTRAL

ab Scheifele 2003 {published data only}

Scheifele DW, Duval B, Russell ML, Warrington R, DeSerres G, Skowronski DM, et al. Ocular and respiratory symptoms attributable to inactivated split influenza vaccine: evidence from a controlled trial involving adults. Clinical Infectious Diseases 2003;36(7):850‐7. CENTRAL

ab Spencer 1977 {published data only}

Spencer MJ, Cherry JD, Powell KR. Clinical trial with "R‐75" strain live, attenuated, serum inhibitor‐resistant intranasal influenza B vaccine. Journal of Clinical Microbiology 1977;5(6):584‐7. CENTRAL

bb Dauvilliers 2013 {published data only}

Dauvilliers Y, Arnulf I, Lecendreux M, Monaca Charley C, Franco P, Drouot X, et al. Increased risk of narcolepsy in children and adults after pandemic H1N1 vaccination in France. Brain 2013;139(Pt 8):2486‐96. CENTRAL

bb DeStefano 2003 {published data only}

DeStefano F, Verstraeten T, Jackson LA, Okoro CA, Benson P, Black SB, et al. Vaccinations and risk of central nervous system demyelinating diseases in adults. Archives of Neurology 2003;60(4):504‐9. CENTRAL

bb Dieleman 2011a {published data only}

Dieleman J, Romio S, Johansen K, Weibel D, Bonhoeffer J, Sturkenboom M, et al. Guillain‐Barré syndrome and adjuvanted pandemic influenza A (H1N1) 2009 vaccine: multinational case‐control study in Europe. BMJ 2012;343:d3908. CENTRAL

bb Dieleman 2011b {published data only}

Dieleman J, Romio S, Johansen K, Weibel D, Bonhoeffer J, Sturkenboom M, et al. Guillain‐Barré syndrome and adjuvanted pandemic influenza A (H1N1) 2009 vaccine: multinational case‐control study in Europe. BMJ 2012;343:d3908. CENTRAL

bb Dieleman 2011c {published data only}

Dieleman J, Romio S, Johansen K, Weibel D, Bonhoeffer J, Sturkenboom M, et al. Guillain‐Barré syndrome and adjuvanted pandemic influenza A (H1N1) 2009 vaccine: multinational case‐control study in Europe. BMJ 2012;343:d3908. CENTRAL

bb Dieleman 2011d {published data only}

Dieleman J, Romio S, Johansen K, Weibel D, Bonhoeffer J, Sturkenboom M, et al. Guillain‐Barré syndrome and adjuvanted pandemic influenza A (H1N1) 2009 vaccine: multinational case‐control study in Europe. BMJ 2012;343:d3908. CENTRAL

bb Dieleman 2011e {published data only}

Dieleman J, Romio S, Johansen K, Weibel D, Bonhoeffer J, Sturkenboom M, et al. Guillain‐Barré syndrome and adjuvanted pandemic influenza A (H1N1) 2009 vaccine: multinational case‐control study in Europe. BMJ 2012;343:d3908. CENTRAL

bb Galeotti 2013 {published data only}

Galeotti F, Massari M, D'Alessandro R, Beghi E, Chiò A, Logroscino G, et al. Risk of Guillain‐Barré syndrome after 2010‐2011 influenza vaccination. European Journal of Epidemiology 2013;28(5):433‐44. CENTRAL

bb Garbe 2012 {published data only}

Garbe E, Andersohn F, Bronder E, Klimpel A, Thomae M, Kurtal H. Association between drug use and acute immune thrombocytopenia in adults: results from the Berlin case‐control surveillance study. Pharmacoepidemiology and Drug Safety 2011;20:S147. CENTRAL
Garbe E, Andersohn F, Bronder E, Salama A, Klimpel A, Thomae M, et al. Drug‐induced immune thrombocytopaenia: results from the Berlin Case‐Control Surveillance Study. European Journal of Clinical Pharmacology 2012;68(5):821‐32. CENTRAL

bb Grimaldi‐Bensouda 2011 {published data only}

Grimaldi‐Bensouda L, Alpérovitch A, Besson G, Vial C, Cuisset JM, Papeix C, et al. Guillain‐Barre syndrome, influenzalike illnesses, and influenza vaccination during seasons with and without circulating A/H1N1 viruses. American Journal of Epidemiology 2011;174(3):326‐35. CENTRAL

bb Grimaldi‐Bensouda 2012 {published data only}

Grimaldi‐Bensouda L, Michel M, Aubrun E, Leighton P, Viallard JF, Adoue D, et al. A case‐control study to assess the risk of immune thrombocytopenia associated with vaccines. Blood 2012;120(25):4938‐44. CENTRAL
Grimaldi‐Bensouda L, Michel M, Viallard J‐F, Adoue D, Magy‐Bertrand N, Khellaf M, et al. A multicenter case‐control prospective study to assess the risk of immune thrombocytopenia (ITP) associated with vaccines in adults using the PGRx‐ITP registry. Blood 2011;118(21):1169. CENTRAL

bb Hernan 2004 {published data only}

Hernan MA, Jick SS, Olek MJ, Jick H. Recombinant hepatitis B vaccine and the risk of multiple sclerosis: a prospective study. Neurology 2004;63(5):838‐42. CENTRAL

bb MacIntyre 2013 {published data only}

MacIntyre CR, Heywood AE, Kovoor P, Ridda I, Seale H, Tan T, et al. Ischaemic heart disease, influenza and influenza vaccination: a prospective case control study. Heart 2013;99(24):1843‐8. CENTRAL

bb Mastrangelo 2000 {published data only}

Mastrangelo G, Rossi CR, Pfahlberg A, Marzia V, Barba A, Baldo M, et al. Is there a relationship between influenza vaccinations and risk of melanoma? A population‐based case‐control study. European Journal of Epidemiology 2000;16(9):777‐82. CENTRAL

bb Mutsch 2004 {published data only}

Mutsch M, Zhou W, Rhodes P, Bopp M, Chen RT, Linder T, et al. Use of the inactivated intranasal influenza vaccine and the risk of Bell's palsy in Switzerland. New England Journal of Medicine 2004;350(9):896‐903. CENTRAL

bb Payne 2006 {published data only}

Payne DC, Rose CE, Kerrison J, Aranas A, Duderstadt S, McNeil MM. Anthrax vaccination and risk of optic neuritis in the United States military, 1998‐2003. Archives of Neurology 2006;63(6):871‐5. CENTRAL

bb Ray 2011 {published data only}

Ray P, Black S, Shinefield H, Dillon A, Carpenter D, Lewis E, et al. Vaccine Safety Datalink Team. Risk of rheumatoid arthritis following vaccination with tetanus, influenza and hepatitis B vaccines among persons 15‐59 years of age. Vaccine 2011;29(38):6592‐7. CENTRAL

bb Rouleau 2014 {published data only}

Rouleau I, De Serres G, Skowronski DM, Drolet JP, Lemire C, Toth E, et al. Risk factors associated with anaphylaxis and other allergic‐like events following receipt of 2009 monovalent AS03‐adjuvanted pandemic influenza vaccine in Quebec, Canada. Vaccine 2014;32(28):3480‐7. CENTRAL

bb Siscovick 2000 {published data only}

Siscovick DS, Raghunathan TE, Lin D, Weinmann S, Arbogast P, Lemaitre RN, et al. Influenza vaccination and the risk of primary cardiac arrest. American Journal of Epidemiology 2000;152(7):674‐7. CENTRAL

bb Zorzon 2003 {published data only}

Zorzon M, Zivadinov R, Nasuelli D, Dolfini P, Bosco A, Bratina A, et al. Risk factors of multiple sclerosis: a case‐control study. Neurological Sciences 2003;24(4):242‐7. CENTRAL

cb Bardage 2011 {published data only}

Bardage C, Persson I, Ortqvist A, Bergman U, Ludvigsson JF, Granath F. Neurological and autoimmune disorders after vaccination against pandemic influenza A (H1N1) with a monovalent adjuvanted vaccine: population based cohort study in Stockholm, Sweden. BMJ 2011;343:d5956. CENTRAL

cb Baxter 2012 {published data only}

Baxter R, Toback SL, Sifakis F, Hansen J, Bartlett J, Aukes L, et al. A postmarketing evaluation of the safety of Ann Arbor strain live attenuated influenza vaccine in adults 18‐49 years of age. Vaccine 2012;30(20):3053‐60. CENTRAL

cb Kaplan 1982 {published data only}

Kaplan JE, Katona P, Hurwitz ES, Schonberger LB. Guillain‐Barre syndrome in the United States, 1979‐1980 and 1980‐1981. Lack of an association with influenza vaccination. JAMA 1982;248(6):698‐700. CENTRAL

cb Lasky 1998 {published data only}

Lasky T, Terracciano GJ, Magder L, Koski CL, Ballesteros M, Nash D, et al. The Guillain‐Barre syndrome and the 1992‐1993 and 1993‐1994 influenza vaccines. New England Journal of Medicine 1998;339(25):1797‐802. CENTRAL

cb Moro 2013 {published data only}

Moro ML, Nobilio L, Voci C, Di Mario S, Candela S, Magrini N. A population based cohort study to assess the safety of pandemic influenza vaccine Focetria in Emilia‐Romagna region, Italy ‐ part two. Vaccine 2013;31(10):1438‐46. CENTRAL

cb O'Flanagan 2014 {published data only}

O'Flanagan D, Barret AS, Foley M, Cotter S, Bonner C, Crowe C, et al. Investigation of an association between onset of narcolepsy and vaccination with pandemic influenza vaccine, Ireland April 2009‐December 2010. Eurosurveillance 2014;19(17):15‐25. CENTRAL

cb Persson 2014 {published data only}

Persson I, Granath F, Askling J, Ludvigsson JF, Olsson T, Feltelius N. Risks of neurological and immune‐related diseases, including narcolepsy, after vaccination with Pandemrix: a population‐ and registry‐based cohort study with over 2 years of follow‐up. Journal of Internal Medicine 2014;275(2):172‐90. CENTRAL

cb Ray 2011 {published data only}

Ray P, Black S, Shinefield H, Dillon A, Carpenter D, Lewis E, et al. Risk of rheumatoid arthritis following vaccination with tetanus, influenza and hepatitis B vaccines among persons 15‐59 years of age. Vaccine 2011;29(38):6592‐7. CENTRAL

cb Shonberger 1979 {published data only}

Schonberger LB, Bregman DJ, Sullivan‐Bolyai JZ, Keenlyside RA, Ziegler DW, Retailliau HF, et al. Guillain‐Barre syndrome following vaccination in the National Influenza Immunization Program, United States, 1976 ‐ 1977. American Journal of Epidemiology 1979;110(2):105‐23. CENTRAL

paa Ma 2014 {published data only}

Ma F, Zhang L, Jiang R, Zhang J, Wang H, Gao X, et al. Prospective cohort study of the safety of an influenza A (H1N1) vaccine in pregnant Chinese women. Clinical and Vaccine Immunology 2014;21(9):1282‐7. CENTRAL

paa Madhi 2014 {published data only}

Madhi SA, Cutland CL, Kuwanda L, Weinberg A, Hugo A, Jones S, et al. Influenza vaccination of pregnant women and protection of their infants. New England Journal of Medicine 2014;371(10):918‐31. CENTRAL

pba Benowitz 2010 {published data only}

Benowitz I, Esposito DB, Gracey KD, Shapiro ED, Vázquez M. Influenza vaccine given to pregnant women reduces hospitalization due to influenza in their infants. Clinical Infectious Diseases 2010;51(12):1355‐61. CENTRAL

pba Poehling 2011 {published data only}

Poehling KA, Szilagyi PG, Staat MA, Snively BM, Payne DC, Bridges CB, et al. Impact of maternal immunization on influenza hospitalizations in infants. American Journal of Obstetrics and Gynecology 2001;204(Suppl 6):141‐8. CENTRAL

pbb Irving 2013 {published data only}

Irving SA, Kieke BA, Donahue JG, Mascola MA, Baggs J, Destefano F, et al. Trivalent inactivated influenza vaccine and spontaneous abortion. Obstetrics and Gynecology 2013;121(1):159‐65. CENTRAL

pca Ahrens 2014 {published data only}

Ahrens KA, Louik C, Kerr S, Mitchell AA, Werler MM. Seasonal influenza vaccination during pregnancy and the risks of preterm delivery and small for gestational age birth. Paediatric and Perinatal Epidemiology 2014;28(6):498‐509. CENTRAL

pca Black 2004 {published data only}

Black SB, Shinefield HR, France EK, Fireman BH, Blatt ST, Shay D. Effectiveness of influenza vaccine during pregnancy in preventing hospitalizations and outpatient visits for respiratory illness in pregnant women and their infants. American Journal of Perinatology 2004;21(6):333‐9. CENTRAL

pca Eick 2011 {published data only}

Eick AA, Uyeki TM, Klimov A, Hall H, Reid R, Santosham M, et al. Maternal influenza vaccination and effect on influenza virus infection in young infants. Archives of Pediatrics and Adolescent Medicine 2011;165(2):104‐11. CENTRAL

pca France 2006 {published data only}

France EK, Smith‐Ray R, McClure D, Hambidge S, Xu S, Yamasaki K, et al. Impact of maternal influenza vaccination during pregnancy on the incidence of acute respiratory illness visits among infants. Archives of Pediatrics and Adolescent Medicine 2006;160(12):1277‐83. CENTRAL

pca Hulka 1964 {published data only}

Hulka JF. Effectiveness of polyvalent influenza vaccine in pregnancy. Report of a controlled study during an outbreak of Asian influenza. Obstetrics and Gynecology 1964;23:830‐7. CENTRAL

pca Munoz 2005 {published data only}

Munoz FM, Greisinger AJ, Wehmanen OA, Mouzoon ME, Hoyle JC, Smith FA, et al. Safety of influenza vaccination during pregnancy. American Journal of Obstetrics and Gynecology 2005;192(4):1098‐106. CENTRAL

pca Yamada 2012 {published data only}

Yamada T, Yamada T, Morikawa M, Cho K, Endo T, Sato SS, et al. Pandemic (H1N1) 2009 in pregnant Japanese women in Hokkaido. Journal of Obstetrics and Gynaecology Research 2012;38(1):130‐6. CENTRAL

pcb Beau 2014 {published data only}

Beau AB, Hurault‐Delarue C, Vidal S, Guitard C, Vayssiere C, Petiot D. Pandemic A/H1N1 influenza vaccination during pregnancy: a comparative study using the EFEMERIS database. Vaccine 2014;32(11):1254‐8. CENTRAL

pcb Cantu 2013 {published data only}

Cantu J, Biggio J, Jauk V, Wetta L, Andrews W, Tita A. Selective uptake of influenza vaccine and pregnancy outcomes. Journal of Maternal‐Fetal and Neonatal Medicine 2013;26(12):1207‐11. CENTRAL

pcb Chambers 2013 {published data only}

Chambers CD, Johnson D, Xu R, Luo Y, Louik C, Mitchell AA, et al. Risks and safety of pandemic H1N1 influenza vaccine in pregnancy: birth defects, spontaneous abortion, preterm delivery, and small for gestational age infants. Vaccine 2013;31(44):5026‐32. CENTRAL

pcb Cleary 2014 {published data only}

Cleary BJ, Rice U, Eogan M, Metwally N, McAuliffe F. 2009 A/H1N1 influenza vaccination in pregnancy: uptake and pregnancy outcomes ‐ a historical cohort study. European Journal of Obstetrics & Gynecology and Reproductive Biology 2014;178:163‐8. CENTRAL

pcb Deinard 1981 {published data only}

Deinard AS, Ogburn P. A/NJ/8/76 influenza vaccination program: effects on maternal health and pregnancy outcome. American Journal of Obstetrics and Gynecology 1981;140(3):240‐5. CENTRAL

pcb Dodds 2012 {published data only}

Dodds L, Macdonald N, Scott J, Spencer A, Allen VM, McNeil S. The association between influenza vaccine in pregnancy and adverse neonatal outcomes. Journal of Obstetrics and Gynaecology Canada 2012;34(8):714‐20. CENTRAL

pcb Fell 2012 {published data only}

Fell DB, Sprague AE, Liu N, Yasseen AS, Wen SW, Smith G, et al. Better Outcomes Registry & Network (BORN) Ontario. H1N1 influenza vaccination during pregnancy and fetal and neonatal outcomes. American Journal of Public Health 2012;102(6):e33‐40. CENTRAL

pcb Håberg 2013 {published data only}

Håberg SE, Trogstad L, Gunnes N, Wilcox AJ, Gjessing HK, Samuelsen SO, et al. Risk of fetal death after pandemic influenza virus infection or vaccination. New England Journal of Medicine 2013;368(4):333‐40. CENTRAL
Håberg SE, Trogstad L, Gunnes N, Wilcox AJ, Gjessing HK, Samuelsen SO, et al. Risk of fetal death after pandemic influenza virus infection or vaccination. Obstetrical and Gynecological Survey 2013;68(5):348‐9. CENTRAL

pcb Heikkinen 2012 {published data only}

Heikkinen T, Young J, van Beek E, Franke H, Verstraeten T, Weil JG, et al. Safety of MF59‐adjuvanted A/H1N1 influenza vaccine in pregnancy: a comparative cohort study. American Journal of Obstetrics & Gynecology 2012;207(3):177.e1‐8. CENTRAL

pcb Källén 2012 {published data only}

Källén B, Olausson PO. Vaccination against H1N1 influenza with Pandemrix during pregnancy and delivery outcome: a Swedish register study. British Journal of Obstetrics and Gynaecology 2012;119(13):1583‐90. CENTRAL

pcb Launay 2012 {published data only}

Launay O, Krivine A, Charlier C, Truster V, Tsatsaris V, Lepercq J, et al. Low rate of pandemic A/H1N1 2009 influenza infection and lack of severe complication of vaccination in pregnant women: a prospective cohort study. PLoS ONE 2012;7(12):e52303. CENTRAL

pcb Lin 2012 {published data only}

Lin TH, Lin SY, Lin CH, Lin RI, Lin HC, Chiu TH, et al. AdimFlu‐S influenza A (H1N1) vaccine during pregnancy: the Taiwanese Pharmacovigilance Survey. Vaccine 2012;30(16):2671‐5. CENTRAL

pcb Louik 2013 {published data only}

Louik C, Ahrens K, Kerr S, Pyo J, Chambers C, Jones KL, et al. Risks and safety of pandemic H1N1 influenza vaccine in pregnancy: exposure prevalence, preterm delivery, and specific birth defects. Vaccine 2013;31(44):5033‐40. CENTRAL

pcb Ludvigsson 2013 {published data only}

Ludvigsson JF, Zugna D, Cnattingius S, Richiardi L, Ekbom A, Ortqvist A, et al. Influenza H1N1 vaccination and adverse pregnancy outcome. European Journal of Epidemiology 2013;28(7):579‐88. CENTRAL

pcb Nordin 2013 {published data only}

Nordin JD, Kharbanda EO, Benitez GV, Nichol K, Lipkind H, Naleway A, et al. Maternal safety of trivalent inactivated influenza vaccine in pregnant women. Obstetrics and Gynecology 2013;121(3):519‐25. CENTRAL

pcb Nordin 2014 {published data only}

Nordin JD, Kharbanda EO, Vazquez Benitez G, Lipkind H, Vellozzi C, Destefano F. Maternal influenza vaccine and risks for preterm or small for gestational age birth. Journal of Pediatrics 2014;164(5):1051‐7.e2. CENTRAL

pcb Omer 2011 {published data only}

Omer SB, Goodman D, Steinhoff MC, Rochat R, Klugman KP, Stoll BJ, et al. Maternal influenza immunization and reduced likelihood of prematurity and small for gestational age births: a retrospective cohort study. PLoS Medicine 2011;8(5):e1000441. CENTRAL

pcb Oppermann 2012 {published data only}

Oppermann M, Fritzsche J, Weber‐Schoendorfer C, Keller‐Stanislawski B, Allignol A, Meister R, et al. A(H1N1)v2009: a controlled observational prospective cohort study on vaccine safety in pregnancy. Vaccine 2012;30(30):4445‐52. CENTRAL

pcb Pasternak 2012 {published data only}

Pasternak B, Svanström H, Mølgaard‐Nielsen D, Krause TG, Emborg HD, Melbye M, et al. Risk of adverse fetal outcomes following administration of a pandemic influenza A (H1N1) vaccine during pregnancy. JAMA 2012;308(2):165‐74. CENTRAL

pcb Richards 2013 {published data only}

Richards JL, Hansen C, Bredfeldt C, Bednarczyk RA, Steinhoff MC, Adjaye‐Gbewonyo D, et al. Neonatal outcomes after antenatal influenza immunization during the 2009 H1N1 influenza pandemic: impact on preterm birth, birth weight, and small for gestational age birth. Clinical Infectious Diseases 2013;56(9):1216‐22. CENTRAL

pcb Rubinstein 2013 {published data only}

Rubinstein F, Micone P, Bonotti A, Wainer V, Schwarcz A, Augustovski F. Influenza A/H1N1 MF59 adjuvanted vaccine in pregnant women and adverse perinatal outcomes: multicentre study. BMJ Online 2013;346(7896):f393. CENTRAL

pcb Sheffield 2012 {published data only}

Sheffield JS, Greer LG, Rogers VL, Roberts SW, Lytle H, McIntire DD, et al. Effect of influenza vaccination in the first trimester of pregnancy. Obstetrics and Gynecology 2012;120(3):532‐7. CENTRAL

pcb Toback 2012 {published data only}

Toback SL, Beigi R, Tennis P, Sifakis F, Calingaert B, Ambrose CS. Maternal outcomes among pregnant women receiving live attenuated influenza vaccine. Influenza and Other Respiratory Viruses 2012;6(1):44‐51. CENTRAL

pcb Trotta 2014 {published data only}

Trotta F, Da Cas R, Spila Alegiani S, Gramegna M, Venegoni M, Zocchetti C, et al. Evaluation of safety of A/H1N1 pandemic vaccination during pregnancy: cohort study. BMJ 2014;348:g3361. CENTRAL

ab López‐Macías 2011a {published data only}

López‐Macías C, Ferat‐Osorio E, Tenorio‐Calvo A, Isibasi A, Talavera J, Arteaga‐Ruiz O, et al. Safety and immunogenicity of a virus‐like particle pandemic influenza A (H1N1) 2009 vaccine in a blinded, randomised, placebo‐controlled trial of adults in Mexico. Vaccine 2011;29(44):7826‐34. CENTRAL

ab López‐Macías 2011b {published data only}

López‐Macías C, Ferat‐Osorio E, Tenorio‐Calvo A, Isibasi A, Talavera J, Arteaga‐Ruiz O, et al. Safety and immunogenicity of a virus‐like particle pandemic influenza A (H1N1) 2009 vaccine in a blinded, randomised, placebo‐controlled trial of adults in Mexico. Vaccine 2011;29(44):7826‐34. CENTRAL

ab Mallory 2010 {published data only}

Mallory RM, Malkin E, Ambrose CS, Bellamy T, Shi L, Yi T, et al. Safety and immunogenicity following administration of a live, attenuated monovalent 2009 H1N1 influenza vaccine to children and adults in two randomised controlled trials. PLoS ONE 2010;5(10):e13755. CENTRAL

ab Plennevaux 2010 {published data only}

Plennevaux E, Sheldon E, Blatter M, Reeves‐Hoché MK, Denis M. Immune response after a single vaccination against 2009 influenza A H1N1 in USA: a preliminary report of two randomised controlled phase 2 trials. Lancet 2010;375(9708):41‐8. CENTRAL

ab Precioso 2011 {published data only}

Precioso AR, Miraglia JL, Campos LM, Goulart AC, Timenetsky Mdo C, Cardoso MR, et al. A phase I randomised, double‐blind, controlled trial of 2009 influenza A (H1N1) inactivated monovalent vaccines with different adjuvant systems. Vaccine 2011;29(48):8974‐81. CENTRAL

ab Treanor 2010 {published data only}

Treanor JJ, Taylor DN, Tussey L, Hay C, Nolan C, Fitzgerald T, et al. Safety and immunogenicity of a recombinant hemagglutinin influenza‐flagellin fusion vaccine (VAX125) in healthy young adults. Vaccine 2010;28(52):8268‐74. CENTRAL

ab Turley 2011 {published data only}

Turley CB, Rupp RE, Johnson C, Taylor DN, Wolfson J, Tussey L, et al. Safety and immunogenicity of a recombinant M2e‐flagellin influenza vaccine (STF2.4xM2e) in healthy adults. Vaccine 2011;29(32):5145‐52. CENTRAL

ab Wacheck 2010 {published data only}

Wacheck V, Egorov A, Groiss F, Pfeiffer A, Fuereder T, Hoeflmayer D, et al. A novel type of influenza vaccine: safety and immunogenicity of replication‐deficient influenza virus created by deletion of the interferon antagonist NS1. Journal of Infectious Diseases 2010;201(3):354‐62. CENTRAL

Al‐Dabbagh 2013 {published data only}

Al‐Dabbagh M, Lapphra K, Scheifele DW, Halperin SA, Langley JM, Cho P, et al. Elevated inflammatory mediators in adults with oculo‐respiratory syndrome following influenza immunization: a public health agency of Canada/Canadian Institutes of Health Research Influenza Research Network (PCIRN) Study. Clinical and Vaccine Immunology 2013;20(8):1108‐14. CENTRAL

Ambrosch 1976 {published data only}

Ambrosch F, Balluch H. Studies of the non‐specific clinical effectiveness of influenza vaccination. Laryngologie, Rhinologie, Otologie 1976;55:57‐61. CENTRAL

Ambrose 2012 {published data only}

Ambrose CS, Wu X. The safety and effectiveness of self‐administration of intranasal live attenuated influenza vaccine in adults. Vaccine 2013;31(6):857‐60. CENTRAL

Andersson 2015 {published data only}

Andersson L. Response on the author's reply to the letter to the editor: Contradictory data on type 1 diabetes in a recently published article "Risks of neurological and immune‐related diseases, including narcolepsy, after vaccination with Pandemrix". Journal of Internal Medicine 2015;277(2):272‐3. CENTRAL

Aoki 1986 {published data only}

Aoki FY, Sitar DS, Milley EV, Hammond GW, Milley EV, Vermeersch C, et al. Potential of influenza vaccine and amantadine to prevent influenza A illness in Canadian forces personnel 1980‐83. Military Medicine 1986;151(9):459‐65. CENTRAL

Arnou 2010 {published data only}

Arnou R, Eavis P, Pardo JR, Ambrozaitis A, Kazek MP, Weber F. Immunogenicity, large scale safety and lot consistency of an intradermal influenza vaccine in adults aged 18‐60 years: randomized, controlled, phase III trial. Human Vaccines 2010;6(4):346‐54. CENTRAL

Atmar 1995 {published data only}

Atmar RL, Keitel WA, Cate TR, Quarles JM, Couch RB. Comparison of trivalent cold‐adapted recombinant (CR) influenza virus vaccine with monovalent CR vaccines in healthy unselected adults. Journal of Infectious Diseases 1995;172(1):253‐7. CENTRAL

Atmar 2011 {published data only}

Atmar RL, Keitel WA, Quarles JM, Cate TR, Patel SM, Nino D, et al. Evaluation of age‐related differences in the immunogenicity of a G9 H9N2 influenza vaccine. Vaccine 2011;29(45):8066‐72. CENTRAL

Atsmon 2012 {published data only}

Atsmon J, Kate‐Ilovitz E, Shaikevich D, Singer Y, Volokhov I, Haim KY, et al. Safety and immunogenicity of multimeric‐001 ‐ a novel universal influenza vaccine. Journal of Clinical Immunology 2012;32(3):595‐603. CENTRAL

Ausseil 1999 {published data only}

Ausseil F. Immunization against influenza among working adults: the Philippines experience. Vaccine 1999;17(Suppl 1):59‐62. CENTRAL

Banzhoff 2001 {published data only}

Banzhoff A, Kaniok W, Muszer A. Effectiveness of an influenza vaccine used in Poland in the 1998‐1999 influenza season. Immunological Investigations 2001;30(2):103‐13. CENTRAL

Baxter 2010 {published data only}

Baxter R, Ray GT, Fireman BH. Effect of influenza vaccination on hospitalizations in persons aged 50 years and older. Vaccine 2010;28(45):7267‐72. CENTRAL

Baxter 2011 {published data only}

Baxter R, Patriarca PA, Ensor K, Izikson R, Goldenthal KL, Cox MM. Evaluation of the safety, reactogenicity and immunogenicity of FluBlok trivalent recombinant baculovirus‐expressed hemagglutinin influenza vaccine administered intramuscularly to healthy adults 50‐64 years of age. Vaccine 2011;29(12):2272‐8. CENTRAL

Baxter 2012 {published data only}

Baxter R, Lewis N, Bakshi N, Vellozzi C, Klein NP. Recurrent Guillain‐Barre syndrome following vaccination. Clinical Infectious Diseases 2012;54(6):800‐4. CENTRAL

Baxter 2013 {published data only}

Baxter R, Bakshi N, Fireman B, Lewis E, Ray P, Vellozzi C, et al. Lack of association of Guillain‐Barre syndrome with vaccinations. Clinical Infectious Diseases 2013;57(2):197‐204. CENTRAL

Belongia 2009 {published data only}

Belongia EA, Kieke BA, Donahue JG, Greenlee RT, Balish A, Foust A, et al. Effectiveness of inactivated influenza vaccines varied substantially with antigenic match from the 2004‐2005 season to the 2006‐2007 season. Journal of Infectious Diseases 2009;199(2):159‐67. CENTRAL

Belshe 2001 {published data only}

Belshe RB, Gruber WC. Safety, efficacy and effectiveness of cold‐adapted, live, attenuated, trivalent, intranasal influenza vaccine in adults and children. Philosophical Transactions of the Royal Society of London 2001;356(1416):1947‐51. CENTRAL

Benke 2004 {published data only}

Benke G, Abramson M, Raven J, Thien FCK, Walters EH. Asthma and vaccination history in a young adult cohort. Australian and New Zealand Journal of Public Health 2004;28(4):336‐8. CENTRAL

Beran 2013 {published data only}

Beran JI, Peeters M, Dewe W, Raupachova J, Hobzova L, Devaster JM. Immunogenicity and safety of quadrivalent versus trivalent inactivated influenza vaccine: a randomised, controlled trial in adults. BMC Infectious Diseases 2013;13(1):224. CENTRAL

Betts 1977b {published data only}

Betts RF, Douglas RG. Comparative study of reactogenicity and immunogenicity of influenza A/New Jersey/8/76 (Hsw1N1) virus vaccines in normal volunteers. Journal of Infectious Diseases 1977;136(Suppl):443‐9. CENTRAL

Beyer 1996 {published data only}

Beyer WEP, Palache AM, Kerstens R, Masurel N. Gender differences in local and systemic reactions to inactivated influenza vaccine, established by a meta‐analysis of fourteen independent studies. European Journal of Clinical Microbiology & Infectious Diseases 1996;15(1):65‐70. CENTRAL

Carlson 1979 {published data only}

Carlson AJ, Davidson WL, McLean AA, Vella PP, Weibel RE, Woodhour AF, et al. Pneumococcal vaccine: dose, revaccination, and coadministration with influenza vaccine. Proceedings of the Society for Experimental Biology and Medicine 1979;161(4):558‐63. CENTRAL

Cate 1977 {published data only}

Cate TR, Couch RB, Kasel JA, Six HR. Clinical trials of monovalent influenza A/New Jersey/76 virus vaccines in adults: reactogenicity, antibody response, and antibody persistence. Journal of Infectious Diseases 1977;136(Suppl):450‐5. CENTRAL

Chavant 2013 {published data only}

Chavant F, Ingrand I, Jonville‐Bera AP, Plazanet C, Gras‐Champel V, Lagarce L, et al. The PREGVAXGRIP study: a cohort study to assess foetal and neonatal consequences of in utero exposure to vaccination against A (H1N1) v2009 influenza. Drug Safety 2013;36(6):455‐65. CENTRAL

Chichester 2012 {published data only}

Chichester JA, Jones RM, Green BJ, Stow M, Miao F, Moonsammy G, et al. Safety and immunogenicity of a plant‐produced recombinant hemagglutinin‐based influenza vaccine (HAI‐05) derived from A/Indonesia/05/2005 (H5N1) influenza virus: a phase 1 randomised, double‐blind, placebo‐controlled, dose‐escalation study in healthy adults. Viruses 2012;4(11):3227‐44. CENTRAL

Chlibek 2002 {published data only}

Chlibek R, Beran J, Splino M. Effectiveness of influenza vaccination in healthy adults ‐ a fourfold decrease in influenza morbidity during one influenza season. Epidemiologie, Mikrobiologie, Imunologie 2002;51(2):47‐51. CENTRAL

Choe 2011a {published data only}

Choe YJ, Cho H, Song KM, Kim JH, Han OP, Kwon YH, et al. Active surveillance of adverse events following immunization against pandemic influenza A (H1N1) in Korea. Japanese Journal of Infectious Diseases 2011;64(4):297‐303. CENTRAL

Choe 2011b {published data only}

Choe YJ, Cho H, Bae GR, Lee JK. Guillain‐Barre syndrome following receipt of influenza A (H1N1) 2009 monovalent vaccine in Korea with an emphasis on Brighton Collaboration case definition. Vaccine 2011;29(11):2066‐70. CENTRAL

Choe 2011c {published data only}

Choe YJ, Cho H, Kim SN, Bae GR, Lee JK. Serious adverse events following receipt of trivalent inactivated influenza vaccine in Korea, 2003‐2010. Vaccine 2011;29(44):7727‐32. CENTRAL

Chou 2007 {published data only}

Chou CH, Liou WP, Hu KI, Loh CH, Chou CC, Chen YH. Bell’s palsy associated with influenza vaccination: two case reports. Vaccine 2007;25:2839–41. CENTRAL

Clover 1991 {published data only}

Clover RD, Crawford S, Glezen WP, Taber LH, Matson CC, Couch RB. Comparison of heterotypic protection against influenza A/Taiwan/86 (H1N1) by attenuated and inactivated vaccines to A/Chile/83‐like viruses. Journal of Infectious Diseases 1991;163(2):300‐4. CENTRAL

Confavreux 2001 {published data only}

Confavreux C, Suissa S, Saddier P, Bourdes V, Vukusic S. Vaccinations and the risk of relapse in multiple sclerosis. Vaccines in Multiple Sclerosis Study Group. New England Journal of Medicine 2001;344(5):319‐26. CENTRAL

Conlin 2013 {published data only}

Conlin AMS, Bukowinski AT, Sevick CJ, DeScisciolo C, Crum‐Cianflone NF. Safety of the pandemic H1N1 influenza vaccine among pregnant U.S. military women and their newborns. Obstetrics and Gynecology 2013;121(3):511‐8. CENTRAL

Couch 2012 {published data only}

Couch RB, Patel SM, Wade‐Bowers CL, Niño D. A randomised clinical trial of an inactivated avian influenza A (H7N7) vaccine. PLoS ONE 2012;7(12):e49704. CENTRAL

Das Gupta 2002 {published data only}

Das Gupta R, Guest JF. A model to estimate the cost benefit of an occupational vaccination programme for influenza with Influvac in the UK. Pharmacoeconomics 2002;20(7):475‐84. CENTRAL

Davidson 2011 {published data only}

Davidson LE, Fiorino AM, Snydman DR, Hibberd PL. Lactobacillus GG as an immune adjuvant for live‐attenuated influenza vaccine in healthy adults: a randomized double‐blind placebo‐controlled trial. European Journal of Clinical Nutrition 2011;65(4):501‐7. CENTRAL

Davies 1972 {published data only}

Davies JE, Howell RW, Meichen FW. A clinical trial of inhaled inactivated influenza vaccine. British Journal of Clinical Practice 1972;26(10):469‐71. CENTRAL

Davies 1973 {published data only}

Davies JE, Howell RH, Meichen FW. A clinical trial of inhaled inactivated influenza vaccine. British Journal of General Practice 1970;27(6):219‐21. CENTRAL

De Serres 2003a {published data only}

De Serres GI, Boulianne N, Duval B, Rochette L, Grenier JL, Roussel R, et al. Oculo‐respiratory syndrome following influenza vaccination: evidence for occurrence with more than one influenza vaccine. Vaccine 2003;21(19‐20):2346‐53. CENTRAL

De Serres 2003b {published data only}

De Serres GI, Grenier JL, Toth E, Menard S, Roussel R, Tremblay M, et al. The clinical spectrum of the oculo‐respiratory syndrome after influenza vaccination. Vaccine 2003;21(19‐20):2354‐61. CENTRAL

De Serres 2004 {published data only}

De Serres G, Skowronski DM, Guay M, Rochette L, Jacobsen K, Fuller T, et al. Recurrence risk of oculorespiratory syndrome after influenza vaccination: randomized controlled trial of previously affected persons. Archives of Internal Medicine 2004;164(20):2266‐72. CENTRAL

De Wals 2012 {published data only}

De Wals P, Deceuninck G, Toth E, Boulianne N, Brunet D, Boucher RM, et al. Risk of Guillain‐Barre syndrome following H1N1 influenza vaccination in Quebec. JAMA 2012;308(2):175‐81. CENTRAL

Dolin 1977 {published data only}

Dolin R, Wise TG, Mazur MH, Tuazon CU, Ennis FA. Immunogenicity and reactogenicity of influenza A/New Jersey/76 virus vaccines in normal adults. Journal of Infectious Diseases 1977;136(Suppl):435‐42. CENTRAL

Dominguez 2012 {published data only}

Dominguez A, Castilla J, Godoy P, Delgado‐Rodriguez M, Martin V, Saez M, et al. Effectiveness of pandemic and seasonal influenza vaccines in preventing pandemic influenza‐associated hospitalization. Vaccine 2012;30(38):5644‐50. CENTRAL

Duffy 2014 {published data only}

Duffy J, Weintraub E, Vellozzi C, DeStefano F. Narcolepsy and influenza A (H1N1) pandemic 2009 vaccination in the United States. Neurology 2014;83(20):1823‐30. CENTRAL

Eames 2012 {published data only}

Eames KT, Brooks‐Pollock E, Paolotti D, Perosa M, Gioannini C, Edmunds WJ. Rapid assessment of influenza vaccine effectiveness: analysis of an internet‐based cohort. Epidemiology and Infection 2012;140(7):1309‐15. CENTRAL

Edmonson 1970 {published data only}

Edmonson KW, Graham SM, Warburton MF. A clinical trial of influenza vaccine in Canberra. Medical Journal of Australia 1970;4:6‐13. CENTRAL

Eick‐Cost 2012 {published data only}

Eick‐Cost AA, Tastad KJ, Guerrero AC, Johns MC, Lee S, MacIntosh VH, et al. Effectiveness of seasonal influenza vaccines against influenza‐associated illnesses among US military personnel in 2010‐11: a case‐control approach. PLoS ONE 2012;7(7):e41435. CENTRAL

El'shina 1998 {published data only}

El'shina GA, Gorbunov MA, Shervarli VI, Lonskaia NI, Pavlova LI, Khaitov RM, et al. Evaluation of the effectiveness of influenza trivalent polymer subunit vaccine "Grippol". Zhumal Mikrobiologii Epidemiologii Immunobiologii 1998;3:40‐3. CENTRAL

Englund 1993 {published data only}

Englund JA, Mbawuike IN, Hammill H, Holleman MC, Baxter BD, Glezen WP. Maternal immunization with influenza or tetanus toxoid vaccine for passive antibody protection in young infants. Journal of Infectious Diseases 1993;168(3):647‐56. CENTRAL

Finklea 1969 {published data only}

Finklea JF, Sandifer SH, Peck FB, Manos JP. A clinical and serologic comparison of standard and purified bivalent inactivated influenza vaccines. Journal of Infectious Diseases 1969;120(6):708‐12. CENTRAL

Fisher 2012 {published data only}

Fisher BM, Van Bockern J, Hart J, Lynch AM, Winn VD, Gibbs RS, et al. Pandemic influenza A H1N1 2009 infection versus vaccination: a cohort study comparing immune responses in pregnancy. PLoS ONE 2012;7(3):e33048. CENTRAL

Foy 1981 {published data only}

Foy HM, Cooney MK, Allan ID, Frost F, Blumhagen JM, Fox JP. Influenza B virus vaccines in children and adults: adverse reactions, immune response, and observations in the field. Journal of Infectious Diseases 1981;143(5):700‐6. CENTRAL

Frank 1981 {published data only}

Frank AL, Webster RG, Glezen WP, Cate TR. Immunogenicity of influenza A/USSR (H1N1) subunit vaccine in unprimed young adults. Journal of Medical Virology 1981;7(2):135‐42. CENTRAL

Freestone 1976 {published data only}

Freestone DS. Clinical trials with intranasally administered WRL 105 strain live influenza vaccine in volunteers. Developments in Biological Standardization 1976;33:207‐12. CENTRAL

Gerstoft 2001 {published data only}

Gerstoft J. Influenza vaccination of healthy adults. Ugeskrift for Laeger 2001;163(19):2615‐7. CENTRAL

Greenbaum 2002 {published data only}

Greenbaum E, Furst A, Kiderman A, Stewart B, Levy R, Schlesinger M, et al. Mucosal [SIgA] and serum [IgG] immunologic responses in the community after a single intra‐nasal immunization with a new inactivated trivalent influenza vaccine. Vaccine 2002;20(7‐8):1232‐9. CENTRAL

Greene 2013 {published data only}

Greene SK, Rett MD, Vellozzi C, Li L, Kulldorff M, Marcy SM, et al. Guillain‐Barre syndrome, influenza vaccination, and antecedent respiratory and gastrointestinal infections: a case‐centered analysis in the Vaccine Safety Datalink, 2009‐2011. PLoS ONE 2013;8(6):e67185. CENTRAL

Gross 1999 {published data only}

Gross PA, Sperber SJ, Donabedian A, Dran S, Morchel G, Cataruozolo P, et al. Paradoxical response to a novel influenza virus vaccine strain: the effect of prior immunization. Vaccine 1999;17(18):2284‐9. CENTRAL

Grotto 1998 {published data only}

Grotto I, Mandel Y, Green MS, Varsano N, Gdalevich M, Ashkenazi I, et al. Influenza vaccine efficacy in young, healthy adults. Clinical Infectious Diseases 1998;26(4):913‐7. CENTRAL

Gruber 1994 {published data only}

Gruber WC, Campbell PW, Thompson JM, Reed GW, Roberts B, Wright PF. Comparison of live attenuated and inactivated influenza vaccines in cystic fibrosis patients and their families: results of a 3‐year study. Journal of Infectious Diseases 1994;169(2):241‐7. CENTRAL

Gwini 2011 {published data only}

Gwini SM, Coupland CA, Siriwardena AN. The effect of influenza vaccination on risk of acute myocardial infarction: self‐controlled case‐series study. Vaccine 2011;29(6):1145‐9. CENTRAL

Haber 2004 {published data only}

Haber P, DeStefano F, Angulo FJ, Iskander J, Shadomy SV, Weintraub E, et al. Guillain‐Barre syndrome following influenza vaccination. JAMA 2004;292(20):2478‐81. CENTRAL

Haigh 1973 {published data only}

Haigh W, Howell RW, Meichen FW. A comparative trial of influenza immunization by inhalation and hypojet method. The Practitioner 1973;211:365‐70. CENTRAL

Halperin 2002 {published data only}

Halperin SA, Smith B, Mabrouk T, Germain M, Trepanier P, Hassell T, et al. Safety and immunogenicity of a trivalent, inactivated, mammalian cell culture‐derived influenza vaccine in healthy adults, seniors, and children. Vaccine 2002;20(7‐8):1240‐7. CENTRAL

Hambidge 2011 {published data only}

Hambidge SJ, Ross C, McClure D, Glanz J. Trivalent inactivated influenza vaccine is not associated with sickle cell hospitalizations in adults from a large cohort. Vaccine 2011;29(46):8179‐81. CENTRAL

Heinonen 1973 {published data only}

Heinonen OP, Shapiro S, Monson RR, Hartz SC, Rosenberg L, Slone D. Immunization during pregnancy against poliomyelitis and influenza in relation to childhood malignancy. International Journal of Epidemiology 1973;2(3):229‐35. CENTRAL

Hellenbrand 2012 {published data only}

Hellenbrand W, Jorgensen P, Schweiger B, Falkenhorst G, Nachtnebel M, Greutelaers B, et al. Prospective hospital‐based case‐control study to assess the effectiveness of pandemic influenza A(H1N1)pdm09 vaccination and risk factors for hospitalization in 2009‐2010 using matched hospital and test‐negative controls. BMC Infectious Diseases 2012;12:127. CENTRAL

Hobson 1970 {published data only}

Hobson D, Baker FA, Chivers CP, Reed SE, Sharp D. A comparison of monovalent Hong Kong influenza virus vaccine with vaccines containing only pre‐1968 Asian strains in adult volunteers. Journal of Hygiene 1970;68(3):369‐78. CENTRAL

Hobson 1973 {published data only}

Hobson D, Baker FA, Curry RL, Beare AS, Massey PM. The efficacy of live and inactivated vaccines of Hong Kong influenza virus in an industrial community. Journal of Hygiene 1973;71(4):641‐7. CENTRAL

Hoskins 1973 {published data only}

Hoskins TW, Davies JR, Allchin A, Miller CL, Pollock TM. Controlled trial of inactivated influenza vaccine containing the A/Hong Kong strain during an outbreak of influenza due to the A/England/42/72 strain. Lancet 1973;2(7821):116‐20. CENTRAL

Hoskins 1976 {published data only}

Hoskins TW, Davies JR, Smith AJ, Allchin A, Miller CL, Pollock TM. Influenza at Christ's Hospital: March, 1974. Lancet 1976;1(7951):105‐8. CENTRAL

Hoskins 1979 {published data only}

Hoskins TW, Davies JR, Smith AJ, Miller CL, Allchin A. Assessment of inactivated influenza A vaccine after three outbreaks of influenza A at Christ's Hospital. Lancet 1979;1(8106):33‐5. CENTRAL

Howell 1967 {published data only}

Howell RW. Long term efficacy of oil‐adjuvant influenza vaccine in an industrial population. British Journal of Industrial Medicine 1967;24:66‐70. CENTRAL

Huang 2011 {published data only}

Huang WT, Chen WC, Teng HJ, Huang WI, Huang YW, Hsu CW. Adverse events following pandemic A (H1N1) 2009 monovalent vaccines in pregnant women ‐ Taiwan, November 2009‐August 2010. PLoS ONE 2011;6(8):e23049. CENTRAL

Hurwitz 1983 {published data only}

Hurwitz ES, Holman RC, Nelson DB, Schonberger LB. National surveillance for Guillain‐Barré syndrome: January 1978‐March 1979. Neurology 1983;33(2):150‐7. CENTRAL

Jackson 2011 {published data only}

Jackson LA, Patel SM, Swamy GK, Frey SE, Creech CB, Munoz FM, et al. Immunogenicity of an inactivated monovalent 2009 H1N1 influenza vaccine in pregnant women. Journal of Infectious Diseases 2011;204(6):854‐63. CENTRAL

Janjua 2012 {published data only}

Janjua NZ, Skowronski DM, De Serres G, Dickinson J, Crowcroft NS, Taylor M, et al. Estimates of influenza vaccine effectiveness for 2007‐2008 from Canada's sentinel surveillance system: cross‐protection against major and minor variants. Journal of Infectious Diseases 2012;205(12):1858‐68. CENTRAL

Jianping 1999 {published data only}

Jianping H, Xin F, Changshun L, Bo Z, Linxiu G, Wei X, et al. Assessment of effectiveness of Vaxigrip. Vaccine 1999;17(Suppl 1):57‐8. CENTRAL

Jimenez‐Jorge 2012 {published data only}

Jimenez‐Jorge S, de Mateo S, Pozo F, Casas I, Garcia Cenoz M, Castilla J, et al. Early estimates of the effectiveness of the 2011/12 influenza vaccine in the population targeted for vaccination in Spain, 25 December 2011 to 19 February 2012. Euro Surveillance 2012;17(12):20129. CENTRAL

Keitel 2001 {published data only}

Keitel WA, Cate TR, Nino D, Huggins LL, Six HR, Quarles JM, et al. Immunization against influenza: comparison of various topical and parenteral regimens containing inactivated and/or live attenuated vaccines in healthy adults. Journal of Infectious Diseases 2001;183(2):329‐32. CENTRAL

Kelly 2012 {published data only}

Kelly HA, Sullivan SG, Grant KA, Fielding JE. Moderate influenza vaccine effectiveness with variable effectiveness by match between circulating and vaccine strains in Australian adults aged 20‐64 years, 2007‐2011. Influenza and Other Respiratory Viruses 2012;7(5):729‐37. CENTRAL

Khazeni 2009 {published data only}

Khazeni N, Hutton D, Garber A, Hupert N, Owens D. Effectiveness and cost‐effectiveness of vaccination against pandemic influenza (H1N1) 2009. Annals of Internal Medicine 2009;151:829‐39. CENTRAL

Kiderman 2001 {published data only}

Kiderman A, Furst A, Stewart B, Greenbaum E, Morag A, Zakay‐Rones Z. A double‐blind trial of a new inactivated, trivalent, intra‐nasal anti‐influenza vaccine in general practice: relationship between immunogenicity and respiratory morbidity over the winter of 1997‐98. Journal of Clinical Virology 2001;20(3):155‐61. CENTRAL

Kim 2012 {published data only}

Kim J‐H, Cho H‐Y, Hennessey KA, Lee HJ, Bae GR, Kim HC. Adverse events following immunization (AEFI) with the novel influenza A (H1N1) 2009 vaccine: findings from the national registry of all vaccine recipients and AEFI and the passive surveillance system in South Korea. Japanese Journal of Infectious Diseases 2012;65(2):99‐104. CENTRAL

Kissling 2012 {published data only}

Kissling E, Valenciano M. Early estimates of seasonal influenza vaccine effectiveness in Europe among target groups for vaccination: results from the I‐MOVE multicentre case‐control study, 2011/12. Euro Surveillance 2012;17(15):20146. CENTRAL

Kunz 1977 {published data only}

Kunz C, Hofmann H, Bachmayer H, Liehl E, Moritz AJ. Clinical trials with a new influenza subunit vaccine in adults and children. Developments in Biological Standardization 1977;39:297‐302. CENTRAL

Langley 2004 {published data only}

Langley JM, Faughnan ME. Prevention of influenza in the general population. Canadian Medical Association Journal 2004;171(10):1213‐22. CENTRAL

Lavallee 2014 {published data only}

Lavallee PC, Labreuche J, Fox KM, Lavados P, Mattle H, Steg PG, et al. Influenza vaccination and cardiovascular risk in patients with recent TIA and stroke. Neurology 2014;82(21):1905‐13. CENTRAL

Lee 2011 {published data only}

Lee GM, Greene SK, Weintraub ES, Baggs J, Kulldorff M, Fireman BH, et al. H1N1 and seasonal influenza vaccine safety in the Vaccine Safety Datalink project. American Journal of Preventive Medicine 2011;41(2):121‐8. CENTRAL

Leeb 2011 {published data only}

Leeb A, Carcione D, Richmond PC, Jacoby P, Effler PV. Reactogenicity of two 2010 trivalent inactivated influenza vaccine formulations in adults. Vaccine 2011;29(45):7920‐4. CENTRAL

Leroux‐Roels 2010a {published data only}

Leroux‐Roels I, Roman F, Forgus S, Maes C, De Boever F, Drame M, et al. Priming with AS03 A‐adjuvanted H5N1 influenza vaccine improves the kinetics, magnitude and durability of the immune response after a heterologous booster vaccination: an open non‐randomised extension of a double‐blind randomised primary study. Vaccine 2010;28(3):849‐57. CENTRAL

Leroux‐Roels 2010b {published data only}

Leroux‐Roels I, Vets E, Freese R, Seiberling M, Weber F, Salamand C, et al. Corrigendum to: "Seasonal influenza vaccine delivered by intradermal microinjection: a randomised controlled safety and immunogenicity trial in adults" by Leroux‐Roels et al. [Vaccine 26 (2008) 6614‐6619]. Vaccine 2010;28(50):8033. CENTRAL

Liem 1973 {published data only}

Liem KS, Marcus EA, Jacobs J, Strik RV. The protective effect of intranasal immunization with inactivated influenza virus vaccine. Postgraduate Medical Journal 1973;49(569):175‐9. CENTRAL

Lind 2014 {published data only}

Lind A, Ramelius A, Olsson T, Arnheim‐Dahlstrom L, Lamb F, Khademi M, et al. A/H1N1 antibodies and TRIB2 autoantibodies in narcolepsy patients diagnosed in conjunction with the Pandemrix vaccination campaign in Sweden 2009‐2010. Journal of Autoimmunity 2014;50:99‐106. CENTRAL

Liu 2012 {published data only}

Liu N, Sprague AE, Yasseen AS, Fell DB, Wen SW, Smith GN, et al. Vaccination patterns in pregnant women during the 2009 H1N1 influenza pandemic: a population‐based study in Ontario, Canada. Canadian Journal of Public Health 2012;103(5):e353‐8. CENTRAL

Louik 2013 {published data only}

Louik C, Chambers C, Jacobs D, Rice F, Johnson D, Mitchell AA. Influenza vaccine safety in pregnancy: can we identify exposures?. Pharmacoepidemiology and Drug Safety 2013;22(1):33‐9. CENTRAL

Mackenzie 1975 {published data only}

Mackenzie JS, Mackenzie I, Lloyd J, Dent V. Comparative trials of live attenuated and detergent split influenza virus vaccines. Journal of Hygiene 1975;75(3):425‐43. CENTRAL

Mackenzie 2012 {published data only}

Mackenzie IS, Macdonald TM, Shakir S, Dryburgh M, Mantay BJ, Mcdonnell P, et al. Influenza H1N1 (swine flu) vaccination: a safety surveillance feasibility study using self‐reporting of serious adverse events and pregnancy outcomes. British Journal of Clinical Pharmacology 2012;73(5):801‐11. CENTRAL

Mair 1974 {published data only}

Mair HJ, Sansome DAW, Tillett HE. A controlled trial of inactivated monovalent influenza A vaccines in general practice. Journal of Hygiene 1974;73:317‐27. CENTRAL

Maynard 1968 {published data only}

Maynard JE, Dull HB, Hanson ML, Feltz ET, Berger R, Hammes L. Evaluation of monovalent and polyvalent influenza vaccines during an epidemic of type A2 and B influenza. American Journal of Epidemiology 1968;87(1):148‐57. CENTRAL

McCarthy 2004 {published data only}

McCarthy MW, Kockler DR. Trivalent intranasal influenza vaccine, live. Annals of Pharmacotherapy 2004;38(12):2086‐93. CENTRAL

Mendelman 2001 {published data only}

Mendelman PM, Cordova J, Cho I. Safety, efficacy and effectiveness of the influenza virus vaccine, trivalent, types A and B, live, cold‐adapted (CAIV‐T) in healthy children and healthy adults. Vaccine 2001;19(17‐19):2221‐6. CENTRAL

Merelli 2000 {published data only}

Merelli E, Casoni F. Prognostic factors in multiple sclerosis: role of intercurrent infections and vaccinations against influenza and hepatitis B. Neurological Sciences 2000;21(4 Suppl 2):853‐6. CENTRAL

Meyers 2003a {published data only}

Meyers DG. Could influenza vaccination prevent myocardial infarction, stroke and sudden cardiac death?. American Journal of Cardiovascular Drugs 2003;3(4):241‐4. CENTRAL

Meyers 2003b {published data only}

Meyers DG. Myocardial infarction, stroke, and sudden cardiac death may be prevented by influenza vaccination. Current Atherosclerosis Reports 2003;5(2):146‐9. CENTRAL

Micheletti 2011 {published data only}

Micheletti F, Moretti U, Tridente G, Zanoni G. Consultancy and surveillance of post‐immunisation adverse events in the Veneto region of Italy for 1992‐2008. Human Vaccines 2011;7(Suppl):234‐9. CENTRAL

Monto 2000 {published data only}

Monto AS. Preventing influenza in healthy adults: the evolving story. JAMA 2000;284(13):1699‐701. CENTRAL

Montplaisir 2014 {published data only}

Montplaisir J, Petit D, Quinn M‐J, Ouakki M, Deceuninck G, Desautels A, et al. Risk of narcolepsy associated with inactivated adjuvanted (AS03) A/H1N1 (2009) pandemic influenza vaccine in Quebec. PLoS ONE 2014;9(9):e108489. CENTRAL

Moro 2011 {published data only}

Moro PL, Broder K, Zheteyeva Y, Revzina N, Tepper N, Kissin D, et al. Adverse events following administration to pregnant women of influenza A (H1N1) 2009 monovalent vaccine reported to the Vaccine Adverse Event Reporting System. American Journal of Obstetrics & Gynecology 2011;205(5):473.e1‐9. CENTRAL

Morris 1975 {published data only}

Morris CA, Freestone DS, Stealey VM, Oliver PR. Recombinant WRL 105 strain live attenuated influenza vaccine. Immunogenicity, reactivity, and transmissibility. Lancet 1975;2(7927):196‐9. CENTRAL

Mostow 1977 {published data only}

Mostow SR, Eichkoff TC, Chelgren GA, Retailliau HF, Castle M. Studies of inactivated influenza virus vaccines in hospital employees: reactogenicity and absenteeism. Journal of Infectious Diseases 1977;136 Suppl:S533‐8. CENTRAL

Muennig 2001 {published data only}

Muennig PA, Khan K. Cost‐effectiveness of vaccination versus treatment of influenza in healthy adolescents and adults. Clinical Infectious Diseases 2001;33(11):1879‐85. CENTRAL

Murray 1979 {published data only}

Murray DL, Imagawa DT, Okada DM, St Geme JW. Antibody response to monovalent A/New Jersey/8/76 influenza vaccine in pregnant women. Journal of Clinical Microbiology 1979;10(2):184‐7. CENTRAL

Nazareth 2013 {published data only}

Nazareth I, Tavares F, Rosillon D, Haguinet F, Bauchau V. Safety of AS03‐adjuvanted split‐virion H1N1 (2009) pandemic influenza vaccine: a prospective cohort study. BMJ Open 2013;3(2):e001912. CENTRAL

Nichol 1996 {published data only}

Nichol KL, Margolis KL, Lind A, Murdoch M, McFadden R, Hauge M, et al. Side effects associated with influenza vaccination in healthy working adults. Archives of Internal Medicine 1996;156(14):1546‐50. CENTRAL

Nichol 1999b {published data only}

Nichol KL. Clinical effectiveness and cost effectiveness of influenza vaccination among healthy working adults. Vaccine 1999;17(Suppl 1):67‐73. CENTRAL

Nichol 2001 {published data only}

Nichol KL. Live attenuated influenza virus vaccines: new options for the prevention of influenza. Vaccine 2001;19(31):4373‐7. CENTRAL

Nichol 2003 {published data only}

Nichol KL, Mallon KP, Mendelman PM. Cost benefit of influenza vaccination in healthy, working adults: an economic analysis based on the results of a clinical trial of trivalent live attenuated influenza virus vaccine. Vaccine 2003;21(17‐8):2207‐17. CENTRAL

Nichol 2004 {published data only}

Nichol KL, Mendelman P. Influence of clinical case definitions with differing levels of sensitivity and specificity on estimates of the relative and absolute health benefits of influenza vaccination among healthy working adults and implications for economic analyses. Virus Research 2004;103(1‐2):3‐8. CENTRAL

Omon 2011 {published data only}

Omon E, Damase‐Michel C, Hurault‐Delarue C, Lacroix I, Montastruc JL, Oustric S, et al. Non‐adjuvanted 2009 influenza A (H1N1)v vaccine in pregnant women: the results of a French prospective descriptive study. Vaccine 2011;29(52):9649‐54. CENTRAL

Petrie 2011 {published data only}

Petrie JG, Ohmit SE, Johnson E, Cross RT, Monto AS. Efficacy studies of influenza vaccines: effect of end points used and characteristics of vaccine failures. Journal of Infectious Diseases 2011;203(9):1309‐15. CENTRAL

Phillips 2013 {published data only}

Phillips CJ, Woolpert T, Sevick C, Faix D, Blair PJ, Crum‐Cianflone NF. Comparison of the effectiveness of trivalent inactivated influenza vaccine and live, attenuated influenza vaccine in preventing influenza‐like illness among US military service members, 2006‐2009. Clinical Infectious Diseases 2013;56(1):11‐9. CENTRAL

Phonrat 2013 {published data only}

Phonrat B, Pitisuttithum P, Chamnanchanunt S, Puthavathana P, Ngaosuwankul N, Louisirirotchanakul S, et al. Safety and immune responses following administration of H1N1 live attenuated influenza vaccine in Thais. Vaccine 2013;31(11):1503‐9. CENTRAL

Pleguezuelos 2012 {published data only}

Pleguezuelos O, Robinson S, Stoloff GA, Caparrós‐Wanderley W. Synthetic influenza vaccine (FLU‐v) stimulates cell mediated immunity in a double‐blind, randomised, placebo‐controlled Phase I trial. Vaccine 2012;30(31):4655‐60. CENTRAL

Puig‐Barbera 2012 {published data only}

Puig‐Barbera J, Diez‐Domingo J, Arnedo‐Pena A, Ruiz‐Garcia M, Perez‐Vilar S, Mico‐Esparza JL, et al. Effectiveness of the 2010‐2011 seasonal influenza vaccine in preventing confirmed influenza hospitalizations in adults: a case‐case comparison, case‐control study. Vaccine 2012;30(39):5714‐20. CENTRAL

Puleston 2010 {published data only}

Puleston RL, Bugg G, Hoschler K, Konje J, Thornton J, Stephenson I, et al. Observational study to investigate vertically acquired passive immunity in babies of mothers vaccinated against H1N1v during pregnancy. Health Technology Assessment 2010;14(55):1‐82. CENTRAL

Pyhala 2001 {published data only}

Pyhala R, Haanpaa M, Kleemola M, Tervahauta R, Visakorpi R, Kinnunen L. Acceptable protective efficacy of influenza vaccination in young military conscripts under circumstances of incomplete antigenic and genetic match. Vaccine 2001;19(23‐4):3253‐60. CENTRAL

Reynales 2012 {published data only}

Reynales H, Astudillo P, de Valliere S, Hatz C, Schlagenhauf P, Rath B, et al. A prospective observational safety study on MF59 adjuvanted cell culture‐derived vaccine, Celtura during the A/H1N1 (2009) influenza pandemic. Vaccine 2012;30(45):6436‐43. CENTRAL

Rimmelzwaan 2000 {published data only}

Rimmelzwaan GF, Nieuwkoop N, Brandenburg A, Sutter G, Beyer WE, Maher D, et al. A randomized, double blind study in young healthy adults comparing cell mediated and humoral immune responses induced by influenza ISCOM vaccines and conventional vaccines. Vaccine 2000;19(9‐10):1180‐7. CENTRAL

Rocchi 1979c {published data only}

Rocchi G, Carlizza L, Andreoni M, Ragona G, Piga C, Pelosio A, et al. Protection from natural infection after live influenza virus immunization in an open population. Journal of Hygiene 1979;82(2):231‐6. CENTRAL

Rowhani‐Rahbar 2012 {published data only}

Rowhani‐Rahbar A, Klein NP, Lewis N, Fireman B, Ray P, Rasgon B, et al. Immunization and Bell's palsy in children: a case‐centered analysis. American Journal of Epidemiology 2012;175(9):878‐85. CENTRAL

Ruben 1972 {published data only}

Ruben FL, Jackson GG. A new subunit influenza vaccine: acceptability compared with standard vaccines and effect of dose on antigenicity. Journal of Infectious Diseases 1972;125(6):656‐64. CENTRAL

Ruben 1973 {published data only}

Ruben FL, Akers LW, Stanley ED, Jackson GG. Protection with split and whole virus vaccines against influenza. Archives of Internal Medicine 1973;132(4):568‐71. CENTRAL

Safranek 1991 {published data only}

Safranek TJ, Lawrence DN, Kurland LT, Culver DH, Wiederholt WC, Hayner NS, et al. Reassessment of the association between Guillain‐Barre syndrome and receipt of swine influenza vaccine in 1976‐1977: results of a two‐state study. Expert Neurology Group. American Journal of Epidemiology 1991;133(9):940‐51. CENTRAL

Sarateanu 1980 {published data only}

Sarateanu DE, Ehrengut W, Pressler K, Peukert M, Schenk KD. Serological response to whole, split and subunit influenza vaccines of persons with and without immunological experience towards influenza A/U.S.S.R. 90/77 virus. Comparative Immunology, Microbiology and Infectious Diseases 1980;3(1‐2):225‐36. CENTRAL

Scheifele 2013 {published data only}

Scheifele DW, Dionne M, Ward BJ, Cooper C, Vanderkooi OG, Li Y, et al. Safety and immunogenicity of 2010‐2011 A/H1N1pdm09‐containing trivalent inactivated influenza vaccine in adults previously given AS03‐adjuvanted H1N1 2009 pandemic vaccine: results of a randomised trial. Human Vaccines and Immunotherapy 2013;9(1):136‐43. CENTRAL

Schonberger 1981 {published data only}

Schonberger LB, Hurwitz ES, Katona P, Holman RC, Bregman DJ. Guillain‐Barré syndrome: its epidemiology and associations with influenza vaccination. Annals of Neurology 1981;9(Suppl):31‐8. CENTRAL

Schwartz 1996 {published data only}

Schwartz K. Influenza vaccine for healthy adults. Journal of Family Practice 1996;42(4):351‐2. CENTRAL

Simpson 2012 {published data only}

Simpson CR, Ritchie LD, Robertson C, Sheikh A, McMenamin J. Effectiveness of H1N1 vaccine for the prevention of pandemic influenza in Scotland, UK: a retrospective observational cohort study. Lancet Infectious Diseases 2012;12(9):696‐702. CENTRAL

Sipilä 2015 {published data only}

Sipilä JO, Soilu‐Hanninen M. The incidence and triggers of adult‐onset Guillain‐Barre syndrome in southwestern Finland 2004‐2013. European Journal of Neurology 2015;22(2):292‐8. CENTRAL

Skowronski 2002 {published data only}

Skowronski DM, Strauss B, Kendall P, Duval B, De Serres G. Low risk of recurrence of oculorespiratory syndrome following influenza revaccination. Canadian Medical Association Journal 2002;167(8):853‐8. CENTRAL

Skowronski 2003 {published data only}

Skowronski DM, De Serres G, Scheifele D, Russell ML, Warrington R, Davies HD, et al. Randomized, double‐blind, placebo‐controlled trial to assess the rate of recurrence of oculorespiratory syndrome following influenza vaccination among persons previously affected. Clinical Infectious Diseases 2003;37(8):1059‐66. CENTRAL

Smith 1977a {published data only}

Smith AJ, Davies JR. The response to inactivated influenza A (H3N2) vaccines: the development and effect of antibodies to the surface antigens. Journal of Hygiene 1977;78:363‐75. CENTRAL

Smith 1977b {published data only}

Smith CD, Leighton HA, Shiromoto RS. Antigenicity and reactivity of influenza A/New Jersey/8/76 virus vaccines in military volunteers at Fort Ord, California. Journal of Infectious Diseases 1977;136(Suppl):460‐5. CENTRAL

Song 2011 {published data only}

Song JY, Cheong HJ, Woo HJ, Wie SH, Lee JS, Chung MH, et al. Immunogenicity and safety of trivalent inactivated influenza vaccine: a randomised, double‐blind, multi‐center, phase 3 clinical trial in a vaccine‐limited country. Journal of Korean Medical Science 2011;26(2):191‐5. CENTRAL

Souayah 2011 {published data only}

Souayah N, Michas‐Martin PA, Nasar A, Krivitskaya N, Yacoub HA, Khan H, et al. Guillain‐Barré syndrome after Gardasil vaccination: data from Vaccine Adverse Event Reporting System 2006‐2009. Vaccine 2011;29(5):886‐9. CENTRAL

Spencer 1975 {published data only}

Spencer MJ, Cherry JD, Powell KR, Sumaya CV, Garakian AJ. Clinical trials with Alice strain, live attenuated, serum inhibitor‐resistant intranasal influenza A vaccine. Journal of Infectious Diseases 1975;132(4):415‐20. CENTRAL

Spencer 1979 {published data only}

Spencer MJ, Cherry JD, Powell KR, Sumaya CV. A clinical trial with Alice/R‐75 strain, live attenuated serum inhibitor‐resistant intranasal bivalent influenza A/B vaccine. Medical Microbiology and Immunology 1979;167(1):1‐9. CENTRAL

Steinhoff 2012 {published data only}

Steinhoff MC, Omer SB, Roy E, El Arifeen S, Raqib R, Dodd C, et al. Neonatal outcomes after influenza immunization during pregnancy: a randomised controlled trial. Canadian Medical Association Journal 2012;184(6):645‐53. CENTRAL

Sumaya 1979 {published data only}

Sumaya CV, Gibbs RS. Immunization of pregnant women with influenza A/New Jersey/76 virus vaccine: reactogenicity and immunogenicity in mother and infant. Journal of Infectious Diseases 1979;140(2):141‐6. CENTRAL

Talaat 2010 {published data only}

Talaat KR, Greenberg ME, Lai MH, Hartel GF, Wichems CH, Rockman S, et al. A single dose of unadjuvanted novel 2009 H1N1 vaccine is immunogenic and well tolerated in young and elderly adults. Journal of Infectious Diseases 2010;202(9):1327‐37. CENTRAL

Tavares 2011 {published data only}

Tavares F, Nazareth I, Monegal JS, Kolte I, Verstraeten T, Bauchau V. Pregnancy and safety outcomes in women vaccinated with an AS03‐adjuvanted split virion H1N1 (2009) pandemic influenza vaccine during pregnancy: a prospective cohort study. Vaccine 2011;29(37):6358‐65. CENTRAL

Taylor 1969 {published data only}

Taylor PJ, Miller CL, Pollock TM, Perkins FT, Westwood MA. Antibody response and reactions to aqueous influenza vaccine, simple emulsion vaccine and multiple emulsion vaccine. A report to the Medical Research Council Committee on influenza and other respiratory virus vaccines. Journal of Hygiene 1969;67(3):485‐90. CENTRAL

Taylor 2012 {published data only}

Taylor DN, Treanor JJ, Sheldon EA, Johnson C, Umlauf S, Song L, et al. Development of VAX128, a recombinant hemagglutinin (HA) influenza‐flagellin fusion vaccine with improved safety and immune response. Vaccine 2012;30(39):5761‐9. CENTRAL

Thompson 2014 {published data only}

Thompson MG, Li DK, Shifflett P, Sokolow LZ, Ferber JR, Kurosky S, et al. Effectiveness of seasonal trivalent influenza vaccine for preventing influenza virus illness among pregnant women: a population‐based case‐control study during the 2010‐2011 and 2011‐2012 influenza seasons. Clinical Infectious Diseases 2014;58(4):449‐57. CENTRAL

Tokars 2012 {published data only}

Tokars JI, Lewis P, Destefano F, Wise M, Viray M, Morgan O, et al. The risk of Guillain‐Barré syndrome associated with influenza a (H1N1) 2009 monovalent vaccine and 2009‐2010 seasonal influenza vaccines: results from self‐controlled analyses. Pharmacoepidemiology and Drug Safety 2012;21(5):546‐52. CENTRAL

Treanor 2001 {published data only}

Treanor JJ, Wilkinson BE, Masseoud F, Hu‐Primmer J, Battaglia R, O'Brien D, et al. Safety and immunogenicity of a recombinant hemagglutinin vaccine for H5 influenza in humans. Vaccine 2001;19(13‐4):1732‐7. CENTRAL

Treanor 2002 {published data only}

Treanor J, Keitel W, Belshe R, Campbell J, Schiff G, Zangwill K, et al. Evaluation of a single dose of half strength inactivated influenza vaccine in healthy adults. Vaccine 2002;20(7‐8):1099‐105. CENTRAL

Treanor 2012 {published data only}

Treanor JJ, Talbot HK, Ohmit SE, Coleman LA, Thompson MG, Cheng PY, et al. Effectiveness of seasonal influenza vaccines in the United States during a season with circulation of all three vaccine strains. Clinical Infectious Diseases 2012;55(7):951‐9. CENTRAL

Tsai 2010 {published data only}

Tsai T, Kyaw MH, Novicki D, Nacci P, Rai S, Clemens R. Exposure to MF59‐adjuvanted influenza vaccines during pregnancy ‐ a retrospective analysis. Vaccine 2010;28(7):1877‐80. CENTRAL

Tsatsaris 2011 {published data only}

Tsatsaris V, Capitant C, Schmitz T, Chazallon C, Bulifon S, Riethmuller D, et al. Maternal immune response and neonatal seroprotection from a single dose of a monovalent nonadjuvanted 2009 influenza A(H1N1) vaccine: a single‐group trial. Annals of Internal Medicine 2011;155(11):733‐41. CENTRAL

Tyrrell 1970 {published data only}

Tyrrell DA, Buckland R, Rubenstein D, Sharpe DM. Vaccination against Hong Kong influenza in Britain, 1968‐9. A report to the Medical Research Council Committee on Influenza and other Respiratory Virus Vaccines. Journal of Hygiene 1970;68(3):359‐68. CENTRAL

Vesikari 2012 {published data only}

Vesikari T, Forstén A, Herbinger KH, Cioppa GD, Beygo J, Borkowski A, et al. Safety and immunogenicity of an MF59‐adjuvanted A/H5N1 pre‐pandemic influenza vaccine in adults and the elderly. Vaccine 2012;30(7):1388‐96. CENTRAL

Warren‐Gash 2013 {published data only}

Warren‐Gash C, Geretti AM, Hamilton G, Rakhit RD, Smeeth L, Hayward AC. Influenza‐like illness in acute myocardial infarction patients during the winter wave of the influenza A H1N1 pandemic in London: a case‐control study. BMJ Open 2013;3(5):e002604. CENTRAL

Warshauer 1976 {published data only}

Warshauer DM, Minor TE, Inhorn SL, Reed CE, Dick EC. Use of an inhibitor‐resistant live attenuated influenza vaccine in normal and asthmatic adults. 14th Congress of the International Association of Biological Standardization, Douglas, Isle of Man 1975. Developments in Biological Standardization 1975;33:184‐90. CENTRAL

Wilde 1999 {published data only}

Wilde JA, McMillan JA, Serwint J, Butta J, O'Riordan MA, Steinhoff MC. Effectiveness of influenza vaccine in health care professionals. JAMA 1999;281(10):908‐13. CENTRAL

Williams 1973 {published data only}

Williams MC, Davignon L, McDonald JC, Pavilanis PV, Boudreault A, Clayton AJ. Trial of aqueous killed influenza vaccine in Canada, 1968‐69. WHO Bulletin 1973;49:333‐40. CENTRAL

Williams 2011 {published data only}

Williams SE, Pahud BA, Vellozzi C, Donofrio PD, Dekker CL, Halsey N, et al. Causality assessment of serious neurologic adverse events following 2009 H1N1 vaccination. Vaccine 2011;29(46):8302‐8. CENTRAL

Wise 2012 {published data only}

Wise ME, Viray M, Sejvar JJ, Lewis P, Baughman AL, Connor W, et al. Guillain‐Barre syndrome during the 2009‐2010 H1N1 influenza vaccination campaign: population‐based surveillance among 45 million Americans. American Journal of Epidemiology 2012;175(11):1110‐9. CENTRAL

Wood 1999 {published data only}

Wood SC, Alexseiv A, Nguyen VH. Effectiveness and economical impact of vaccination against influenza among a working population in Moscow. Vaccine 1999;17(Suppl 3):81‐7. CENTRAL

Wood 2000 {published data only}

Wood SC, Nguyen VH, Schmidt C. Economic evaluations of influenza vaccination in healthy working‐age adults. Employer and society perspective. Pharmacoeconomics 2000;18(2):173‐83. CENTRAL

Xu 2012 {published data only}

Xu R, Luo Y, Chambers C. Assessing the effect of vaccine on spontaneous abortion using time‐dependent covariates Cox models. Pharmacoepidemiology and Drug Safety 2012;21(8):844‐50. CENTRAL

Yang 2012 {published data only}

Yang Z, Wang S, Li W, Li C, Dong J, Li F, et al. The long‐term immunogenicity of an inactivated split‐virion 2009 pandemic influenza A H1N1 vaccine: randomized, observer‐masked, single‐center clinical study. Results in Immunology 2012;2:184‐9. CENTRAL

Yeager 1999 {published data only}

Yeager DP, Toy EC, Baker B. Influenza vaccination in pregnancy. American Journal of Perinatology 1999;16(6):283‐6. CENTRAL

Yih 2012 {published data only}

Yih WK, Lee GM, Lieu TA, Ball R, Kulldorff M, Rett M, et al. Surveillance for adverse events following receipt of pandemic 2009 H1N1 vaccine in the Post‐Licensure Rapid Immunization Safety Monitoring (PRISM) System, 2009‐2010. American Journal of Epidemiology 2012;175(11):1120‐8. CENTRAL

Zaman 2008 {published data only}

Zaman K, Roy E, Arifeen SE, Rahman M, Raqib R, Wilson E, et al. Effectiveness of maternal influenza immunization in mothers and infants. New England Journal of Medicine 2008;359(15):1555‐64. CENTRAL

ACIP 2006

Centers for Disease Control and Prevention. Prevention and control of influenza: recommendations of the Advisory Committee on Immunization Practices (ACIP). Recommendations and Reports: Morbidity and Mortality Weekly Report 2006;55:1‐41.

ACIP 2010

Fiore AE, Uyeki TM, Broder K, Finelli L, Euler GL, Singleton JA, et al. Prevention and control of influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices (ACIP), 2010. Morbidity and Mortality Weekly Report 2010;59(RR‐8):1‐62.

ACIP 2015

Advisory Committee on Immunization Practices. Prevention and control of influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices, United States, 2015‐16 influenza season. Morbidity and Mortality Weekly Report 2015;64(30):818‐25.

AIH 2013

Australian Technical Advisory Group on Immunisation (ATAGI). The Australian Immunisation Handbook. 10th Edition. Australian Government: National Health and Medical Research Council, 2013.

Atkins 2004

Atkins D, Best D, Briss PA, Eccles M, Falck‐Ytter Y, Flottorp S, et al. GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490.

Demicheli 2014

Demicheli V, Jefferson T, Al‐Ansary LA, Ferroni E, Rivetti A, Di Pietrantonj C. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 2014, Issue 3. [DOI: 10.1002/14651858.CD001269.pub5]

DiazGranados 2012

DiazGranados CA, Denis M, Plotkin S. Seasonal influenza vaccine efficacy and its determinants in children and non‐elderly adults: a systematic review with meta‐analyses of controlled trials. Vaccine 2012;31(1):49‐57.

DoH 2015

Department of Health, UK. Influenza. Immunisation Against Infectious Diseases: The Green Book. Vol. 19, Department of Health, UK, 2015:18‐9.

EMA 2014

Committee for Medicinal Products for Human Use. Guideline on influenza vaccines. Non‐clinical and clinical module. European Medicines Agency, 2014, EMA/CHMP/VWP/457259/2014. [web:: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2016/07/WC500211324.pdf]

Farez 2011

Farez MF, Correale J. Immunizations and risk of multiple sclerosis: systematic review and meta‐analysis. Journal of Neurology 2011;258(7):1197‐206.

GRADEpro GDT 2014 [Computer program]

GRADE Working Group, McMaster University. GRADEpro GDT. Hamilton (ON): GRADE Working Group, McMaster University, 2014.

Grohskopf 2016

Grohskopf LA, Sokolow LZ, Broder KR, Olsen SJ, Karron RA, Jernigan DB, et al. Prevention and control of seasonal influenza with vaccines. Morbidity and Mortality Weekly Report 2016;65(5):1‐54. [DOI: 10.15585/mmwr.rr6505a1]

Higgins 2011

Higgins JP, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Jefferson 2009a

Jefferson TO. Mistaken identity: seasonal influenza versus influenza‐like illness. Clinical Evidence 2009;329:1‐4.

Jefferson 2009b

Jefferson T, Di Pietrantonj C, Debalini MG, Rivetti A, Demicheli V. Relation of study quality, concordance, take home message, funding, and impact in studies of influenza vaccines: systematic review. BMJ 2009;338:b354. [DOI: 10.1136/bmj.b354]

Jefferson 2012

Jefferson T, Rivetti A, Di Pietrantonj C, Demicheli V, Ferroni E. Vaccines for preventing influenza in healthy children. Cochrane Database of Systematic Reviews 2012, Issue 8. [DOI: 10.1002/14651858.CD004879.pub4]

Jones 2016

Jones M, Fowler R. Immortal time bias in observational studies of time to event outcomes. Journal of Critical Care 2016;36:195‐9.

Kissling 2009a

Kissling E, Moren A, Valenciano M. Protocol for case‐control studies to measure pandemic and seasonal influenza vaccine effectiveness in the European Union and European Economic Area Member States. ECDC Technical Document. Paris: European Centre for Disease Prevention and Control, 2009. [web: http://ecdc.europa.eu/en/publications/Publications/0907_TED_Influenza_AH1N1_Measuring_Influenza_Vaccine_Effectiveness_Protocol_Case_Control_Studies.pdf last accessed on May 25th, 2017]

Kissling 2009b

Kissling E, Moren A, Valenciano M. Protocol for cohort database studies to measure pandemic and seasonal influenza vaccine effectiveness in the European Union and European Economic Area Member States. ECDC Technical Document. Paris: European Centre for Disease Prevention and Control, 2009. [web: http://ecdc.europa.eu/en/publications/Publications/0907_TER_Influenza_AH1N1_Measuring_Influenza_Vaccine_Effectiveness_Protocol_Cohort_Database_Studies.pdf Last accessed on May 25th, 2017]

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group. Preferred reporting items for systematic reviews and meta‐analyses: The PRISMA Statement. BMJ 2009;339:2535.

NACI 2014

National Advisory Committee on Immunization (NACI). An Advisory Committee Statement (ACS) National Advisory Commitee on Immunization: statement on seasonal influenza vaccine for 2014‐2015. Canada Communicable Diseases Report, 2014. http:/www.phac‐aspc.gcca/naci‐ccni/assets/pdf/flu‐grippe‐eng.pdf (accessed September 3, 2015).

Osterholm 2012

Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta‐analysis. Lancet Infectious Diseases 2012;12:36‐44.

RevMan 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Skowronski 2009

Skowronski DM, De Serres G. Is routine immunisation warranted in early pregnancy?. Vaccine 2009;27:4754‐70.

STIKO 2010

Ständigen Impfkommission (STIKO) am Robert Koch‐Institut. Recommendations update for immunisation against influenza [Änderung der Empfehlungen zur Impfung gegen Influenza]. Epidemiologisches Bulletin 2010;31:299‐308.

Toback 2012

Toback SL, Levin MJ, Block SL, Belshe RB, Ambrose CS, Falloon J. Quadrivalent Ann Arbor strain live‐attenuated influenza vaccine. Expert Reviews Vaccines 2012;11(11):1293‐303.

Treanor 2016

Treanor JJ. Influenza vaccination. New England Journal of Medicine 2016;375(13):1261‐8. [DOI: 10.1056/NEJMcp1512870]

Wijnans 2016

Wijnans L, Voordouw B. A review of the changes to the licensing of influenza vaccines in Europe. Viruses 2016;10(1):2‐8.

Wiselka 1994

Wiselka M. Influenza: diagnosis, management and prophylaxis. BMJ 1994;308:1341‐5.

Demicheli 1998

Demicheli V, Jefferson T. Influenza vaccines in healthy adults. Cochrane Database of Systematic Reviews 1998, Issue 4. [DOI: 10.1002/14651858.CD001269]

Demicheli 1999

Demicheli V, Rivetti D, Deeks JJ, Jefferson TO. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 1999, Issue 4. [DOI: 10.1002/14651858.CD001269]

Demicheli 2001

Demicheli V, Rivetti D, Deeks JJ, Jefferson TO. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 2001, Issue 4. [DOI: 10.1002/14651858.CD001269]

Demicheli 2004

Demicheli V, Rivetti D, Deeks JJ, Jefferson TO. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 2004, Issue 3. [DOI: 10.1002/14651858.CD001269.pub2]

Jefferson 2007

Jefferson TO, Rivetti D, Di Pietrantonj C, Rivetti A, Demicheli V. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 2007, Issue 2. [DOI: 10.1002/14651858.CD001269.pub3]

Jefferson 2010

Jefferson TO, Di Pietrantonj C, Rivetti A, Bawazeer GA, Al‐Ansary LA, Feroni E. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 2010, Issue 7. [DOI: 10.1002/14651858.CD001269.pub4]

Jefferson 2014

Jefferson T, Di Pietrantonj C, Rivetti A, Bawazeer GA, Al‐Ansary LA, Ferroni E. Vaccines for preventing influenza in healthy adults. Cochrane Database of Systematic Reviews 2014, Issue 3. [DOI: 10.1002/14651858.CD001269.pub5]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

aa Barrett 2011

Methods

Double‐blind, placebo‐controlled, multicentric RCT performed at 36 centres in the USA assessing effectiveness, reactogenicity, and antibodies responses of a Vero cell‐derived, trivalent, split influenza vaccine

Participants

Healthy adults aged 18 to 48 years recruited at 36 centres throughout the USA.

Individuals were excluded if they belonged to a CDC risk category for complications of influenza illness, had a history of surgical or functional asplenia, had been treated with any blood product or immune globulin in the previous 90 days, had a history of allergy to vaccine components, had received a live vaccine within 4 weeks or an inactivated vaccine within 2 weeks of study entry, or had dermatological disorders or tattoos that would obscure the assessment of injection‐site reactions. Individuals were not specifically excluded because of egg allergy. Immunisation in previous seasons was not judged to be an exclusion criterion.

Interventions

Inactivated, Vero cell‐derived, trivalent split influenza vaccine containing 15 µg haemagglutinin of the following strains, which were recommended by WHO for the season 2008 to 2009 in the Northern Hemisphere:

A‐H1N1: A/Brisbane/59/2007

A‐H3N2: A/Uruguay/716/2007 (A/Brisbane/10/2007‐like) (A/H3N2)

B: B/Florida/4/2006

The vaccine was manufactured by Baxter AG, Vienna. Vaccine strains were egg‐derived wild type strains provided by the National Institute for Biological Standard and Control. Placebo consisted of phosphate‐buffered saline.

Participants were randomly allocated to receive one 0.5 mL dose of either vaccine or placebo into the deltoid muscle. Vaccinations were performed between 1 and 15 December 2008.

Outcomes

Safety: participants were provided with a diary card, on which they had to record their temperature daily for the first 7 days following immunisation and to report fever and other adverse events for 21 days after immunisation. Participants returned for a final study visit 166 to 194 days after vaccination for a physical examination and final assessment of adverse events.

Serological: the first serum samples were presumably collected before vaccine administration (this is not well described in any of the 3 reports), and the second 18 to 24 days later. Haemagglutination‐inhibiting titres and GMT against vaccine strains were assessed by Focus Diagnostics (Cypress, CA, USA). Haemagglutination‐inhibiting assays were done in triplicate with egg‐derived antigen. Titres of less than 1:10 were expressed as 1:5 and judged to be negative.

Effectiveness: during the visit at days 18 to 24 after immunisation, participants were instructed to return to the clinic within 48 hours after the onset of symptoms of an influenza‐like illness, should they have fever with cough, sore throat, muscle ache, headache, fatigue, nausea, or bloodshot eyes, or any 2 of these symptoms without fever. At every visit for an influenza‐like illness until 15 May 2009, nasopharyngeal swabs were obtained for culturing and typing viruses.

Nasopharyngeal swab specimens were sent to BioAnalytical Research (Lake Success, NY, USA), for culture using Rapid R‐Mix (Diagnostic Hybrids, Athens, OH, USA) and traditional culture methods, and for virus typing with RT‐PCR analyses. Influenza type A/H1N1 or A/H3N2 isolates were sent to the laboratory of the Influenza Division, National Center for Immunization and Respiratory Diseases, CDC, Atlanta, GA, USA, for analyses of HI using ferret antiserum to assess the antigenic relatedness of the isolate to the vaccine strains.

Notes

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Individuals were randomly assigned by use of a centralised telephone system"

"Randomisation was done in blocks, with block sizes greater than two"

Allocation concealment (selection bias)

Low risk

"The allocation sequence was generated by Baxter, using an interactive voice response system with the random number generator algorithm of Wichmann and Hill, as modified by Mcleod"

Blinding (performance bias and detection bias)
All outcomes

Low risk

"At each study site, an investigator, subinvestigator, or study nurse who was masked to treatment allocation was designated to vaccinate participants, and was then prohibited from participation in data collection or the study. To ensure masking, the participants were enrolled by investigators who were not involved in the randomisation process.

Because the syringes containing the test and the control products were different in appearance both studies employed an observational blinding procedure such that study personnel who administered vaccinations were not involved in recording or reviewing study data"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Both efficacy and safety estimates were calculated on ITT study population. We know that all treated participants (3623 to influenza vaccine and 3620 to placebo) had been included in the safety analysis, whereas 3619 and 3617 had been considered for the effectiveness estimate calculation (i.e. those vaccinated and with at least 21 days' follow‐up after immunisation). Participants in the per‐protocol population (those who completed the study without major protocol deviations) were 3316 and 3318 in the vaccine and placebo arms, respectively.
Reasons for non‐inclusion in the per‐protocol population were not specified for 150 vaccine and 135 placebo recipients.

Summary assessment

Low risk

Low risk of bias

aa Beran 2009a

Methods

Randomised, double‐blind, placebo‐controlled study conducted in the Czech Republic during the 2005 to 2006 influenza season. This was defined retrospectively as starting the first week with 2 culture‐confirmed cases in the study area and ending the last week with 1 culture‐confirmed case in the study area. Randomisation was generated by GSK (sponsor) using the SAS program, in a 2:1 blocking scheme using a minimisation procedure (with no explanation of why such a method or the ratio was used). The allocation concealment method was not explicitly mentioned. However, the authors mentioned that placebo and vaccine treatments were indistinguishable in appearance and that blinding to treatment assignment was maintained until study analysis.

Participants

Self referred healthy adults (n = 6203), predominately Caucasian (understood to be white) (99.8%), aged between 18 and 64 years (mean 35 + 13 years) of both genders (TIV group: female 55.3%, placebo group: female 54.2%) and with no history of influenza vaccination within the last 3 influenza seasons. A subset of participants who were randomly selected for vaccine safety and reactogenicity were given a calibrated thermometer and a diary card to record symptoms. The method of selection of this subset was not explained. Use of antimicrobial/influenza antiviral therapy seemed to be allowed but was not quantified.

Interventions

TIV vaccine: 0.5 mL single dose by IM injection or placebo (normal saline). Use of more than 1 lot was not reported.

TIV contained haemagglutinin antigens of:

  • A/New Caledonia/20/99 (H1N1) IVR‐116 virus as an A/New Caledonia/20/99‐like strain;

  • A/New York/55/2004 (H3N2) X‐157 virus as an A/California/7/2004‐like strain;

  • B/Jiangsu/10/2003 virus as a B/Shanghai/361/2002‐like strain.

2 modes of surveillance were used.

  • Passive: started on the day of vaccination, participants self report of ILI symptoms through a toll‐free number.

  • Active: started 2 weeks after vaccination day: a biweekly telephone contact of the participants by someone (not clear who) for ILI symptoms.

  • It is not clear if the surveillance included the entire cohort or just a subset, or why the authors carried out harms surveillance using the 2 surveillance methods already in place.

Outcomes

Serological

Blood samples were collected for the specified subset and were tested/analysed at GSK Biologicals SSW Dresden, Germany.

Blood sample obtained prior to vaccination and at 21 days following vaccination. Serum samples were stored at ‐20 °C until blinded analyses were conducted.

A haemagglutination‐inhibition test was done using chicken red blood cells with the 3 virus strains present in the TIV used as antigens. The serum titre was expressed as the reciprocal of the highest dilution that showed complete inhibition of haemagglutination.

Serology was not a primary outcome in this study.

Effectiveness

Incidence of culture‐confirmed ILI (primary outcome, reported as the attack rate in the efficacy cohort)

Nasal and throat swab collected by a nurse on the same day.

Swab samples were stored at 28 °C and transferred within 5 days of the onset of ILI symptoms.

Sample sent to the National Reference Laboratory for Influenza (NRL, Prague, Czech Republic) for conventional influenza virus culture using MDCK cells.

Confirmation of influenza A or B was determined using the following:

  • haemagglutination assay with turkey and guinea pig erythrocytes;

  • haemagglutination inhibition to identify virus type, subtype, and drift variant;

  • direct immunoperoxidase assay using anti‐influenza A and anti‐influenza B nucleoprotein antibodies.

There were 814 reported ILI episodes, only 46 gave positive culture. 

Clinical

Incidence of ILI symptoms (secondary outcome, reported as attack rate in the ATP cohort)

Influenza‐like illness was defined as fever (oral temperature greater or equal to 37.8 °C) plus cough and/or sore throat. An ILI episode was defined as the period from the first day of ILI symptoms until the last day of ILI symptoms. A new episode was taken into account only after the complete resolution of the previous one. To count as a separate episode at least 7 days free of any symptoms should pass.

Number of events was 370 reported events (254 in TIV and 120 in placebo).

Number of participants reporting at least 1 event (240 in TIV and 113 in placebo) was used to calculate the attack rate.

Reasons to exclude from the ATP cohort included:

  • protocol violation (inclusion/exclusion criteria): seems that the selected subset have certain criteria but not mentioned by the authors;

  • underlying medical condition: not specified what? Or why not excluded from the efficacy cohort as well since participants are reported to be healthy;

  • forbidden by the protocol: protocol not clear;

  • participants not exposed during the influenza season: unclear what this means (did the participant travel after getting the study treatment?).

Immunogenicity

Blood sample obtained prior to vaccination and at 21 days following vaccination. Performed only for a subset of participants, not all efficacy cohort.

Safety

Data on SAEs began at the receipt of vaccine/placebo and continued until the end of the study. However, safety was solicited from a subset of participants (no mention of method used to randomly select them, no justification for not collecting SAEs from all participants, especially with the presence of 2 surveillance methods).

Reactogenicity

Defined as the presence and intensity of the following symptoms within 4 days of vaccination: pain, redness, and swelling (found to occur more in the TIV group), other general symptoms of fatigue, fever, headache, muscle aches, shivering, and joint pain (found to occur more in the TIV group).

The intensities of adverse events were recorded according to a standard 0 to 3 grade scale: "absent", "easily tolerated", "interferes with normal activity", and "prevents normal activity".

Notes

The authors report that due to the atypical nature of the influenza season during this study they were unable to assess TIV efficacy.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A randomisation list was generated by the sponsor by SAS program and used to number the vaccine and placebo treatments"; "A randomization blocking scheme (2:1) was employed to ensure that balance between treatments was maintained."

Allocation concealment (selection bias)

Unclear risk

No explicit description of the method of concealment, authors only mentioned that treatments were numbered and that they were indistinguishable in appearance.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Authors reported that the blinding assignment was maintained until study analysis.

Authors mentioned that the treatments were indistinguishable in appearance.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Exclusion of allocated participants from the analysis of the trial:

a) did the report mention explicitly the exclusion of allocated participants from the analysis of trial results? Yes;

b) if so did the report mention the reason(s) for exclusion? Yes. Details were reported in the study flow chart.

Summary assessment

Unclear risk

Unclear risk of bias

aa Beran 2009b

Methods

A randomised, double‐blind, placebo‐controlled study conducted during the 2006 to 2007 influenza season at 15 centres located in the Czech Republic and Finland. The protocols and study documents were approved by the ethics committee of each country. Participants were randomised to receive 1 dose of TIV (lot 1 or lot 2 of Fluarix) or placebo (normal saline solution) at the first study visit (day 0) by intramuscular injection. Each 0.5 mL dose of TIV contained 15 mg of each of the haemagglutinin antigens of strains A/New Caledonia/20/99(H1N1) IVR‐116, A/Wisconsin/67/2005(H3N2), and B/Malaysia/2506/2004 (from the Victoria lineage).

From the day of vaccination, passive and active surveillance (biweekly contact) to detect ILI cases. For each case of suspected ILI, a nasal and throat swab specimen (composed of a swab of both nasal sinuses and a second swab of the throat) was collected for culture (as much as possible on the same day as the ILI report and, at the latest, 5 days after the ILI onset). Each participant was provided with a calibrated thermometer to measure temperature and a diary card to record temperatures and symptoms during the ILI episode. Blinded analysis was carried out at GSK Biologicals in Dresden, Germany.

Blood samples for the evaluation of influenza vaccine immunogenicity were obtained from the randomly selected, planned subset of an estimated 500 participants just prior to vaccination and 21 to 28 days later. Frozen aliquots of culture supernatants from positive viral cultures were sent to J Treanor's laboratory (University of Rochester Vaccine Evaluation Unit Influenza Serology Laboratory, Rochester, NY, USA) for identification of virus‐matching isolates by conventional haemagglutination‐inhibition testing (using H1 and H3 antisera from the CDC and B/Malaysia antiserum from the WHO).

Participants

Eligible participants were self referred women or men who were between 18 and 64 years of age and had no significant clinical disease at the time of vaccination.

WHO provided written informed consent.

Interventions

Intervention 1 dose of TIV (lot 1 or lot 2 of Fluarix), IM injection, at the first day of the study (day 0)
Each 0.5 mL dose of TIV contained 15 mg of each of the haemagglutinin antigens of strains A/New/Caledonia/20/99(H1N1) IVR‐116, A/Wisconsin/67/2005(H3N2), and B/Malaysia/2506/2004 (from the Victoria lineage).

Comparator placebo (normal saline solution), IM injection, at the first day of the study (day 0)

Outcomes

Serological (only carried out for the TIV group)

Effectiveness 

Evaluate efficacy of TIV versus placebo in the prevention of culture‐confirmed influenza A and/or B due to strains antigenically matched to the vaccine (their primary objective)

Secondary objectives were evaluation of TIV in the prevention of:

  • culture‐confirmed influenza due to strains antigenically matched to the vaccine for each of the 2 vaccine lots;

  • culture‐confirmed influenza A and/or B attributable to any influenza A or B strain;

  • ILI, which was less stringently defined as at least 1 systemic symptom (fever or myalgia, or both) and 1 respiratory symptom (cough or sore throat, or both).

Safety vaccine reactogenicity and immunogenicity in a random subset of participants by obtaining blood samples prior to vaccination and 21 to 28 days later. However, no harms data were reported.

Notes

The authors concluded that TIV is efficacious against culture‐confirmed influenza in healthy adults.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided.

Allocation concealment (selection bias)

Unclear risk

No details provided.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

There is no mention of appearance of the injection content.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition reasons for the whole cohort are provided by the participant flow.

Summary assessment

Unclear risk

Unclear risk of bias

aa Bridges 2000a

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1997 to 1998 influenza season. Follow‐up lasted from November to March. Influenza period was defined as the period during which clinical specimens collected from ill participants yielded influenza viruses (8 December 1997 through 2 March 1998) and lasted 12 weeks. Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers. Pharyngeal swab and paired sera were collected from ill people.

Participants

1184 healthy factory employees: 595 treated and 589 placebo. Age of participants was 18 to 64.

Interventions

Commercial trivalent, inactivated, intramuscular vaccine. Schedule and dose were not indicated. Vaccine composition was: A/Johannesburg/82/96, A/Nanchang/933/95, and B/Harbin/7/94. Placebo was sterile saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Influenza‐like illness, influenza, days ill, physician visits, times any drug was prescribed, times antibiotic was prescribed, working days lost, admissions, adverse effects. They were defined as follows: influenza‐like illness: fever = 37.7 °C with cough or sore throat); upper respiratory illness: cough with sore throat or fever = 37.7 °C. Local adverse effects were arm soreness and redness. Systemic adverse effects were: fever, sore throat, coryza, myalgia, headache, and fatigue, but authors reported no data. Surveillance was passive.

Notes

For analysis we chose the influenza‐like illness definition. Intention‐to‐treat analysis was performed. Systemic adverse effects were not reported. Circulating strain was A/Sidney/5/97‐like.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient description

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition reasons for the whole cohort are provided by the participant flow.

Summary assessment

Low risk

Low risk of bias

aa Bridges 2000b

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1998 to 1999 influenza season. Follow‐up lasted from November to March. The influenza period was defined as the period during which clinical specimens collected from ill participants yielded influenza viruses (4 January 1998 through 14 March 1999) and lasted 10 weeks. Pharyngeal swabs and paired sera were collected from ill people.

Participants

1191 healthy factory employees: 587 treated and 604 placebo. Age of participants was 19 to 64.

Interventions

Commercial trivalent, inactivated, intramuscular vaccine. Schedule and dose were not indicated. Vaccine composition was: A/Beijing/262/95, A/Sydney/5/97, and B/Harbin/7/94. Placebo was sterile saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Influenza‐like illness, influenza, days ill, physician visits, times any drug was prescribed, times antibiotic was prescribed, working days lost, admissions, adverse effects. They were defined as follows: influenza‐like illness: fever = 37.7 °C with cough or sore throat; upper respiratory illness: cough with sore throat or fever = 37.7 °C. Local adverse effects were arm soreness and redness. Systemic adverse effects were: fever, sore throat, coryza, myalgia, headache, and fatigue, but authors reported no data. Surveillance was passive.

Notes

For analysis we chose the influenza‐like illness definition. Intention‐to‐treat analysis was performed. Systemic adverse effects were not reported. Circulating strain was A/Sydney/5/97‐like and B/Beijing/184/93‐like.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers.

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Placebo was sterile saline for injection. Probably adequate

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition reasons for the whole cohort are provided by the participant flow.

Summary assessment

Low risk

Low risk of bias

aa Eddy 1970

Methods

Controlled clinical trial, single‐blind, conducted in South Africa during the 1969 influenza season. Follow‐up lasted from May to July. The first clinical case of influenza appeared on 21 May 1969 and the last 6 weeks later. The epidemic period lasted 6 weeks. The control participants were selected by drawing a 1‐in‐4 systematic sample from a ranked list of the personnel numbers.

Participants

1758 healthy male black African employees: 1254 treated and 413 placebo. Age of participants was 18 to 65.

Interventions

Monovalent inactivated parenteral vaccine. Schedule and dose were single injection, 1 mL. Vaccine composition was: A2/Aichi/2/68 (Hong Kong variant). Placebo was sterile water. Vaccine was recommended and matched circulating strain.

Outcomes

Influenza‐like illness, working days lost, days ill. Influenza‐like illness was not defined; case features were generically described in results section. All ill people were admitted to hospital until recovery. Surveillance was passive.

Notes

The word 'double‐blinding' was not used, but the control group received an injection of "dummy vaccine". Poor reporting, poor‐quality study. Circulating strain was A2/Hong Kong/68 virus.
Efficacy data only were extracted.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Systematic selection

Allocation concealment (selection bias)

High risk

Inadequate

Blinding (performance bias and detection bias)
All outcomes

High risk

No descriptions

Incomplete outcome data (attrition bias)
All outcomes

High risk

Insufficient description

Summary assessment

High risk

High risk of bias

aa Edwards 1994a

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1986 to 1987 influenza season. Follow‐up lasted the whole epidemic period. The epidemic period in any study year started on the day that the first influenza A virus isolate was obtained in Nashville and ended on the day that the last isolate was obtained and lasted 8 weeks. Participants were recruited from 7 organisations and assigned to 1 of the study groups using a permuted block randomisation scheme that was stratified by treatment centre and age group. Sealed randomisation envelopes contained vaccine codes. Pharyngeal swab and paired sera were collected from ill people.

Participants

1311 healthy children and adults of metropolitan Nashville: 872 treated and 439 placebo. Age of participants was 1 to 65. 85% of participants were older than 16.

Interventions

Bivalent, live, cold‐adapted, aerosol‐administered influenza A vaccine and the commercial inactivated intramuscularly administered influenza vaccine. Schedule and dose were: single‐dose; cold‐adapted 107 to 107.6 pfu/mL; inactivated 15 µg each strain. Vaccine composition was: cold‐adapted: Texas/1/85 H1N1 and Bethesda/1/85 H3N2; inactivated: Chile/1/83 H1N1 and Mississippi/1/85 H3N2. Placebo was allantoic fluid. Vaccine was recommended but did not match circulating strain.

Outcomes

Influenza‐like illness, influenza. They were defined as follows: fever of abrupt onset with at least 1 of the following: chills, headache, malaise, myalgia, cough, pharyngitis, or other respiratory complaints (only participants who presented for culture were considered); throat culture. Surveillance was passive.

Notes

Influenza B strain contained in the commercial and monovalent vaccines was not described. Strains used yearly to develop cold‐adapted and inactivated vaccines were antigenically comparable. Since cold‐adapted influenza B vaccines were not sufficiently characterised to include in the study, the authors used monovalent inactivated influenza B vaccine in all participants in the cold‐adapted arm and as placebo in the control group inactivated arm. Only the cold‐adapted comparison was included in the analysis. The circulating strain was Taiwan/1/86. Effectiveness data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient description: "permutated block randomization scheme that was stratified by treatment centre and age group"

Allocation concealment (selection bias)

Low risk

Adequate: participants and clinical staff were kept unaware of the assigned vaccine group through the use of sealed randomisation envelopes that contained vaccines codes.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

Insufficient description

aa Edwards 1994b

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1987 to 1988 influenza season. Follow‐up lasted the whole epidemic period. The epidemic period in any study year started on the day that the first influenza A virus isolate was obtained in Nashville and ended on the day that the last isolate was obtained and lasted 14 weeks. Participants were recruited from 7 organisations and assigned to 1 of the study groups using a permuted block randomisation scheme that was stratified by treatment centre and age group. Sealed randomisation envelopes contained vaccine codes. Pharyngeal swab and paired sera were collected from ill people.

Participants

1561 healthy children and adults of metropolitan Nashville: 1029 treated and 532 placebo. Age of participants was 1 to 65. 85% of participants were older than 16.

Interventions

Bivalent, live, cold‐adapted, aerosol‐administered influenza A vaccine and the commercial inactivated intramuscularly administered influenza vaccine. Schedule and dose were: single dose; cold‐adapted 107 to 107.6 pfu/mL; inactivated 15 µg each strain. Vaccine composition was: cold‐adapted: Kawasaki/9/86 H1N1 and Bethesda/1/85 H3N2; inactivated: Taiwan/1/86 H1N1 and Leningrad/360/86 H3N2. Placebo was allantoic fluid. Vaccine was recommended but did not match the circulating strain.

Outcomes

Influenza‐like illness, influenza. They were defined as follows: fever of abrupt onset with at least 1 of the following: chills, headache, malaise, myalgia, cough, pharyngitis, or other respiratory complaints (ILI symptoms retrospectively reported were considered); 4‐fold antibody rise between postvaccination and spring sera. Surveillance was passive.

Notes

Influenza B strain contained in the commercial and monovalent vaccines was not described. Strains used yearly to develop cold‐adapted and inactivated vaccines were antigenically comparable. Since cold‐adapted influenza B vaccines were not sufficiently characterised to include in the study, the authors used monovalent inactivated influenza B vaccine in all participants in the cold‐adapted arm and as placebo in the control group inactivated arm. Only the cold‐adapted comparison was included in the analysis. The circulating strain was Sichuan/2/87 (H3N2) (antigen drift from vaccine strain) and B/Victoria/2/87.
Effectiveness data only were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient description: "permutated block randomization scheme that was stratified by treatment centre and age group"

Allocation concealment (selection bias)

Low risk

Adequate: participants and clinical staff were kept unaware of the assigned vaccine group through the use of sealed randomisation envelopes that contained vaccines codes.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

Insufficient description

Summary assessment

Unclear risk

Unclear

aa Edwards 1994c

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1988 to 1989 influenza season. Follow‐up lasted the whole epidemic period. The epidemic period in any study year started on the day that the first influenza A virus isolate was obtained in Nashville and ended on the day that the last isolate was obtained and lasted 11 weeks. Participants were recruited from 7 organisations and assigned to 1 of the study groups using a permuted block randomisation scheme that was stratified by treatment centre and age group. Sealed randomisation envelopes contained vaccine codes. Pharyngeal swab and paired sera were collected from ill people.

Participants

1676 healthy children and adults of metropolitan Nashville: 1114 treated and 562 placebo. Age of participants was 1 to 65. 85% of participants were older than 16.

Interventions

Bivalent, live, cold‐adapted, aerosol‐administered influenza A vaccine and the commercial inactivated intramuscularly administered influenza vaccine. Schedule and dose were: single dose; cold‐adapted 107 to 107.6 pfu/mL; inactivated 15 µg each strain. Vaccine composition was: cold‐adapted: Kawasaki/9/86 H1N1 and Los Angeles/2/87 H3N2; inactivated: Taiwan/1/86 H1N1 and Sichuan/2/87 H3N2. Placebo was allantoic fluid. Vaccine was recommended and matched circulating strain.

Outcomes

Influenza‐like illness, influenza. They were defined as follows: fever of abrupt onset with at least 1 of the following: chills, headache, malaise, myalgia, cough, pharyngitis, or other respiratory complaints (ILI symptoms retrospectively reported were considered); 4‐fold antibody rise between postvaccination and spring sera. Surveillance was passive.

Notes

Influenza B strain contained in the commercial and monovalent vaccines was not described. Strains used yearly to develop cold‐adapted and inactivated vaccines were antigenically comparable. Since cold‐adapted influenza B vaccines were not sufficiently characterised to include in the study, the authors used monovalent inactivated influenza B vaccine in all participants in the cold‐adapted arm and as placebo in the control group inactivated arm. Only the cold‐adapted comparison was included in the analysis. The circulating strain was Taiwan/1/86 (H1N1) and B/Yamata/16/88. Effectiveness data only were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient description: "permutated block randomization scheme that was stratified by treatment centre and age group"

Allocation concealment (selection bias)

Low risk

Adequate: participants and clinical staff were kept unaware of the assigned vaccine group through the use of sealed randomisation envelopes that contained vaccines codes.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

Insufficient description

Summary assessment

Unclear risk

Unclear

aa Edwards 1994d

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1989 to 1990 influenza season. Follow‐up lasted the whole epidemic period. The epidemic period in any study year started on the day that the first influenza A virus isolate was obtained in Nashville and ended on the day that the last isolate was obtained and lasted 11 weeks. Participants were recruited from 7 organisations and assigned to 1 of the study groups using a permuted block randomisation scheme that was stratified by treatment centre and age group. Sealed randomisation envelopes contained vaccine codes. Pharyngeal swab and paired sera were collected from ill people.

Participants

1507 healthy children and adults of metropolitan Nashville: 999 treated and 508 placebo. Age of participants was 1 to 65. 85% of participants were older than 16.

Interventions

Bivalent, live, cold‐adapted, aerosol‐administered influenza A vaccine and the commercial inactivated intramuscularly administered influenza vaccine. Schedule and dose were: single dose; cold‐adapted 107 to 107.6 pfu/mL; inactivated 15 µg each strain. Vaccine composition was: Kawasaki/9/86 H1N1 and Los Angeles/2/87 H3N2; inactivated: Taiwan/1/86 H1N1 and Shanghai/11/87 H3N2. Placebo was allantoic fluid. Vaccine was recommended and matched circulating strain.

Outcomes

Influenza‐like illness, influenza. They were defined as follows: fever of abrupt onset with at least 1 of the following: chills, headache, malaise, myalgia, cough, pharyngitis, or other respiratory complaints (ILI symptoms retrospectively reported were considered); 4‐fold antibody rise between postvaccination and spring sera. Surveillance was passive.

Notes

Influenza B strain contained in the commercial and monovalent vaccines was not described. Strains used yearly to develop cold‐adapted and inactivated vaccines were antigenically comparable. Since cold‐adapted influenza B vaccines were not sufficiently characterised to include in the study, the authors used monovalent inactivated influenza B vaccine in all participants in the cold‐adapted arm and as placebo in the control group inactivated arm. Only the cold‐adapted comparison was included in the analysis. The circulating strain was Shanghai/11/87 (H3N2). Effectiveness data only were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient description: "permutated block randomization scheme that was stratified by treatment centre and age group"

Allocation concealment (selection bias)

Low risk

Adequate: participants and clinical staff were kept unaware of the assigned vaccine group through the use of sealed randomisation envelopes that contained vaccines codes.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

Insufficient description

Summary assessment

Unclear risk

Unclear

aa Frey 2010

Methods

Randomised, controlled, multicentre, observer‐blind trial assessing effectiveness, immunogenicity, and safety of both CCIV and TIV containing the strain recommended by WHO for the current season (2007 to 2008)

Participants

Participants were recruited at 56 centres in the USA, Finland, and Poland.
Major exclusion criteria: health condition for which inactivated vaccine is recommended, employment prone to influenza transmission, influenza vaccination or laboratory‐confirmed influenza within 6 months of enrolment, history of Guillain‐Barré syndrome, a temperature of 37.8 °C and/or acute illness within 3 days of enrolment, and pregnancy or breastfeeding.
A total of 11,404 participants were randomised: 11,382 were vaccinated and 10,844 (95%) completed the study.

Interventions

Individuals aged 18 to 49 years were randomised equally, with use of an interactive voice response system, to receive a single dose of CCIV, TIV, or placebo.
Both CCIV and TIV (Novartis Vaccines and Diagnostics) contained 15 µg of haemagglutinin per 0.5 mL dose of each of the following virus strains:
A/Solomon Islands/3/2006 (H1N1)–like
A/Wisconsin/67/2005 (H3N2)–like
B/Malaysia/2506/2004–like
Preparations were administered in the deltoid muscle of the non‐dominant arm. Only the vaccine administrator had access to the randomisation code.

Outcomes

Safety
Study participants were monitored for 30 minutes after vaccination for immediate reactions. Participants recorded the occurrence, duration, and severity of local injection site and systemic reactions for 7 days after vaccination. Solicited reactions were graded as follows: mild, no limitation of normal daily activities; moderate, some limitation; or severe, unable to perform normal daily activities. Unsolicited reactions were recorded for 21 days after vaccination. Serious adverse events were monitored for the entire study (9 months).
Effectiveness
Influenza surveillance began 21 days after vaccination. Participants had to report to investigators the occurrence of influenza‐like illness symptoms (fever 37.8 °C plus sore throat or cough, as well as body aches, chills, headache, and runny or stuffy nose). An active survey was also performed by means of weekly phone calls.
Participants reporting influenza‐like illness symptoms underwent clinical evaluations; nasal and throat specimens were obtained for laboratory confirmation of influenza virus. Specimens were targeted for collection within 24 hours after symptom onset, with a window of 120 hours. Specimens were cultured on RhMK and tested by PCR.
Each study participant was observed during the 6‐month study surveillance period or for 6 months after vaccination, whichever was longer. Study duration was around 9 months.
Immunogenicity
It was assessed on the first 1045 participants enrolled at USA sites and randomised 8:25:2 to receive CCIV, TIV, or placebo. Serum samples were collected at baseline and 3 weeks after immunisation for seroprotection, seroconversion, and GMT determination.

Notes

Financial support: "Novartis Vaccines was the funding source and was involved in all stages of the study conduct and analysis"
Potential conflicts of interest: "M.L., A.I., N.G., and S.H. are employees of Novartis Vaccines and Diagnostics. T.V. has received consultancy fees from MedImmune and speaker fees from MedImmune, Novartis, and Crucell in relation to meetings on influenza vaccination. S.F. and A.S.‐M.: no conflicts"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Low risk

“Individuals ... were randomised equally, with use of an interactive voice response system, to receive a single dose of CCIV, TIV, or placebo.”

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

“This randomized, placebo‐controlled, observer‐blind trial evaluated ...”
“Only the vaccine administrator had access to the randomization code.”
No information about the appearance of the preparation is provided in the text.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Flow of participants during the study is reported and described. Loss to follow‐up amounts to about 5% at study end and is balanced through the 3 arms.

Summary assessment

Unclear risk

Unclear

aa Hammond 1978

Methods

Controlled clinical trial, double‐blinded conducted in Australia during the 1976 influenza season. Follow‐up lasted the whole epidemic period. Epidemic influenza was defined by virus isolation and serology tests and lasted from middle of April to middle of August 1976 (17 weeks). Coded, identical‐looking vials were sequentially administered to enrolled participants. A throat swab was collected from ill people. Serological confirmation was performed on all participants.

Participants

225 medical students or staff members: 116 treated and 109 placebo. Age of participants was not indicated.

Interventions

Trivalent parenteral subunit vaccine. Schedule and dose were: single dose; vaccine composition was: 250 IU of A/Victoria/3/75, 250 IU of A/Scotland/840/74, and 300 IU of B/Hong Kong/8/73. Placebo was diphtheria and tetanus toxoids. Vaccine was recommended and matched circulating strain.

Outcomes

Influenza‐like illness, influenza. Clinical illnesses were not defined. Influenza was defined as respiratory illness that was associated with the isolation of influenza virus, a 4‐fold or greater rise in antibody titre occurring between postvaccination and postepidemic sera, or both. Surveillance was active.

Notes

Clinical illness was not defined, and data were included in the analysis as "clinical cases without clear definition". Circulating strain was A/Vic/3/75‐like. Efficacy data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Alternate

Allocation concealment (selection bias)

High risk

No description

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No description

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description

Summary assessment

High risk

No description

aa Jackson 2010a

Methods

Randomised, multicentre, double‐blind, placebo‐controlled trial assessing the effectiveness and safety of a trivalent inactivated vaccine in preventing confirmed influenza. The study was performed during 2 influenza seasons (2005 to 2006 and 2006 to 2007) in the USA.

Participants

Healthy adults aged between 18 and 49 years without significant acute or chronic medical or psychiatric illness. Individuals with cancer; systolic blood pressure ≥ 140 mmHg, diastolic blood pressure ≥ 90 mmHg; belonging to a risk group for which routine influenza vaccination is recommended (chronic pulmonary, cardiovascular, renal, hepatic, haematological, or metabolic disorders; immunosuppressive illness, recent/ongoing receipt of immunosuppressive therapy, immunoglobulin, other vaccines, or with HIV infection were excluded. Participants enrolled for the first season were not included in the second season.
In season I (2005 to 2006), 3514 participants were recruited at 37 centres from 17 September 2005 onwards.
In season II (2006 to 2007), 4144 participants were recruited at 44 centres from 16 October 2006 onwards.

Interventions

Recruited participants were randomised at the beginning of each season to receive 1 dose of trivalent inactivated split influenza vaccine (FluLaval, a trademark of the GlaxoSmithKline group of companies; manufactured by ID Biomedical Corporation of Quebec, Canada) or saline placebo injection.
Each 0.5 mL dose of TIV contained 15 μg of HA antigen of each recommended influenza strain.
For season I (2005 to 2006) antigens were:
A/New Caledonia/20/1999 (H1N1)
A/New York/55/2004 (H3N2, A/California/7/2004‐like)
B/Jiangsu/10/2003 (B/Shanghai/361/2002‐like)

Outcomes

Effectiveness
During the influenza seasons, participants were instructed to report symptoms meeting the ILI definition by using a toll‐free, study‐specific phone number within 48 hours from their onset and to record them together with temperature. Influenza‐like illness symptoms were moreover solicited by weekly outbound phone contact. Visits from nurses were dispatched to participants who filled ILI definition within 24 hours after symptoms onset, and nasopharyngeal and oropharyngeal swabs for viral culture were drawn. During season I surveillance for influenza was conducted between 14 November 2005 and 30 April 2006; during season II between 13 November and 30 April.

Primary effectiveness study endpoint was:
VMCCI (vaccine‐matched, culture‐confirmed influenza). The case definition required the presence of ILI, defined as symptoms that interfered with normal daily activities and that included cough and at least 1 additional symptom from among fever (oral temperature > 37.7 °C/99.9 °F), headache, myalgia and/or arthralgia, chills, rhinorrhoea/nasal congestion, and sore throat. Participants meeting the definition for ILI and with concurrent isolation from a nasopharyngeal swab of an influenza A and/or B virus isolate antigenically matching a vaccine strain for the relevant year were considered to be cases of VMCCI.
Secondary effectiveness endpoints were:
CCI (culture‐confirmed influenza illness). ILI with any influenza A or B virus isolate by culture.
LCI (laboratory‐confirmed influenza illness). 1 or both of CCI or ILI with a 4‐fold increase in HI serum antibody titres to a circulating influenza virus strain between day 21 (±4 days) postvaccination and final visit specimens obtained after the end of the influenza season.

Immunogenicity
Serum samples were collected from study participants at day 0, 21, and about 4 weeks after the end of the surveillance period.
Immunogenicity was assessed determining GMT, seroconversion and seroprotection rate between samples collected at day 21 and at day 0 on a randomly selected subset of participants.

Safety

Local and systemic reactions (events) occurred within 3 days after immunisation. Participants were observed for the first 30 minutes following immunisation. Participants recorded further reactions occurring no later than 8 days following vaccination by means of an interactive voice response system. The following symptoms were reported (3 days).

  • Fever (at least 37.5 °C)

  • Injection site pain/soreness

  • Injection site redness

  • Injection site swelling

  • Myalgia or arthralgia, or both

  • Headache

  • Tiredness

  • Chills

  • Malaise

  • Red eyes

  • Swelling of the face

  • Cough

  • Chest tightness or difficulty in breathing

  • Sore throat, hoarseness, or pain on swallowing

Participants with at least 1 vaccine reactogenicity event
Data were provided pooled for the 2 study seasons.
Unsolicited spontaneous adverse events, for which follow‐up was extended for at least 135 days following immunisation.
Pregnancy outcomes
Pregnancies
Spontaneous abortion
Full‐term birth

Notes

Per‐protocol: participants who received the treatment to which they were randomised, responded to ≥ 1 postvaccination active surveillance telephone calls, and had no major protocol deviations considered to affect the efficacy or immunogenicity data (determined before unblinding) (for effectiveness estimates).
Intention‐to‐immunise: the per‐protocol set plus participants with protocol deviations and treatment errors and analysed as randomised.
The safety set included participants who received any study treatment and had any postvaccination safety data. If an incorrect treatment was conclusively documented, participants in the safety set were analysed based on the treatment they had actually received.

Funding source was pharmaceutical.
"GSK Biologicals was the funding source and was involved in all stages of the study conduct and analysis. GSK Biologicals also took in charge all costs associated with the development and the publishing of this manuscript. The corresponding author had full access to the data, and final responsibility for submission of the manuscript for publication"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Treatment allocation was determined by blocked, stratified randomization with a 1:1 distribution to TIV or placebo; randomization was stratified by study center, age (18‐34 and 35‐49 years), and the subject's report of previous recent receipt (within ≤ 2 years) of TIV.”

Allocation concealment (selection bias)

Unclear risk

Insufficient description of allocation concealment: “Each study center had a pre‐determined sequence of randomization numbers which were allocated sequentially to eligible participants. Participants were allocated equally among 3 different vaccine lots”

Blinding (performance bias and detection bias)
All outcomes

Low risk

“Clinic staff (excluding the nurse giving the vaccine), were blinded to the treatment group until the study was complete.”

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Participant flow

Summary assessment

Unclear risk

Unclear

aa Jackson 2010b

Methods

See aa Jackson 2010a (the following data refer to the second study season)

Participants

In season II (2006 to 2007), 4144 participants were recruited at 44 centres from 16 October 2006 onwards.

Interventions

Recruited participants were randomised at the beginning of each season to receive 1 dose of trivalent inactivated split influenza vaccine (FluLaval, a trademark of the GlaxoSmithKline group of companies; manufactured by ID Biomedical Corporation of Quebec, Canada) or saline placebo injection.
Each 0.5 mL dose of TIV contained 15 μg of haemagglutinin antigen of each recommended influenza strain
Antigens for season II (2006 to 2007) were:
A/New Caledonia/20/1999 (H1N1) virus
A/Wisconsin/67/2005 (H3N2)
B/Malaysia/2506/2004

Outcomes

See aa Jackson 2010a

Notes

See aa Jackson 2010a

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

See aa Jackson 2010a

Allocation concealment (selection bias)

Unclear risk

See aa Jackson 2010a

Blinding (performance bias and detection bias)
All outcomes

Low risk

See aa Jackson 2010a

Incomplete outcome data (attrition bias)
All outcomes

Low risk

See aa Jackson 2010a

Summary assessment

Unclear risk

See aa Jackson 2010a

aa Keitel 1988a

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1983 to 1984 influenza season. Follow‐up lasted the whole epidemic period. Influenza period was defined as the interval during which community surveillance recovered influenza viruses from 10% or more of people with febrile respiratory illness per calendar week (from 8 January to 17 March 1984) and lasted 9 weeks. Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers according to prior vaccination experience. Specimens for culture and acute‐convalescent blood specimens were obtained from ill people. At spring time volunteers were asked to record any illness that occurred during the epidemic period, and blood specimens were collected.

Participants

598 healthy employees working in the Texas Medical Center in Houston, Texas, or in surrounding industrial companies: 300 treated and 298 placebo. Age of participants was 30 to 60.

Interventions

Trivalent, killed, whole, intramuscularly administered vaccine. Schedule and dose were: single dose; 15 µg of haemagglutinin of each influenza strain. Vaccine composition was: A/Philippines/2/82 (H3N2), A/Brazil/11/78 (H1N1), and B/Singapore/222/79. Placebo was sterile saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Outcomes were: ILI, influenza. Illnesses were classified as "any", "flu‐like" (lower respiratory or systemic illness, or both), and "febrile" (oral temperature of 37.8 °C or higher). Laboratory confirmation was based on culture and/or 4‐fold or greater rise in antibody titre occurring between postvaccination (pre‐epidemic), acute, convalescent and/or spring (postepidemic) sera.

Notes

Influenza‐like illness and influenza were detected in 3 groups: first vaccinated, multivaccinated, and placebo. Febrile illnesses were included in the analysis; the first 2 groups' cases were combined. Circulating strain was A/Victoria/7/83 (H1N1) and B/USSR/100/83. Efficacy data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No description

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description

Summary assessment

Unclear risk

No description

aa Keitel 1988b

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1984 to 1985 influenza season. Follow‐up lasted the whole epidemic period. The influenza period was defined as the interval during which community surveillance recovered influenza viruses from 10% or more of people with febrile respiratory illness per calendar week (from 6 January to 9 March 1985) and lasted 9 weeks. Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers according to prior vaccination experience. Specimens for culture and acute‐convalescent blood specimens were obtained from ill people. At spring time volunteers were asked to record any illness that occurred during the epidemic period, and blood specimens were collected.

Participants

697 healthy employees working in the Texas Medical Center in Houston, Texas, or in surrounding industrial companies: 456 treated and 241 placebo. Age of participants was 30 to 60.

Interventions

Trivalent, killed, whole, intramuscularly administered vaccine. Schedule and dose were: single dose; 15 µg of haemagglutinin of each influenza strain. Vaccine composition was: A/Philippines/2/82 (H3N2), A/Chile/1/83 (H1N1), and B/USSR/100/83. Placebo was sterile saline for injection.

Outcomes

Outcomes were: ILI, influenza. Illnesses were classified as "any", "flu‐like" (lower respiratory or systemic illness, or both), and "febrile" (oral temperature of 37.8 °C or higher). Laboratory confirmation was based on culture and/or 4‐fold or greater rise in antibody titre occurring between postvaccination (pre‐epidemic), acute, convalescent and/or spring (postepidemic) sera. Surveillance was passive.

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No description

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description

Summary assessment

Unclear risk

No description

aa Keitel 1997a

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1985 to 1986 influenza season. Follow‐up lasted the whole epidemic period. The influenza period was defined by viral surveillance. Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers according to prior vaccination experience. Specimens for culture and acute‐convalescent blood specimens were obtained from ill people. At spring time, volunteers were asked to record any illness that occurred during the epidemic period, and blood specimens were collected.

Participants

830 healthy employees working in the Texas Medical Center in Houston, Texas, or in surrounding industrial companies: 577 treated and 253 placebo. Age of participants was 30 to 60.

Interventions

Trivalent, killed, whole, intramuscularly administered vaccine. Schedule and dose were: single dose; 15 µg of haemagglutinin of each influenza strain. Vaccine composition was: A/Philippines/2/82 (H3N2), A/Chile/1/83 (H1N1), and B/USSR/100/83. Placebo was sterile saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

ILI, influenza. Illnesses were classified as "any", "flu‐like" (lower respiratory or systemic illness, or both), and "febrile" (oral temperature of 37.8 °C or higher). Laboratory confirmation was based on culture and/or 4‐fold or greater rise in antibody titre occurring between postvaccination (pre‐epidemic), acute, convalescent and/or spring (postepidemic) sera. Surveillance was active.

Notes

Influenza‐like illness and influenza cases were detected in 3 groups: first vaccinated, multivaccinated, and placebo. Febrile illnesses were included in the analysis; the first 2 groups' cases were combined. Circulating strains were B/Ann Arbor/1/86, A/Mississippi/1/85.
Efficacy data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No description

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description

Summary assessment

Unclear risk

No description

aa Keitel 1997b

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1986 to 1987 influenza season. Follow‐up lasted the whole epidemic period. Influenza period was defined by viral surveillance. Specimens for culture and acute‐convalescent blood specimens were obtained from ill people. At spring time, volunteers were asked to record any illness that occurred during the epidemic period, and blood specimens were collected.

Participants

940 healthy employees working in the Texas Medical Center in Houston, Texas, or in surrounding industrial companies: 723 treated and 217 placebo. Age of participants was 30 to 60.

Interventions

Trivalent, killed, whole, intramuscularly administered vaccine. Schedule and dose were: 2 doses; 15 µg of haemagglutinin of each influenza strain. Vaccine composition was: A/Mississippi/1/85/H3N2), A/Chile/1/83 (H1N1), and B/Ann Arbor/1/86 plus A/Taiwan/1/86 (H1N1). Placebo was sterile saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

ILI, influenza. Illnesses were classified as "any", "flu‐like" (lower respiratory or systemic illness, or both), and "febrile" (oral temperature of 37.8 °C or higher). Laboratory confirmation was based on culture and/or 4‐fold or greater rise in antibody titre occurring between postvaccination (pre‐epidemic), acute, convalescent and/or spring (postepidemic) sera. Surveillance was passive.

Notes

Influenza‐like illness and influenza cases were detected in 3 groups: first vaccinated, multivaccinated, and placebo. Febrile illnesses were included in the analysis; the first 2 groups' cases were combined. Circulating strain was A/Taiwan/1/86. Effectiveness data only were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers according to prior vaccination experience.

Allocation concealment (selection bias)

Unclear risk

Insufficent information available to judge

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, but no further details available.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description

Summary assessment

Unclear risk

No description

aa Keitel 1997c

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1987 to 1988 influenza season. Follow‐up lasted the whole epidemic period. Influenza period was defined by viral surveillance. Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers according to prior vaccination experience. Specimens for culture and acute‐convalescent blood specimens were obtained from ill people. At spring time, volunteers were asked to record any illness that occurred during the epidemic period, and blood specimens were collected.

Participants

934 healthy employees working in the Texas Medical Center in Houston, Texas, or in surrounding industrial companies: 789 treated and 145 placebo. Age of participants was 30 to 60.

Interventions

Trivalent, killed, whole, intramuscularly administered vaccine. Schedule and dose were: single dose; 15 µg of haemagglutinin of each influenza strain. Vaccine composition was: A/Leningrad/360/86 (H3N2), A/Taiwan/1/86 (H1N1), B/Ann Arbor/1/86. Placebo was sterile saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

ILI, influenza. Illnesses were classified as "any", "flu‐like" (lower respiratory or systemic illness, or both), and "febrile" (oral temperature of 37.8 °C or higher). Laboratory confirmation was based on culture and/or 4‐fold or greater rise in antibody titre occurring between postvaccination (pre‐epidemic), acute, convalescent and/or spring (postepidemic) sera. Surveillance was passive.

Notes

Influenza‐like illness and influenza cases were detected in 3 groups: first vaccinated, multivaccinated, and placebo. Febrile illnesses were included in the analysis; the first 2 groups' cases were combined. Circulating strains were A/Sichuan/1/87, B/Victoria/2/87. Effectiveness data only were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No description

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description

Summary assessment

Unclear risk

No description

aa Langley 2011

Methods

Randomised, placebo‐controlled trial assessing the protective efficacy of a nasally administered meningococcal outer membrane protein adjuvanted trivalent influenza vaccine (OMP‐TIV) against laboratory‐confirmed influenza infection during the 2003 to 2004 influenza season in Canada in healthy adults.

Participants

Healthy adults aged 18 to 64 years who gave informed consent were eligible to participate (1349 were enrolled at 28 sites in Canada). Exclusion criteria: belonging to a group for which annual influenza vaccination is recommended; presence of significant acute or chronic, uncontrolled medical or psychiatric illness; pregnancy; infection with HIV, hepatitis B, or hepatitis C virus; chronic use of any medication or product for symptoms of rhinitis or nasal congestion or any chronic nasopharyngeal complaint or use of such product within 7 days prior to immunisation; asthma; symptoms or diagnosis suggesting gag reflex impairment or predisposition to aspiration; use of systemic glucocorticosteroids or immunosuppressive medications; receipt of investigational drugs in the prior month; presence of febrile or upper respiratory tract illness on the day of immunisation; and known hypersensitivity to mercurials or chicken eggs.

Interventions

The vaccine contains equal parts of 3 monovalent egg‐grown, formalin‐inactivated influenza antigens formulated with OMPs of Neisseria meningitidis serogroup B strain 8047.

The vaccine tested in this study contained HA from each:

  • A/New Caledonia/20/99 (H1N1)

  • A/Panama/2007/99 (H3N2)

  • B/Shangdong/7/97 (H1N1) (recommended for the 2003 to 2004 season)

Vaccine was tested in 2 formulations: 1 containing 75 ± 15 μg/mL of HA from each of the 3 influenza strains and 1 with 150 ± 30 μg HA/mL. Both formulations are sterile, colourless to yellowish opalescent, and preserved with 0.01% thimerosal.

The placebo control was sterile phosphate‐buffered isotonic saline with 0.01% thimerosal and was colourless.

Participants (n = 1348) were randomised to 1 of the following 3 regimens:

  • Arm 1: meningococcal OMP‐adjuvanted TIV with 15 μg of each HA antigen on days 0 and 14 (n = 455)

  • Arm 2: meningococcal OMP‐adjuvanted TIV with 30 μg of each HA antigen on day 0 and saline placebo on day 14 (n = 450)

  • Control: saline placebo on days 0 and 14 (n = 443)

Vaccine and placebo were administered by means of a VP3/100 nasal spray pump (Valois of America, Greenwich, CT, USA) with the participant in a sitting position, administering 0.10 mL of preparation in each nostril (0.20 mL in all).

Outcomes

Safety

Participants were monitored for 30 minutes after the immunisation on days 0 and 14 for any immediate adverse events and then completed a questionnaire that graded selected complaints as 0 (none), grade 1 (mild), grade 2 (moderate), or grade 3 (severe). From days 0 to 7, participants self monitored evening oral temperature and completed a written memory aid of reactogenicity. On days 3, 7, 17, and 21 participants reported the maximum oral temperature and severity score in the previous days via an interactive voice response system. A clinic visit for participant assessment was initiated if symptom complaints exceeded grade 2. Prior to the day 14 dose participants were questioned about interim adverse events, and a physical exam was performed. Coding for adverse events was according to Medical Dictionary for Regulatory Activities (MeDRA, Chantilly, VA) version 6.1. The following outcomes were reported:

  • Burning or stinging in the nose

  • Burning or stinging in the throat

  • Itching in the nose, throat, or eyes

  • Shortness of breath

  • Lightheadedness or dizziness

  • New rash or a rash becoming itchy

  • Feverishness: temperature (°C) < 37.8; 37.8 to 38.2; 38.3 to 38.9; ≥ 39.0

Immunogenicity
Blood and nasal mucus samples were collected on days 0 and 28 for haemagglutinin inhibition reciprocal titres and salivary secretory IgA (sIgA) measurement, respectively.

Effectiveness
Telephone contacts with participants were made every 2 weeks to solicit adverse events and identify ILI. Spontaneous illness reports were received via toll‐free telephone call centre and reported to investigators. If the participant illness included at least 2 of the illness criteria and was severe enough to impede normal daily activities, then a nurse visit was initiated. The nurse verified symptoms, collected nose and throat swabs, and recorded the participant’s temperature. Samples were cultured on MDCK cells, and a multiplex RT‐PCR test was used to detect influenza A and B viruses (viruses A were subsequently subtyped by another RT‐PCR assay). The primary outcome measure for efficacy was CCI defined as fever (oral temperature > 37.8 °C) and cough and at least 1 of the following: sore throat, runny nose or nasal congestion, muscle or joint ache, headache, fatigue or chills (with symptoms sufficient to impede normal daily activities), and a positive nose and throat swab culture for influenza A or B virus.

A co‐primary endpoint measure was a positive culture, defined as positive nose and throat swab culture for influenza A or B virus and at least 2 of the following 8 symptoms: fever, cough, sore throat, runny nose or nasal congestion, muscle or joint ache, headache, fatigue, or chills.

The secondary outcome measure, ILI with evidence of influenza infection, required laboratory confirmation of influenza by either a positive culture for influenza A or B virus, or positive RT‐PCR for influenza A or B virus, or a 4‐fold rise in reciprocal titre for a circulating influenza strain between days 28 and 180 and fever and cough and at least 1 of sore throat, runny nose or nasal congestion, muscle or joint ache, headache, fatigue, or chills.

Notes

Safety and primary endpoint estimates (CCI) were calculated on the ITI population, which included any participant who received at least 1 dose of test article (n = 1348, 455 in arm 1, 450 in arm 2, 443 in control arm).
For effectiveness estimates of culture positive and ILI, evaluable participants were used, i.e. those who had a complete regimen (i.e. 1 dose of placebo in the placebo group, at least 1 dose of 30 µg, 2 doses of 15µg, n = 1347).
A total of 1326 participants completed the study (452 in arm 1, 442 in arm 2, 432 in control arm).

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

“The study was double‐blind, randomised and placebo controlled.”

Allocation concealment (selection bias)

Low risk

“Subjects were assigned centrally within blocks and stratified within each site by age ≤49 and >49 years, and history of prior influenza immunization within 2 years.”

Blinding (performance bias and detection bias)
All outcomes

Low risk

“Neither the subject nor the site study team (staff performing clinical safety or efficacy evaluations and investigators) were aware of patient assignment. One research nurse at each site was responsible for randomization, maintenance of the treatment log, test article preparation and administration.”
“This staff member did not perform any safety or efficacy observations and could not reveal treatment assignment to participants or other study staff.”
“Both lots are sterile, colorless to yellowish opalescent and preserved with 0.01% thimerosal. The placebo control was sterile phosphate‐buffered isotonic saline with 0.01% thimerosal, and was colorless.”

Incomplete outcome data (attrition bias)
All outcomes

Low risk

About 98% of the initially enrolled participants completed the study.

Summary assessment

Low risk

Low risk of bias

aa Leibovitz 1971

Methods

Controlled clinical trial conducted in the USA during the 1969 to 1970 influenza season. The study period was 30 January to 18 May. Follow‐up lasted first 7 weeks of training. Influenza was detected from 11 February to 13 May and lasted 6 weeks. Participants were allocated to vaccine or control group according to the last non‐zero digit of the Social Security number. Blinding was not mentioned. Specimens for culture and acute‐convalescent blood specimens were obtained from people hospitalised with acute respiratory disease.

Participants

9616 military trainees: 1682 treated and 7934 placebo. Age of participants was 18 to 20.

Interventions

Monovalent inactivated, experimental, intramuscularly administered vaccine. Schedule and dose were: single dose, 556 CCA. Recombinant virus derived from HK/Aichi/68 and A0/PR8/34 was compared against no vaccination. Vaccine was not recommended but matched circulating strain.

Outcomes

Outcomes were: hospitalisation for upper respiratory infection (without definition), hospitalisation for influenza. Laboratory confirmation was based on culture and/or 4‐fold or greater rise in antibody titre occurring between acute and convalescent sera. Surveillance was passive.

Notes

Recruitment and immunisation period overlapped outbreak period. Most of the illnesses were due to adenovirus. Illnesses during the first 1 or 2 weeks after vaccination were not excluded, but the authors stated that this fact did not affect the results. Efficacy data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

High risk

Inadequate

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

High risk

Unclear

aa Mcbride 2016a

Methods

Randomised placebo controlled trial

Participants

Healthy adults aged 18 to 64

Interventions

Trivalent influenza inactivated vaccines containing antigens the 2 A strains and 1 B strain recommended by WHO in 2008 and 2009 for the Southern Hemisphere as follows:

Arm 1: 15 mg of haemagglutinin antigens Solomon Islands/3/2006 (H1N1), A/Brisbane/10/2007 (H3N2), B/Brisbane/3/2007. Fluvax; CSL Limited. 0.5 mL single doses administered intramuscularly into deltoid muscle.

Placebo: 0.5 mL saline, dibasic sodium phosphate and monobasic sodium phosphate.

Outcomes

Influenza cases laboratory confirmed by viral culture and/or real time RT‐PCR were followed up until 30 November each year. Solicited adverse events for 4 days, unsolicited adverse events for 20 days, serious adverse events for 180 days. Harms data were not extractable due to different definitions.

Notes

This record is for the 2008 season. Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Participants were randomized in a 2:1 ratio to receive a single injection of 0.5 mL IIV3 or placebo, administered intramuscularly into the deltoid muscle.”

“The randomization code was prepared by a statistician, employed by CSL Limited, with the use of SAS software (version 9.1.3; SAS Institute, Cary, NC, USA ...”

Allocation concealment (selection bias)

Low risk

“The randomization code was prepared by a statistician, employed by CSL Limited, with the use of SAS software (version 9.1.3; SAS Institute, Cary, NC, USA), using simple block randomization to maintain approximate allocation balance."

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

"As there was a visual difference between IIV3 and placebo, study personnel who were involved in the preparation and administration of the study vaccine had no further involvement in the study conduct. Participants and investigational site staff involved in performing study assessments remained blinded to treatment allocation."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Flow of participants during the study is reported and described. Loss to follow‐up amounts to 0.36% and 0.44% study end and among vaccine and placebo recipients, respectively, and is balanced through the 2 arms.

Summary assessment

Low risk

aa Mcbride 2016b

Methods

Randomised placebo‐controllled trial

Participants

Healthy adults aged 18 to 64

Interventions

Trivalent influenza inactivated vaccines containing antigens the 2 A strains and 1 B strain recommended by WHO in 2008 and 2009 for the Southern Hemisphere as follows:

Arm 1: 15 mg of haemagglutinin antigens A/Brisbane/59/2007 (H1N1), A/Brisbane/10/2007 (H3N2), B/Florida/4/2006. Fluvax; CSL Limited. 0.5 mL single doses administered intramuscularly into deltoid muscle.

Placebo: 0.5 mL saline, dibasic sodium phosphate and monobasic sodium phosphate.

Outcomes

Influenza cases laboratory‐confirmed by viral culture and/or real time RT‐PCR were followed up until 30 November each year. Solicited adverse events for 4 days, unsolicited adverse events for 20 days, serious adverse events for 180 days. Harms data were not extractable due to different definitions.

Notes

This record is for the 2009 season. Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

“Participants were randomized in a 2:1 ratio to receive a single injection of 0.5 mL IIV3 or placebo, administered intramuscularly into the deltoid muscle.”

“The randomization code was prepared by a statistician, employed by CSL Limited, with the use of SAS software (version 9.1.3; SAS Institute, Cary, NC, USA ...”

Allocation concealment (selection bias)

Low risk

“The randomization code was prepared by a statistician, employed by CSL Limited, with the use of SAS software (version 9.1.3; SAS Institute, Cary, NC, USA), using simple block randomization to maintain approximate allocation balance.”

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

“As there was a visual difference between IIV3 and placebo, study personnel who were involved in the preparation and administration of the study vaccine had no further involvement in the study conduct. Participants and investigational site staff involved in performing study assessments remained blinded to treatment allocation.”

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Flow of participants is reported and described. Attrition was 2.52% and 1.6% for the intervention and placebo arms, respectively. In this season (2009) 104/5001 and 40/2499 received H1N1 pandemic vaccine, respectively in the 2 arms and were excluded from efficacy assessment.

Summary assessment

Low risk

aa Mesa Duque 2001

Methods

Randomised controlled trial, double‐blind, conducted in Colombia during the 1997 influenza season. Follow‐up lasted from 15 March to 31 August. Influenza period was not defined. Virological surveillance was not performed.

Participants

493 bank employees: 247 treated and 246 placebo. Age of participants was 18 to 60.

Interventions

Subunit inactivated, intramuscularly administered vaccine. Schedule and dose were: single dose. Vaccine composition was: A/Wahan/359/95, A/Texas/36/91, and B/Beijing/184/93. Placebo was vitamin C. Vaccine was recommended and matched circulating strain.

Outcomes

Episodes of clinical illness, WDL, and adverse effects. Clinical disease was defined as upper respiratory illness (fever, sore throat, and cough lasting more than 24 hours) according to ICD‐9 codes 381, 382, 460, 466, 480 and from 487 to 490. Local adverse effects were oedema, erythema, pain, and swelling. Systemic adverse effects were fever, headache, and indisposition within 5 days of vaccination. Surveillance was passive.

Notes

Circulating strains were not isolated from local cases but by WHO and Colombia surveillance system and matched vaccine components. Working days lost were detected all year round, so they were not included in the analysis. Efficacy and safety data were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Volunteers were randomly allocated to receive vaccine or placebo using a table of random numbers.

Allocation concealment (selection bias)

Low risk

Given details provided regarding randomisation process and other aspects of the study design, we believe the allocation concealment was probably adequate.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinding was ensured by pre‐labelled, coded, identical‐looking vials.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Adequate

Summary assessment

Low risk

Low risk

aa Mixéu 2002

Methods

Randomised controlled trial, double‐blind, conducted in Brazil during the 1997 influenza season. Follow‐up lasted 6 to 7 months. Influenza period was not defined. Virologic surveillance was not performed.

Participants

813 flight crews of an airline company: 405 vaccinated and 408 given placebo. Age of participants was 18 to 64.

Interventions

Split trivalent, intramuscularly administered vaccine. Schedule and dose were: single dose. Vaccine composition was: A/Nanchang/933/95, A/Texas/36/91, and B/Harbin/7/94. Placebo was vaccine diluent. Vaccine was recommended and matched circulating strain.

Outcomes

ILI, WDL. Clinical illness was defined as follows: fever > 37.6 °C and cough, headache, myalgia, rhinorrhoea, sore throat lasting at least 24 hours. Surveillance was passive.

Notes

Local and systemic effects were reported together and therefore not included in the review. Only 294 treated participants and 299 controls completed follow‐up. Efficacy data were extracted.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Authors did not describe the methods used to ensure randomisation.

Allocation concealment (selection bias)

Unclear risk

Authors did not describe the methods used to ensure randomisation.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Authors did not describe the methods used to ensure blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Low rates of attrition unlikely to affect study results.

Summary assessment

Unclear risk

Unclear

aa Mogabgab 1970a

Methods

Randomised study conducted in the USA during the 1968 to 1969 influenza season. Influenza outbreak lasted 9 weeks, from 9 December to 3 February. Randomisation methods were not described. Laboratory confirmation was obtained (by culture or 4‐fold antibody titre increase in acute convalescent sera) for 20 men randomly selected each week from among the ill.

Participants

1402 airmen previously unvaccinated: 881 vaccinated and 521 given placebo. Age of participants was 18 to 21.

Interventions

Monovalent inactivated parenteral influenza A vaccine. Schedule and dose were: single dose. Vaccine composition was: A2/Aichi 2/68 300 CCA units. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

ILI and influenza, complications and admissions. All respiratory illnesses were classified as febrile (38.3 °C or greater), afebrile, pharyngitis, bronchitis, or pneumonia (complications). Surveillance was passive.

Notes

Cases occurring during the first 15 days after vaccination were not included in the analysis. Circulating strain was A2/Hong Kong. Efficacy data were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Mogabgab 1970b

Methods

Randomised study conducted in the USA during the 1968 to 1969 influenza season. Influenza outbreak lasted 9 weeks, from 9 December to 3 February. Randomisation methods were not described. Laboratory confirmation was obtained (by culture or 4‐fold antibody titre increase in acute convalescent sera) for 20 men randomly selected each week from among the ill.

Participants

1551 airmen previously unvaccinated: 1030 vaccinated and 521 given placebo. Age of participants was 18 to 21.

Interventions

Polyvalent inactivated influenza A and B vaccine (the 1967 military formula). Schedule and dose were: single dose. Vaccine composition was: A/Swine/33 100 CCA units, A/PR8/34 100 CCA units, A1/AA/1/57 100 CCA units, A2/Taiwan 1/64 400 CCA units, B/Lee/40 100 CCA units, B/Mass 3/66 200 CCA units. Placebo was saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

ILI and influenza cases, complications and admissions. All respiratory illnesses were classified as febrile (38.3 °C or greater), afebrile, pharyngitis, bronchitis, or pneumonia (complications). Surveillance was passive.

Notes

Cases occurring during the first 15 days after vaccination were not included in the analysis. Circulating strain was A2/Hong Kong. Efficacy data were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Monto 1982

Methods

Randomised, single‐blind study conducted in the USA during the 1979 to 1980 influenza season. Follow‐up lasted for the whole epidemic period. The epidemic period was defined by first and last isolation (11 February to 18 March) and lasted 5 weeks. Each participant was given a serial number that had previously been assigned randomly by a code to either the vaccine or the placebo group. Specimens for culture were obtained from ill people. At spring time blood specimens were collected.

Participants

306 students: 154 vaccinated and 152 given placebo. Age of participants was not reported.

Interventions

Monovalent, live attenuated, intranasal influenza B. Schedule and dose were: single dose. Vaccine composition was: the vaccine virus, cold recombinant, was produced by recombining the attenuated B/Ann Arbor/1/66 with a wild strain B/Hong Kong/8/73. Placebo was vaccine diluent. Vaccine was not recommended and did not match the circulating strain.

Outcomes

Clinical and laboratory confirmed cases and adverse effects. Participants suffered a respiratory illness if they had at least 2 respiratory symptoms. Cases were laboratory confirmed if they had an increase in antibody titre against 3 influenza B virus antigens, i.e. if there was a 4‐fold increase from an initial sample. Side effects were sore throat, coryza, hoarseness, cough, muscle aches, temperature > 100 °F occurring during the first 3 days after vaccination. Surveillance was active.

Notes

Vaccine content was not recommended or matched. Circulating strain was B/Singapore/79‐like and B/Buenos Aires/79‐like.
Efficacy and safety data were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Adequate

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Adequate

Summary assessment

Low risk

Adequate

aa Monto 2009

Methods

Third epidemic season (2007 to 2008) of aa Ohmit 2006 and aa Ohmit 2008

Participants

A total of 1952 healthy adults between the ages of 18 and 49 years were enrolled. Some had been also enrolled in the 2 previous seasons.

Interventions

Newly enrolled participants were recruited from the community around 4 university campuses in Michigan. Allocation methods are the same as for aa Ohmit 2006 and aa Ohmit 2008.

For the 2007 to 2008 season vaccine composition was the following:

  • Fluzone (Sanofi Pasteur, inactivated trivalent vaccine intramuscular): 15 μg of haemagglutinin from each of the following strains in a 0.5 mL dose: A/Solomon Islands/3/2006 (H1N1), A/Wisconsin/67/2005 (H3N2), and B/Malaysia/2506/2004 (B/Victoria lineage).

  • FluMist (MedImmune, live attenuated vaccine, intranasal): 106.5‐7.5 fluorescent focus units of live attenuated influenza virus reassortants of the same strains as used for the inactivated formulation in a 0.2 mL dose.

Outcomes

Same outcomes as aa Ohmit 2008

Notes

Funding source ‐ mixed

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Nichol 1995

Methods

Randomised controlled trial conducted in the USA during the 1994 to 1995 influenza season. Follow‐up lasted from 1 December 1994 through to 31 March 1995. Influenza period was not defined. Virological surveillance was not performed.

Participants

841 full‐time employed: 419 treated and 422 placebo. Age of participants was 18 to 64.

Interventions

Subvirion, trivalent, parenteral influenza A and B vaccine. Schedule and dose were: single dose; 15 µg each strain. Vaccine composition was: A/Texas/36/91, A/Shangdong/9/93, B/Panama/45/90. Placebo was vaccine diluent. Vaccine was recommended and matched circulating strain.

Outcomes

Cases (symptom‐defined), working days lost due to respiratory illness, side effects. Participants were defined as cases if they had at least 1 upper respiratory illness (a sore throat associated with either fever or cough that lasted at least 24 hours). Local adverse effects were defined as arm soreness. Systemic adverse effects were defined as fever, tiredness, "feeling under the weather", muscle ache, headache (within a week after vaccination). Surveillance was active.

Notes

Circulating strain was not indicated. Efficacy and safety data were extracted.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed according to a computer‐generated randomisation schedule.

Allocation concealment (selection bias)

Low risk

Probably adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinding was ensured by preloaded, coded, identical‐looking syringes.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Adequate

Summary assessment

Low risk

Adequate

aa Nichol 1999a

Methods

Randomised controlled trial conducted in the USA during the 1997 to 1998 influenza season. Follow‐up lasted from November to March. Site‐specific peak outbreak period was defined as weeks including 80% of the isolates of a specific area. Total outbreak period lasted from 14 December 1997 through to 21 March 1998. Total outbreak period was included in the analysis and lasted 14 weeks. Participants were recruited from 7 organisations and assigned to 1 of the study groups using a permuted block randomisation scheme that was stratified by treatment centre and age group. Sealed randomisation envelopes contained vaccine codes. Influenza virus surveillance was carried out in the area.

Participants

4561 healthy working adults: 3041 treated and 1520 placebo. Age of participants was 18 to 64.

Interventions

Trivalent, live attenuated influenza A and B vaccine in a single dose. Vaccine composition was: A/Shenzhen/227/95, A/Wuhan/395/95, B/Harbin/7/94‐like. Placebo was egg allantoic fluid. Vaccine was recommended but did not match the circulating strain.

Outcomes

Clinical cases (symptom‐defined), working days lost, and adverse effects. Case definition had 3 specifications: febrile illness (fever for at least 1 day and 2 or more symptoms for at least 2 days: fever, chills, headache, cough, runny nose, sore throat, muscle aches, tiredness); severe febrile illness (3 days of symptoms and 1 day of fever); febrile upper respiratory tract illness (3 days of upper respiratory tract symptoms and 1 day of fever). We chose the febrile illness outcome for analysis. Systemic adverse effects were defined as headache, muscle aches, chills, tiredness, and fever. Surveillance was passive.

Notes

Complete follow‐up data were obtained for 2874 participants in the treatment arm and 1433 participants in the placebo arm. The outcome working days lost is presented as a rate ratio; the data are presented in a way that allows us to compute the difference in mean days lost but not to compute the standard error. Circulating strain was A/Sydney/5/97‐like. Efficacy and safety data were extracted.

Government and industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Adequate

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Adequate

Summary assessment

Low risk

Adequate

aa Ohmit 2006

Methods

Multicentre, randomised, placebo‐controlled trial assessing effectiveness of both inactivated and live attenuated vaccines in preventing laboratory‐confirmed influenza in healthy adults aged below 50.

Participants

For enrolment in the first study year (2004 to 2005), participants were recruited at 4 centres (2 university and 2 community sites) in Michigan. Participants were healthy adults between the ages of 18 and 46 years; those for whom influenza vaccination was recommended or contraindicated were excluded. In all 1247 were enrolled.

Interventions

After informed consent was obtained and a first serum sample drawn, enrolled participants were randomly allocated to receive 1 dose of the following:

  • Inactivated trivalent vaccine (Fluzone, Sanofi Pasteur) containing 15 μg of haemagglutinin from each of the following strains: A/New Caledonia/20/99 (H1N1), A/Wyoming/3/2003 (H3N2, A/Fujian/411/2002‐like strain), and B/Jiangsu/10/2003 (B/Shanghai/361/2002‐like strain (Yamagata lineage)) in each 0.5 mL dose, as intramuscular injection.

  • Placebo saline administered intramuscularly.

  • Live attenuated trivalent vaccine (FluMist, MedImmune) containing a 106.5‐7.5 median tissue‐culture infective dose of live attenuated influenza virus reassortants of the following strains: A/New Caledonia/20/99 (H1N1), A/Wyoming/ 3/2003 (H3N2 A/Fujian/411/2002‐like strain), and B/Jilin/20/2003 (B/Shanghai/361/2002‐like strain (Yamagata lineage)) in each 0.5 mL dose.

  • Placebo saline administered intranasally.

Identical syringes were filled on site with the inactivated vaccine or matching placebo (physiologic saline) by study nurses who were aware of the intervention assignments. The live attenuated influenza vaccine and matching placebo (physiologic saline) were preloaded in identical nasal spray devices by the manufacturer. Both vaccines were licenced for use in the 2004 to 2005 influenza season.

Participants were randomised to vaccine or placebo in ratio of 5:1 using 4 site‐specific randomisation schedules, generated with the use of a random permuted block design with a block size of 12, in order to assign participants sequentially to receive a vaccine or a placebo as they enrolled.

Since the trial was double‐blind, the participants and nurses who administered the study vaccine or placebo were unaware of whether the participant was receiving vaccine or placebo but were aware of the route of administration.

Further serum samples were drawn 3 to 5 weeks after vaccine administration (as participants returned diary cards for local and systemic reactions, preseason sample) and during April to May 2005 (postseason sample).

Outcomes

Local and systemic reactions within 7 days from immunisation (self filled questionnaires): fever, chills, runny nose or congestion, cough, sore throat, headache, muscle aches, weakness, abdominal pain, trouble breathing, red eyes, arm soreness, arm redness.

Laboratory‐confirmed influenza. Active surveillance was maintained between November 2004 and April 2005. Participants were contacted by phone or email twice monthly. Symptomatic influenza was described as the presence of at least 1 respiratory symptom (cough or nasal congestion) and at least 1 systemic symptom (fever, feverishness, chills, body aches) occurring during influenza activity and at least 2 weeks after administration. Participants were instructed to contact study staff when at least 2 respiratory and systemic symptoms were observed. Throat swab specimens were collected from all participants with symptomatic influenza.

Swabs were cultured for identification, and all isolates were typed according to strain using the fluorescence antibody assay and evaluated for antigenic relatedness to vaccine strains by the Influenza Branch at the CDC. In addition, all throat‐swab specimens obtained from participants with symptomatic influenza were tested at the University of Michigan by means of real‐time PCR assays using the TaqMan system (Applied Biosystems).

All collected serum samples were tested with the haemagglutination‐inhibition assay, with the virus strains present in the vaccines used as antigens and against the circulating type A (H3N2) (A/California/07/2004‐like) virus and the circulating type B (B/Hawaii/33/ 2004‐like) virus (i.e. Victoria lineage not included in the vaccine).

For effectiveness the following endpoints were used:

On ITT population: laboratory‐confirmed influenza: culture‐positive or real‐time PCR‐positive, or both.

On per‐protocol population: laboratory‐confirmed influenza: serologically positive; serologically or culture‐positive.

Notes

Intention‐to‐treat analysis: includes all enrolled participants who were randomly assigned to a vaccine or placebo group and who received a vaccine or a placebo (TIV = 513; placebo IM = 103; LAIV = 519; placebo IN = 103).

Per‐protocol analyses: limited to participants having the postintervention (preseason) blood specimen collected at least 3 weeks after receipt of a vaccine or a placebo and at least 2 weeks before the beginning of local influenza activity (TIV = 367; placebo IM = 73; LAIV = 363; placebo IN = 73).

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centralised automated sequence generation

Allocation concealment (selection bias)

Unclear risk

Allocation procedure not described

Blinding (performance bias and detection bias)
All outcomes

Low risk

Blinding apparently successful

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Active surveillance carried out Participants contacted bi monthly

Summary assessment

Unclear risk

Unclear

aa Ohmit 2008

Methods

Multicentre, randomised, placebo‐controlled trial assessing the effectiveness of both inactivated and live attenuated vaccines in preventing laboratory‐confirmed influenza in healthy adults aged below 50 years. Same methods as aa Ohmit 2006.

Participants

For study year 2005 to 2006, healthy men and women aged 18 to 48 years were recruited at 6 study sites (4 university sites and 2 community sites) in Michigan. In all 2058 participants were enrolled. Of these, 972 were already enrolled in the 2004 to 2005 season (see aa Ohmit 2006).

Interventions

Participants who were enrolled in the 2005 to 2006 season were randomised (see aa Ohmit 2006) to receive inactivated vaccine (Fluzone; Sanofi Pasteur), live attenuated vaccine (FluMist; MedImmune), or placebo. Participants already enrolled in the 2004 to 2005 season received the same intervention type (i.e. Fluzone, FluMist, or placebo) as before.

  • Fluzone (intramuscularly administered) contained 15 g haemagglutinin from each of the following strains: A/New Caledonia/20/99 (H1N1), A/New York/55/2004 (H3N2) (A/California/7/2004‐like), and B/Jiangsu/10/2003 (B/Shanghai/361/2002‐like).

  • FluMist (intranasally administered) was formulated to contain a median tissue‐culture infective dose of 106.5 to 107.5 live attenuated influenza virus reassortants of the same strains.

  • Intramuscular or intranasal saline placebo.

Outcomes

  • Local and systemic reactions within 7 days from immunisation (see Ohmit 2006).

  • Symptomatic laboratory‐confirmed influenza A or B illness (primary efficacy outcome). Symptoms were defined as at least 1 respiratory symptom (cough or nasal congestion) plus at least 1 systemic symptom (fever or feverishness, chills, or body aches). Laboratory confirmation was assessed by isolation of the influenza virus in cell culture or by comparison of paired postvaccination (preseason) and postseason serum with at least a 4‐fold increase in haemagglutination‐inhibition antibody titre to 1 circulating influenza strain.

  • Illnesses confirmed by identification of the virus in real‐time PCR assays was considered as a secondary efficacy outcome.

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Powers 1995a

Methods

Randomised controlled trial conducted in the USA during the 1993 to 1994 influenza season. Follow‐up was not indicated. Influenza period was not defined. Participants were randomly assigned to receive 1 of the following 5 vaccine preparations in a double‐blinded manner: 15 mg of rHA0, 15 mg of rHA0 plus alum, 90 mg of rHA0, licensed, and placebo. Spring sera were collected.

Participants

34 healthy university students: 26 treated and 8 placebo. Age of participants was 18 to 45.

Interventions

Subvirion licensed trivalent parenteral AB vaccine. Schedule and dose were: single dose; 15 µg each strain. Vaccine composition was: A/Texas/36/91 (H1N1), A/Beijing/32/92 (H3N2), and B/Panama/45/90. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Clinical and laboratory‐confirmed cases and adverse effects. An "influenza‐like illness" was defined as the presence of any respiratory symptom(s) for >= 2 days, accompanied by fever or systemic symptoms of myalgia or chills. Laboratory evidence of influenza A (H3N2) virus infection was defined as either or both of the isolation of virus from nasopharyngeal secretion and a >= 4‐fold increase in serum HI antibody titre between the 3‐week postvaccination (preseason) specimen and the corresponding postseason specimen collected in the following spring. Local adverse effects were erythema, pain, tenderness, induration, arm stiffness; systemic adverse effects: were headache, generalised myalgia, diarrhoea, nausea, feverishness, temperature > 37.8 °C.

Notes

Efficacy and safety data were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information to judge

Allocation concealment (selection bias)

Unclear risk

Insufficient information to judge

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Insufficient information to judge

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to judge

Summary assessment

Unclear risk

Unclear

aa Powers 1995b

Methods

Single‐blind randomised controlled trial conducted in the USA during the 1974 to 1975 influenza season. Follow‐up lasted from winter to spring. A "two‐month" epidemic period was described by the authors with no reference to a definition and lasted 6 weeks. Study participants were randomly assigned into 3 subgroups to receive either 2 doses of the vaccine (n = 47), 1 dose of vaccine and 1 dose of placebo (n = 48), or 2 doses of placebo (n = 48) at 14 days apart. 6‐month sera were collected on all study participants.

Participants

34 healthy university students: 26 treated and 8 placebo. Age of participants was 18 to 45.

Interventions

Subvirion monovalent parenteral vaccine. Schedule and dose were: single dose; 90 µg rHA0. Vaccine composition was: the recombinant HA vaccine contained HA0 glycoprotein from the influenza A/Beijing/32/92 (H3N2) virus. Placebo was saline for injection. Vaccine was not recommended but matched circulating strain.

Outcomes

Clinical and laboratory‐confirmed cases. An "influenza‐like illness" was defined as the presence of any respiratory symptom(s) for >= 2 days, accompanied by fever or systemic symptoms of myalgia or chills. Laboratory evidence of influenza A (H3N2) virus infection was defined as either or both of the isolation of virus from nasopharyngeal secretion and a >= 4‐fold increase in serum HI antibody titre between the 3‐week postvaccination (preseason) specimen and the corresponding postseason specimen collected in the following spring.

Notes

Safety data were not included; effectiveness data were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Powers 1995c

Methods

Randomised controlled trial conducted in the USA during the 1993 to 1994 influenza season. Follow‐up was not indicated. Influenza period was not defined. Participants were randomly assigned to receive 1 of the following 5 vaccine preparations in a double‐blinded manner: 15 mg of rHA0, 15 mg of rHA0 plus alum, 90 mg of rHA0, licensed, and placebo. Spring sera were collected.

Participants

59 healthy university students: 51 treated and 8 placebo. Age of participants was 18 to 45.

Interventions

Subvirion monovalent parenteral vaccine. Schedule and dose were: single dose; 15 µg rHA0. Vaccine composition was: the recombinant HA vaccine contained HA0 glycoprotein from the influenza A/Beijing/32/92 (H3N2) virus. Placebo was saline for injection. Vaccine was not recommended but matched circulating strain.

Outcomes

Clinical and laboratory‐confirmed cases. An "influenza‐like illness" was defined as the presence of any respiratory symptom(s) for >= 2 days, accompanied by fever or systemic symptoms of myalgia or chills. Laboratory evidence of influenza A (H3N2) virus infection was defined as either or both of the isolation of virus from nasopharyngeal secretion and a >= 4‐fold increase in serum HI antibody titre between the 3‐week postvaccination (preseason) specimen and the corresponding postseason specimen collected in the following spring.

Notes

Efficacy data only were extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Rytel 1977

Methods

Single‐blind randomised controlled trial conducted in the USA during the 1974 to 1975 influenza season. Follow‐up lasted from winter to spring. A "two month" epidemic period was described by the authors with no reference to a definition and lasted 6 weeks. Study participants were randomly assigned into 3 subgroups to receive either 2 doses of the vaccine (n = 47), 1 dose of vaccine and 1 dose of placebo (n = 48), or 2 doses of placebo (n = 48) at 14 days apart. 6‐month sera were collected on all study participants.

Participants

143 young adult female student nurse volunteers: 95 treated and 48 placebo. Age of participants was 18 to 35.

Interventions

Live attenuated, bivalent, intranasal influenza A (containing 107,2 EID50) and B (containing 107,8 EID50) vaccines. Schedule and dose were single or double doses. Vaccine composition was: A/England/42/72 (H3N2) and B/Hong Kong/5/72. Placebo was 5% sucrose. Vaccine was not recommended and did not match the circulating strain.

Outcomes

Influenza and adverse effects. An influenza case was defined as the presence of an influenza‐like illness (3 or more symptoms of acute respiratory disease and temperature greater then 37.2 °C) and virus isolation and/or 4‐fold rise in antibody titre in sera obtained at 30 days and 6 months following immunisation. Local adverse effects were upper respiratory symptoms and cough. These were subdivided into moderate and severe. A definition of general adverse effects (again distinguished between moderate and severe) was not given.

Notes

1 dose and 2 doses were analysed together. Circulating strain was A/PortChalmers/1/73 (H3N2). Efficacy and safety data extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Sumarokow 1971

Methods

Field trial conducted in Russia during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic period was defined as the period of highest influenza morbidity and lasted 11 weeks, from the last 10 days of January to the first 10 days of April. Vaccinations were carried out using coded preparation. Sampling virological and serological survey of ill people was performed.

Participants

19,887 population: 9945 treated and 9942 placebo. Age of participants was 13 to 25.

Interventions

Live allantoic intranasal vaccine. Schedule and dose were: 3 doses. Vaccine composition was not indicated. Placebo was not described. Vaccine was not recommended and did not match the circulating strain.

Outcomes

Clinical cases, deaths, severity of illness. Clinical outcomes were all acute respiratory infections. Laboratory confirmation was obtained on a sample of ill participants by virus isolation or demonstration of seroconversion. Bronchitis, otitis, and pneumonia were considered as complications. Passive surveillance was carried out.

Notes

A first study group with children 3 to 12 years old was excluded. A second study group with participants aged 13 to 25 was included in the analysis. The trial compared 2 live vaccines (allantoic intranasal vaccine and tissue vaccine for oral administration) against placebo. Only intranasal vaccine was included in the analysis. Deaths from flu were not recorded. Circulating strain was A2/Hong Kong/68.
Effectiveness data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient description

Allocation concealment (selection bias)

Unclear risk

Insufficient description

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Insufficient description

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient description

Summary assessment

Unclear risk

Insufficient description

aa Tannock 1984

Methods

Controlled clinical trial, double‐blind, conducted in Australia during the 1981 influenza season. Follow‐up lasted from winter to spring. Influenza period was not defined. Volunteers were alternatively allocated to groups in a double‐blind manner. 6‐month sera were collected.

Participants

88 volunteer staff from Newcastle Hospital and the Commonwealth Steel Corporation: 56 treated and 32 placebo. Age of participants was 16 to 64.

Interventions

Trivalent subunit parenteral vaccine. Schedule and dose were: 7 µg each, 1 or 2 doses. Vaccine composition was: A/Brazil/11/78, A/Bangkok/1/79, B/Singapore/222/79. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Influenza and adverse effects. A case of influenza was defined as a respiratory illness, retrospectively reported, associated with a 4‐fold antibody titre increase between postvaccination and postepidemic sera. Local side effects were redness, swelling, warmth or irritation, pain on contact, pain with pressure, continuous pain, or restriction of arm movement; systemic reactions were fever, chills, sweating, drowsiness, or insomnia.

Notes

1 dose and 2 doses were analysed together; very high dropout. Circulating strain was A/Bangkok/1/79. Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Inadequate

Allocation concealment (selection bias)

High risk

Inadequate

Blinding (performance bias and detection bias)
All outcomes

High risk

Inadequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

Inadequate

Summary assessment

High risk

Inadequate

aa Treanor 2011

Methods

Randomised, placebo‐controlled trial

Participants

Healthy adults between 18 and 49 years of age (n = 4648)

Active arm 1: 295/2344 lost to follow‐up
Controls: 282/2304 lost to follow‐up

Reasons for loss reported for both arms, but numbers do not add up.

Interventions

Trivalent influenza recombinant (haemagglutinin protein) vaccine containing antigens of the 2 A strains and one B strain recommended by WHO in 2007 and 2008 for the Northern Hemisphere. The intervention content was as follows:

Arm 1: 45 mcg of recombinant haemagglutinin antigens (A/Solomon Islands/3/2006 (H1N1), A/Wisconsin/67/2005 (H3N2), and B/Malaysia/2506/2004) with 0.005% polysorbate 20 (Tween‐20) in 10 mM sodium phosphate buffer pH 7.0 ± 0.4 without a preservative. Administered intramuscularly into deltoid muscle.

Placebo: “normal” saline.

Outcomes

Symptomatic influenza cases laboratory confirmed by viral culture. Mild, moderate, and severe adverse events. Industry‐funded study.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No description provided

Allocation concealment (selection bias)

Unclear risk

No description provided

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No description provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Summary assessment

Unclear risk

Loss to follow‐up figures do not match, and there are discrepancies between text and figures.

aa Waldman 1969a

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Randomisation methods were not described. One‐half of the volunteers gave serial blood and nasal wash samples.

Participants

524 schoolteachers: 465 treated and 118 placebo. Age of participants was not indicated.

Interventions

Monovalent inactivated intramuscular vaccine. Schedule and dose were: 1 or 2 doses. Vaccine composition was: A/Hong Kong/68. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Clinical cases and side effects. Clinical case definition was based on the presence of a temperature > 100 °F or a feverish feeling plus any 2 of the following symptoms: sore throat, muscle or joint pain, cough, stuffy or runny nose. Passive surveillance was carried out.

Notes

Data concerning adverse effects were only partially reported by graph. Circulating strain was A2/Hong Kong/68. Effectiveness data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1969b

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. Epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Randomisation methods were not described. One‐half of the volunteers gave serial blood and nasal wash samples.

Participants

590 schoolteachers: 471 treated and 119 placebo. Age of participants was not indicated.

Interventions

Polyvalent inactivated intramuscular vaccine. Schedule and dose were: 1 or 2 doses. Vaccine composition was: A2/Japan/170/62 150 CCA units, A2/Taiwan/1/64 150 CCA units, B/Massachusetts/3/66 300 CCA units. Placebo was saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Clinical cases and side effects. Clinical case definition was based on the presence of a temperature > 100 °F or a feverish feeling plus any 2 of the following symptoms: sore throat, muscle or joint pain, cough, stuffy or runny nose. Passive surveillance was carried out.

Notes

Data concerning adverse effects were only partially reported by graph. Circulating strain was A2/Hong Kong/68. Efficacy data only were extracted.
Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1969c

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Randomisation methods were not described. One‐half of the volunteers gave serial blood and nasal wash samples.

Participants

597 schoolteachers: 479 treated and 118 placebo. Age of participants was not indicated.

Interventions

Monovalent inactivated aerosol vaccine. Schedule and dose were: 1 or 2 doses. Vaccine composition was: A/Hong Kong/68. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Clinical cases and side effects. Clinical case definition was based on the presence of a temperature > 100 °F or a feverish feeling plus any 2 of the following symptoms: sore throat, muscle or joint pain, cough, stuffy or runny nose. Passive surveillance was carried out.

Notes

Data concerning adverse effects were only partially reported by graph. Circulating strain was A2/Hong Kong/68. Efficacy data only were extracted.
Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1969d

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Randomisation methods were not described. One‐half of the volunteers gave serial blood and nasal wash samples.

Participants

590 schoolteachers: 471 treated and 119 placebo. Age of participants was not indicated.

Interventions

Polyvalent inactivated aerosol vaccine. Schedule and dose were: 1 or 2 doses. Vaccine composition was: A2/Japan/170/62 150 CCA units, A2/Taiwan/1/64 150 CCA units, B/Massachusetts/3/66 300 CCA units. Placebo was saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Clinical cases and side effects. Clinical case definition was based on the presence of a temperature > 100 °F or a feverish feeling plus any 2 of the following symptoms: sore throat, muscle or joint pain, cough, stuffy or runny nose. Passive surveillance was carried out.

Notes

Data concerning adverse effects were only partially reported by graph. Circulating strain was A2/Hong Kong/68. Efficacy data only were extracted.
Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1972a

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Identical‐looking, coded vials were used to dispense material. Sampling virological and serological survey of ill people was performed. 2 doses were administered, but as the outbreak occurred mostly between them, only the effectiveness of the first dose was assessed.

Participants

244 volunteer students and staff members: 195 treated and 49 placebo. Age of participants was not indicated.

Interventions

Monovalent A aerosol vaccine. Schedule and dose were: 200 CCA units. Vaccine composition was: A2/Aichi/1/68. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Clinical cases and adverse effects. Clinical cases were defined as febrile respiratory illness with oral temperature higher then 99.5 °F. Local adverse effects were defined as pain and/or tenderness and redness and/or swelling. Systemic adverse effects were defined as general (fever, muscle pain, nausea or vomiting, diarrhoea, and malaise) or respiratory (runny and/or stuffy nose, sore throat, cough, shortness of breath). Passive surveillance was carried out.

Notes

Illness during the first 1 or 2 weeks after vaccination was not excluded, but the authors stated that this fact did not affect the results. Circulating strain was A2/Aichi/2/68. Efficacy and safety data were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1972b

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Identical‐looking, coded vials were used to dispense material. Sampling virological and serological survey of ill people was performed. 2 doses were administered, but as the outbreak occurred mostly between them, only the effectiveness of the first dose was assessed.

Participants

239 volunteer students and staff members: 190 treated and 49 placebo. Age of participants was not indicated.

Interventions

Monovalent A subcutaneous vaccine. Schedule and dose were: 200 CCA units. Vaccine composition was: A2/Aichi/1/69. Placebo was saline for injection. Vaccine was recommended and matched circulating strain.

Outcomes

Clinical cases and adverse effects. Clinical cases were defined as febrile respiratory illness with oral temperature higher then 99.5 °F. Local adverse effects were defined as pain and/or tenderness and redness and/or swelling. Systemic adverse effects were defined as general (fever, muscle pain, nausea or vomiting, diarrhoea, and malaise) or respiratory (runny and/or stuffy nose, sore throat, cough, shortness of breath). Passive surveillance was carried out.

Notes

Illness during the first 1 or 2 weeks after vaccination was not excluded, but the authors stated that this fact did not affect the results. Circulating strain was A2/Aichi/2/68. Efficacy and safety data were extracted. Government funded.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1972c

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Identical‐looking, coded vials were used to dispense material. Sampling virological and serological survey of ill people was performed. 2 doses were administered, but as the outbreak occurred mostly between them, only the effectiveness of the first dose was assessed.

Participants

243 volunteer students and staff members: 194 treated and 49 placebo. Age of participants was not indicated.

Interventions

Bivalent AB aerosol vaccine. Vaccine composition was: A2/Japan/170/62 150 CCA units, A2/Taiwan/1/64 150 CCA units, and B/Massachusetts/3/66 200 CCA units. Placebo was saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Clinical cases and adverse effects. Clinical cases were defined as febrile respiratory illness with oral temperature higher then 99.5 °F. Local adverse effects were defined as pain and/or tenderness and redness and/or swelling. Systemic adverse effects were defined as general (fever, muscle pain, nausea or vomiting, diarrhoea, and malaise) or respiratory (runny and/or stuffy nose, sore throat, cough, shortness of breath). Passive surveillance was carried out.

Notes

Illness during the first 1 or 2 weeks after vaccination was not excluded, but the authors stated that this fact did not affect the results. Circulating strain was A2/Aichi/2/68. Efficacy and safety data were extracted. Government funded.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Waldman 1972d

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1968 to 1969 influenza season. Follow‐up lasted the whole epidemic period. The epidemic curve was traced by absenteeism in the local industries and schools and virus isolation and lasted 7 weeks. Identical‐looking, coded vials were used to dispense material. Sampling virological and serological survey of ill people was performed. 2 doses were administered, but as the outbreak occurred mostly between them, only the effectiveness of the first dose was assessed.

Participants

236 volunteer students and staff members: 187 treated and 49 placebo. Age of participants was not indicated.

Interventions

Bivalent AB subcutaneous vaccine. Vaccine composition was: A2/Japan/170/62 150 CCA units, A2/Taiwan/1/64 150 CCA units, and B/Massachusetts/3/66 200 CCA units. Placebo was saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Clinical cases and adverse effects. Clinical cases were defined as febrile respiratory illness with oral temperature higher then 99.5 °F. Local adverse effects were defined as pain and/or tenderness and redness and/or swelling. Systemic adverse effects were defined as general (fever, muscle pain, nausea or vomiting, diarrhoea, and malaise) or respiratory (runny and/or stuffy nose, sore throat, cough, shortness of breath). Passive surveillance was carried out.

Notes

Illness during the first 1 or 2 weeks after vaccination was not excluded, but the authors stated that this fact did not affect the results. Circulating strain was A2/Aichi/2/68. Efficacy and safety data were extracted. Government funded.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Weingarten 1988

Methods

Randomised controlled trial, double‐blind, conducted in the USA during the 1985 to 1986 influenza season. Follow‐up was not indicated. Epidemic influenza was defined according to population surveillance data (without better explanation), begun in December 1985 and concluded in February 1986. Participants were assigned using a random number generator to receive either the influenza vaccine or placebo. Virological surveillance was not performed.

Participants

179 healthy volunteer hospital employees: 91 treated and 88 placebo. Age of participants was 21 to 65.

Interventions

Split trivalent intramuscular vaccine. Schedule and dose were: single dose; 15 µg each strain. Vaccine composition was: A/Chile/1/83 (H1N1), A/Philippines/2/82 (H3N2), and B/USSR/100/83. Placebo was saline for injection. Vaccine was recommended but did not match the circulating strain.

Outcomes

Clinical cases symptoms defined, WDL regardless of cause, and adverse effects. Influenza illness was defined by the CDC case definition: a documented temperature greater than 100 °F and at least the symptoms of cough or sore throat.

Notes

Data regarding WDL and adverse effects were not complete and they were not considered. Most of the influenza infections were caused by type B.
Efficacy data only were extracted.

Government funded.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Zhilova 1986a

Methods

Semi‐randomised, double‐blind, placebo‐controlled clinical trial conducted in Leningrad, USSR during the 1981 to 1982 influenza season. The study tested the reactogenicity, safety, and effectiveness of an inactivated and a live attenuated vaccine, both administered singly or in combination. Allocation was made on the basis of school classes, and it is unclear whether this is a cluster‐randomised or clinical controlled trial. We have opted for the latter, as the text mentions random selection to maintain "equivalence". "Double blind" is mentioned in the text. During January to May 1982 there was a rise in the level of ILI due to influenza and other agents.

Participants

3961 participants were enrolled. Participants were healthy "students" aged 18 to 23. Numbers in each of the 4 arms are uneven throughout the trial, with no reason provided.

Interventions

Inactivated vaccine trivalent (Ministry of Health USSR) by subcutaneous injection 0.2 mL once (arm 1), or intranasal live "recombinant" "mono" vaccine 0.5 mL spray 2 to 3 times (Ministry of Health USSR) (arm 2), or combined (arm 3), or subcutaneous and intranasal spray sodium chloride saline placebo (arm 4). The strains contained were H1N1, H3N2, and B. Vaccine matching was not good.

Outcomes

Serological
Antibody titres ‐ substudy on 1221 participants
Effectiveness
Influenza‐like illness (not defined and from the text it is unclear how many ILI cases were matched to positive laboratory findings)
Safety

Safety data were not reported in sufficient detail to allow extraction.

Notes

The authors conclude that simultaneous inoculation of the vaccines appeared to produce better humoral antibody responses, especially in the last season. However, the correlation between clinical protection and antibody rises is reported as dubious. The authors make the reasonable point that perhaps live attenuated vaccines work better because they stimulate production of secretory antibodies. This is a poorly reported study. No mention is made of how the placebo could have been correctly used in the schedule (i.e. they should have had 6 arms instead of 4 with subcutaneous placebo, spray placebo administered separately as well as combined; this may be a problem of translation). Efficacy data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

aa Zhilova 1986b

Methods

Semi‐randomised, double‐blind, placebo‐controlled clinical trial conducted in Leningrad, USSR during the 1982 to 1983 influenza season. The study tested the reactogenicity, safety, and effectiveness of an inactivated and a live attenuated vaccine, both administered singly or in combination. Allocation was made based on school classes, and it is unclear whether this is a cluster‐randomised or clinical controlled trial. We have opted for the latter, as the text mentions random selection to maintain "equivalence". "Double blind" is mentioned in the text. In the season there was an outbreak of A (H3N2) lasting 4 to 5 weeks. However, influenza accounted for only up to 30% of isolates from ill people.

Participants

3944 participants were enrolled. Participants were healthy "students" aged 18 to 23. Numbers in each of the 4 arms are uneven throughout the trial, with no reason provided.

Interventions

Inactivated vaccine trivalent (Ministry of Health USSR) by subcutaneous injection 0.2 mL once (arm 1), or intranasal live "recombinant" "mono" vaccine 0.5 mL spray 2 to 3 times (Ministry of Health USSR) (arm 2), or combined (arm 3), or subcutaneous and intranasal spray sodium chloride saline placebo (arm 4). The strains contained were H1N1, H3N2, and B. Vaccine matching was good.

Outcomes

Serological
Antibody titres ‐ substudy on 1221 participants
Effectiveness
Influenza‐like illness (not defined and from the text it is unclear how many ILI cases were matched to positive laboratory findings)
Safety

Safety data were not reported in sufficient detail to allow extraction.
Passive surveillance was carried out.

Notes

The authors conclude that simultaneous inoculation of the vaccines appeared to produce better humoral antibody responses, especially in the last season. However, the correlation between clinical protection and antibody rises is reported as dubious. The authors make the reasonable point that perhaps live attenuated vaccines work better because they stimulate production of secretory antibodies. This is a poorly reported study. No mention is made of how the placebo could have been correctly used in the schedule (i.e. they should have had 6 arms instead of 4 with subcutaneous placebo, spray placebo administered separately as well combined; this may be a problem of translation). Efficacy data only were extracted. Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Atmar 1990

Methods

Double‐blind, placebo‐controlled, randomised trial

Participants

74 healthy volunteers aged 18 to 40 years (data on 17 asthmatics were not extracted)

Interventions

Cold ‐ recombinant vaccine A (H1N1) (n = 16) versus cold ‐ recombinant vaccine A (H3N2) (n = 13) versus cold ‐ recombinant vaccine B (n = 17) versus placebo (n = 26)
Intranasal

Outcomes

Pulmonary function tests (performed on days 0, 3 to 4, 7 after vaccination):

  • FEV1

  • FVC

  • FEV1/FVC

  • Forced expiratory flow rate 25% to 75% (FEF 25 to 75)

Notes

The authors report several non‐significant drops in FEV and FVC up to 7 days' postinoculation and a higher incidence of ILI (17/46 versus 4/26) in the vaccinated arms. Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Betts 1977a

Methods

Randomised controlled trial carried out from April 1976 at Rochester University. Vaccine and placebo were randomly administered in a double‐blind manner, but no description of allocation procedure is given. 36 days after immunisation all participants were challenged with wild type virus (A/Victoria/3/75, H3N2), and antibody response was determined from serum and nasal secretions (before vaccination, 36 hours later, and 21 days after challenge, not for analysis).

Participants

47 healthy male and female university students with absent or low HI titre (i.e. little or no immunity) to both A/Scotland/74 and A/Victoria/3/75.

Interventions

Live attenuated A/Scotland/74 (H3N2) versus placebo, one 0.5 mL dose intranasally. On day 37 after immunisation, participants were challenged with A/Victoria/3/75.

Outcomes

A physician examined the participants 1 day and 4 days after they received vaccine or placebo. Temperature was observed only 1 day after. Observed symptoms were: mild sore throat and rhinorrhoea: vaccine 4/23, placebo 3/24; fever (temperature > 37.50 °C): none had it.

Notes

Safety data only were extracted.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Not used

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Boyce 2000

Methods

Open‐label/single‐blind randomised controlled trial to assess the safety and immunogenicity of adjuvanted and unadjuvanted subunit influenza vaccine, prepared with the strains recommended for and isolated in the 1997 to 1998 season.

Participants

74 healthy adults aged between 10 and 40 years, who did not receive influenza immunisation during the 6 months preceding the trial.

Interventions

  1. M‐59 adjuvanted subunit trivalent flu vaccine (prepared with A/Bayern/795 H1N1, A/Wuhan/359/95 H3N2, B/Beijing/184/93‐like strains, each 15 µg/0.5 mL dose)

  2. Unadjuvanted vaccine (prepared with the same strains at the same concentrations as the adjuvanted preparation)

  3. Placebo (consisting of 0.5 mL sterile saline)

All preparations were intranasally administered in 2 doses 28 days apart. 24 participants received their first dose of adjuvanted (n = 12) or unadjuvanted (n = 12) subunit vaccine in an open‐label manner. After it was determined that they tolerated the first dose, the randomised phase of the trial (n = 50) was begun. In this phase, 18 participants received 2 doses of unadjuvanted vaccine, 19 adjuvanted, and 13 placebo.

Outcomes

After each immunisation, participants were observed for 30 minutes, examined after 2 days, and then completed a diary card reporting symptoms that occurred within 7 days after. Local reactions: nasal symptoms, unpleasant taste, bloody nasal discharge, sneezing. Systemic reactions: chills, pulmonary, nausea, malaise, myalgia or arthralgia, urticarial rash, headache, oral temperature >= 38 °C, stay at home, use of analgesic or antipyretic. Data were not given separately for the randomised and open‐label phase of the study.

Notes

It was not possible to consider the safety data separately for the 2 study phases. Safety data only were extracted.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Caplan 1977

Methods

Randomised controlled trial to assess the reactogenicity and safety of monovalent whole‐virus and split‐virus vaccines prepared with strain A/Victoria/3/75 from different US manufacturers.

Participants

208 healthy adult volunteers aged between 18 and 64 years, recruited from the University of Maryland, USA.

Interventions

Monovalent whole‐virus vaccine (Merck Sharp & Dohme, Merrell‐National Laboratories) or monovalent split‐virus vaccine (Parke‐Davis and Company; Wyeth Laboratories) administered in different antigen concentrations (200, 400, or 800 CCA units) versus placebo. All from A/Victoria75. 1 dose intramuscularly.

Outcomes

Temperature >= 100 °F (37.8 °C), feverishness, pain or burning, tenderness, malaise or myalgia, nausea or vomiting, headache, other. 21‐day follow‐up. Safety outcomes are also given as cumulative % for each category: local, systemic, bothersome; febrile; or scores for systemic reactions.

Notes

Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab El'shina 1996

Methods

Randomised controlled trial

Participants

432 healthy participants aged between 18 and 22 years who had not received any influenza immunisation during the previous 2 to 3 years.

Interventions

Polymer‐subunit influenza vaccine Grippol prepared with the strains A/Victoria/36/88, Wib ‐ 26, B/Panama 45/90. 2 types containing 5 or 2.5 µg haemagglutinin of each strain, respectively were compared with whole‐virion inactivated trivalent vaccine (reference preparation, containing 35 µg of haemagglutinin) and placebo (consisting of sterile physiological solution). One 0.5 mL dose was administered subcutaneously.

Outcomes

After immunisation, participants were placed under medical observation. Fever (48 hours follow‐up): weak (37.1 to 37.5 °C), moderate (37.6 to 38.5 °C), severe (> 38.6 °C). Systemic reactions (3 to 4 days follow‐up): feeling unwell, sore throat, hyperaemia of nasopharynx, head cold, cough, headache, blocked nose, dizziness, shivering, drowsiness, nausea, hoarseness. Local reaction: all (moderate weak); pain at site of injection.

Notes

Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Evans 1976

Methods

Randomised controlled trial

Participants

162 healthy participants aged 18 to 61 years

Interventions

Bivalent live attenuated vaccine WRL 105 (recombinant of A/Okuda/57 and A/Finland/4/74) containing 107.0 EID50 virus/0.5 mL dose versus placebo. Both preparations were administered intranasally 3 to 4 weeks apart.

Outcomes

Reactions to immunisation were observed for 7 days after each dose. Local symptoms (referable to the upper respiratory tract, mainly nasal obstruction, nasal discharge, or sore throat) reported as mild, moderate, or severe. General symptoms (mainly headache, fever, or myalgia). Local and general symptoms are further reported in different intensity classes (mild, moderate, severe, lasting for at least 4 days) reported as mild, moderate, or severe. Use of analgesics.

Notes

Safety data only were extracted.

Funding source ‐ mixed

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Forsyth 1967

Methods

From this report, only the first phase of the first trial is of interest to this review, in which administration of whole‐virus, oil adjuvanted influenza vaccine Invirin (GSK) was compared with placebo in a semi‐randomised allocation. The trial was performed November to December 1962.

Participants

Medical students (n = 380) at the Queen's University of Belfast, UK

Interventions

Trivalent aqueous vaccine (Invirin) one 0.25 mL dose IM containing strains A/Singapore/1/57, A/England/1/61, B/England/939/59. Placebo (phosphate‐buffered saline) was administered as control. Participants born on odd days were given placebo (n = 186); those born on even days received the vaccine (n = 194).

Outcomes

Local reactions: pain, erythema, tenderness, bruises. Stratified by means of scores ranging from 0 to 3 depending on their severity. Systemic reactions: coryza, migraine, paroxysmal tachycardia. All assessed at days 0, 1, 3, 7, 21 after inoculation. Data refer to a 3‐day follow‐up.

Notes

Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Alternate

Allocation concealment (selection bias)

High risk

Not used

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

High risk

Unclear

ab Goodeve 1983

Methods

Randomised controlled trial, double‐blind

Participants

119 healthy young adults from the Medical and Science Faculties of Sheffield University, UK, aged 18 to 19 years without egg allergy

Interventions

Purified subunit monovalent B/Hong Kong/73 flu vaccine prepared in 4 antigen concentrations of 40, 20, 10, 5 µg of HA per each 0.5 mL dose versus saline placebo (0.5 mL dose) subcutaneously administered. Participants were divided into 5 groups of equal dimensions (no further description), each group received 1 of the tested coded preparations. Artificial challenge 1 month later with live attenuated RB77 virus.

Outcomes

Local and systemic reactions were assessed by means of questionnaires completed by participants 24 hours after immunisation. Local reactions (including redness, swelling, itching), local pain (including pain on pressure, pain on contact, continuous pain).

Notes

Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Hrabar 1977

Methods

Randomised controlled trial, double‐blind, carried out during the season 1976 to 1977

Participants

167 students at the technical school in Zagreb, former Republic of Yugoslavia, without sensitivity to egg proteins, pregnancy, acute or chronic diseases

Interventions

Cold‐adapted recombinant A/Victoria/3/75 vaccine administered in 3 different antigen concentrations (107.5, 106.5, 105.5 EID50/0.5 mL) versus placebo. One 0.5 mL dose intranasally.

Outcomes

Participants were medically examined on each of the successive 5 days after immunisation (lasting for at least 1 day). Throat infection, granular palate, oedematous uvula, fever (no cases) as cases and subject‐days. For the following outcomes, authors give the total number of observed cases, without indication of the corresponding arm: malaise, swollen tonsils, fever (1), rhinorrhoea (1), conjunctivitis (7), laryngitis or hoarseness (3), cough (1), swollen tonsils (1), malaise (1). Surveillance was active.

Notes

Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Keitel 1993a

Methods

"The two trials (Keitel 1993a and Keitel 1993b) tested three live attenuated vaccines." This paper reports the results of 2 randomised controlled trials carried out in the USA

Participants

Healthy volunteers recruited at Texas A&M University and Texas Medical Center, aged between 18 and 40 years

Interventions

Two 0.5 mL doses of cold‐adapted recombinant influenza vaccines, 1 month apart, containing 107.1 TCID50 of each strain/dose. 2 studies were conducted in which 4 groups were formed (2 interventions, 2 placebos): 1) placebo 1st and 2nd dose. 2) 1st: A/Kawasaki/9/86 (H1N1, CR 125) + A/Bethesda/1/85 (H3N2, CR90) + B/Ann Arbor/1/86 (B, CRB117)

Outcomes

Mild upper respiratory symptoms. Fever >= 37.8 °C within 1 week after each inoculation

Notes

Safety data only were extracted

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Keitel 1993b

Methods

"The two trials (Keitel 1993a and Keitel 1993b) tested three live attenuated vaccines." This paper reports the results of 2 randomised controlled trials carried out in the USA

Participants

Healthy volunteers recruited at Texas A&M University and Texas Medical Center, aged between 18 and 40 years

Interventions

"Keitel 1993b tested the CR influenza A/Los Angeles/2/87 (H3N2) CR 149 with different lots of CR 125 and CRB 117 used. CR 125, CR 90, and CR 149 express the hemagglutinin and neuraminidase of wild‐type A/Kawasaki (an A/Taiwan/1/86 [H1N1]‐Iike virus), A/Bethesda (an A/Mississippi/1/86 [H3N2]‐l and A/Los Angeles (an A/Sichuan/2/87 [H3N2]‐like virus), respectively, and the internal proteins of cold‐adapted influenza A/Ann Arbor/6/60 (H2N2). CRB 117 expresses the hemagglutinin and neuraminidase of wild‐type influenza B/Ann Arbor/1/ 86 and the internal proteins of cold‐adapted influenza B/Ann Arbor/1/66. Placebo was allantoid fluid”

Outcomes

Mild upper respiratory symptoms. Fever >= 37.8 °C within 1 week after each inoculation

Notes

See Keitel 1993a. Safety data only were extracted

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Langley 2005

Methods

Randomised controlled trial

Participants

Healthy adults aged 18 to 50 years

Interventions

Inactivated A/New Caledonia/20/99 (H1N1) + A/Panama/2007/99 (H3N2) + B/Guangdong/120/2000 non‐covalent associated with outer membrane protein of Neisseria meningitidis. Single nasal dose containing 15, 30, 45 µg versus placebo (phosphate‐buffered saline) intranasally administered.

Outcomes

Local: within 7 days, graphic: rhinorrhoea, congestion, itch/burn, nosebleed, red/puffy eyes, sneezing, sore throat. Systemic: within 7 days: cough, shortness of breath, headache, muscle/joint aches, poor appetite, fatigue within 48 hours, nasal mucosa inflammation, nasal discharge, pharyngeal inflammation, sinusitis, enlarged cervical/postauricular nodes.

Notes

Safety data only were extracted.

Government and industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

High risk

Inadequate

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

High risk

High risk

ab Lauteria 1974

Methods

Controlled trial. Randomisation procedure was neither described nor mentioned. Participants were paired according to age and sex, 1 participant in each pair received vaccine, the other placebo. Double‐blind

Participants

37 volunteers aged 18 to 24 years, with titre of serum neutralising antibodies to A/Hong Kong/8/68 > 1:16

Interventions

Live attenuated A/England/8/68 grown in presence of heated equine serum. Two 0.5 mL doses containing 104 TCID50 of this strain or placebo (0.85% sodium chloride) were administered intranasally 2 to 3 weeks apart

Outcomes

Participants observed for 4 days, beginning 24 hours after immunisation. Fever, myalgia, rhinitis, cough, pharyngitis

Notes

Safety data only were extracted.

Government and industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Miller 1977

Methods

Randomised controlled trial

Participants

43 seronegative healthy adults aged between 22 and 50 years

Interventions

Live attenuated serum inhibitor resistant flu B vaccine R75 (a recombinant of B/Hong Kong/5/72 with B/Russia/69) containing 107.2 EID50 of R75/0.5 mL dose versus placebo (sucrose 5%). Intranasal, 2 doses, 2 weeks apart

Outcomes

Participants were interviewed during the 5 days following each immunisation. Local reaction (defined as immediate complains and comprising bad taste or burning, lasting for a few moments). Systemic reaction (consisting essentially of headache and rhinorrhoea)

Notes

Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Pyrhönen 1981

Methods

Randomised controlled trial conducted in the 1976 to 1977 season in Finland

Participants

307 healthy adults

Interventions

1 of the following 4 preparations was administered to 1 of the 4 study arms. “Volunteers were inoculated with bivalent subunit influenza vaccine containing

1200 IU of strain A/Victoria/3/75 (H3N2) antigen and 800 IU of B/Hongkong/8/73 antigen in 0.5 ml of phosphate or phosphate‐buffered saline solution as placebo”

Outcomes

Harms assessed by questionnaires filled out by each participant within 3 days after immunisation. Fever: vaccine 11/151, placebo 9/154; muscle ache: vaccine 26/151, placebo 12/154; redness: vaccine 53/151, placebo 3/154; tenderness at vaccination site: vaccine 89/151, placebo 12/154; tenderness of axillary glands: vaccine 6/151, placebo 2/154

Notes

Safety data only were extracted

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Reeve 1982

Methods

Randomised controlled trial carried out in Wien

Participants

20 university students aged 20 to 24 years

Interventions

1st phase: cold‐recombinant, live flu vaccine II RB‐77 (B/Ann Arbor/1/66 and B/Tecumse/10/77) containing 107.2 EID50 per 0.5 mL dose versus placebo. 1 dose intranasally. During this phase, participants lived under sequestered condition, and close contact between vaccine and placebo recipients was possible. 2nd phase: 3 weeks after the 1st dose all participants were immunised with 1 dose of the same vaccine

Outcomes

During the 5 days following immunisation, participants were medically observed and temperature recorded morning and evening. Occurring symptoms were attributed scores (0 to 3) depending on their severity (no, light, moderate, severe). Fever (oral temperature > 38 °C): 0/10, 0/10; sneezing: 1/10, 0/10; stuffy nose: 7/10, 1/10; running nose: 3/10, 0/10; afebrile subjective symptoms: 8/10, 2/10

Notes

Safety data only were extracted

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Rocchi 1979a

Methods

Cluster‐randomised controlled trial carried out during the 1976 to 1977 season

Participants

496 healthy military recruits (aged 18 to 20 years) belonging to 4 different companies from "Scuola Allievi Sottoufficiali" in Viterbo, Italy

Interventions

1 of the following 4 preparations were administered to 1 of the 4 study arms: 1. live attenuated A/Victoria/3/75; two 2 mL doses (2 104.5 EID50/dose) oral. 2. Live attenuated recombinant A/Puerto Rico/8/34, A/Victoria/3/75; two 0.5 mL doses intranasally (107 EID50/dose). 3. Inactivated A/Victoria/3/75 (600 IU), B/Hong Kong/5/72 (300 IU) and AlPO4, intramuscular placebo (vaccine diluent) administered intranasally. The 2 doses were administered 2 to 3 weeks apart.

Outcomes

Within 15 days after administration of the 1st dose. Malaise, myalgia, headache, sore throat, cough, fever >= 38.5 °C, fever >= 37.5 °C, 3 or more symptoms, any symptoms. Surveillance was passive.

Notes

Units of randomisation appear to be companies. No description of manner of allocation is mentioned. Blind (only for the cases of intranasal administration). Influenza outbreak occurred when the immunisation began (intraepidermic study).
Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Rocchi 1979b

Methods

See ab Rocchi 1979a

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Not used

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Saxen 1999

Methods

Randomised controlled trial, double‐blind, conducted in Finland during the 1996 to 1997 influenza season. Randomisation methods were not described.

Participants

216 healthcare workers: 211 treated and 427 placebo

Interventions

Trivalent inactivated intramuscular vaccine. Schedule and dose were: single dose; 15 µg each strain. Vaccine composition was: A/Wahan/359/95, A/Singapore/6/86, and B/Beijing/184/93. Placebo was saline for injection. Vaccine was recommended.

Outcomes

Working days lost due to respiratory infections, episodes of respiratory infections, days ill, and antimicrobial prescriptions. Respiratory infection was a common cold; febrile ILIs were not detected. Local adverse effects were defined as local pain. Systemic adverse effects were defined as fever and fatigue.

Notes

Efficacy data were not extracted because episodes of respiratory infections were unclearly defined. Safety data only were extracted.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

ab Scheifele 2003

Methods

Randomised, double‐blind, placebo‐controlled, cross‐over trial assessing the association between exposure to the vaccine and onset of ORS in healthy adults with no previous history of ORS. The trial took place in 5 centres in Canada in September 2001 and was 1 of the conditions of registration of the vaccine, given the high incidence of ORS in the previous season. Centralised randomisation and allocation of centrally prepared, coded, opaque syringes took place. Cross‐over to either vaccine or placebo took place 5 to 7 days after the initial injection.

Participants

The study included 651 adults with a mean age of 45. 17 participants are unaccounted for.

Interventions

Fluviral (Shire) split trivalent containing A/New Caledonia/20/99 (H1N1), A/Panama/2007/99 (H3N2), B/Victoria/504/2000 with additional splitting with Triton X‐100 splitting agent or saline placebo 0.5 mL. Additional splitting was necessary to test the hypothesis that large clumps of virions were responsible for the ORS seen the previous season.

Outcomes

ORS (bilateral conjunctivitis; facial swelling ‐ lip, lid, or mouth; difficulty in breathing and chest discomfort, including cough, wheeze, dysphagia, or sore throat). Local signs/symptoms (redness, swelling, pain). Follow‐up was by phone interview at 24 hours and 6 days after vaccination.

Notes

The authors conclude that (mild) ORS is significantly associated with split TIV immunisation (attributable risk 2.9%, 0.6 to 5.2). Other adverse effects associated with TIV are hoarseness (1.3%, 0.3 to 1.3) and coughing (1.2%, 0.2 to 1.6). The study is good quality, and the authors' conclusions are robust. It is extraordinary that no one has looked for these symptoms before, but it may be that the relatively young age of participants and the hypothesis contributed to this. Safety‐only study.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block randomisation

Allocation concealment (selection bias)

Low risk

Adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Adequate

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Adequate

Summary assessment

Low risk

Adequate

ab Spencer 1977

Methods

Controlled trial, single‐blind

Participants

21 pairs of students and employers at the University of California, aged between 24 and 50 years who lived together or worked in close proximity

Interventions

Recombinant, live attenuated R 75 vaccine (B/Hong Kong/5/72 and B/Russia/69) containing 107.5 EID/dose versus placebo (allantoic fluid). Lyophilised vaccine was supplied by Smith, Kline & French Laboratories and diluted with 2.5 mL of a 5% sucrose solution just before administration. Both preparations were administered intranasally (5 drops/nostril). In each pair 1 individual received vaccine and the other placebo. A second dose was administered 14 days apart.

Outcomes

Any clinical symptoms within 7 days after each immunisation (rhinitis, cough, pharyngitis, headache, malaise, and myalgia were the prominently observed symptoms, but given as aggregates).

Notes

Reported safety data do not allow quantitative analysis.

Industry funded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Unclear

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

bb Dauvilliers 2013

Methods

Case‐control study investigating the association between exposure to monovalent, 2009‐10 pandemic H1N1 vaccines and onset of narcolepsy

Participants

Cases (n = 59): were identified from 14 French expert orphan disease narcolepsy centres among participants referred to 1 of the participating sleep centres to confirm the diagnosis by polysomnography as well as the Multiple Sleep Latency Test between 1 October 2009 and 30 April 2011 (according to the International Classification of Sleep Disorders definition, ICSD 2005). Participating centres identified retrospectively from lists of medical records completed by reference centres for orphan diseases as required by the French government and from hospital statistic databases all their patients with narcolepsy‐cataplexy potentially matching the eligibility criteria. All potentially eligible cases were asked to participate, and their clinical history was revised to confirm the diagnosis of narcolepsy‐cataplexy following the criteria of the Brighton Collaboration, levels 1 to 3.

Controls (n = 135): were selected among patients from the hospitals to which the participating sleep centres belonged and among healthy volunteers from a national database (Narcobank). Up to 4 controls were matched to each case for sex, age, geographic location.

Only 25 cases and 73 controls were at least 18 years old.

Interventions

Exposure to Pandremix (AS03 adjuvanted) or Panenza (not adjuvanted) monovalent p H1N1 influenza vaccines. Vaccination was ascertained by means of a phone interview, during which other data were also recorded (body mass index, smoking, medical history, history of viral or bacterial infections), and confirmed by vaccination certificates. Date of first disease symptoms was reported.

A sensitivity analysis was carried out considering as index date:

  1. the date of narcolepsy‐cataplexy diagnosis;

  2. the date of Multiple Sleep Latency Test; or

  3. the date the first symptoms appeared.

Participants were considered vaccinated if they received vaccination before this latter date (whatever analysis authors performed). Data analysis was performed excluding and including cases for whom symptom onset did occur concomitantly or shortly before vaccination, so that it was not possible to state whether vaccination had effectively been administered before the onset of first symptoms, from analyses 1 and 2. (They remained always included in analysis 3).

Effect estimates were moreover performed considering as exposed those participants who received AS03‐adjuvanted pandemic vaccine only.

Outcomes

Narcolepsy‐cataplexy (Brighton Collaboration levels 1 to 3)

Notes

Mixed (?)

This was not an industry‐supported study. This study was funded by grants from the Agence Nationale de Sécurité du Médicament et des Produits de Santé, the European Centre for Disease Prevention and Control, and the PHRC AOM07‐138 grant from the French Health Ministry. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Y Dauvilliers has received funds for speaking and board engagements with UCB, Cephalon, Jazz, Novartis, and Bioprojet. P Franco has received funds for speaking and board engagements with UCB. MP d Ortho has received funds for speaking from Cephalon and board engagements with Bioprojet. C Monaca Charley has received funds for speaking or board engagements, or both with UCB, Novartis, and Cephalon. M Lecendreux has received funds for speaking and board engagements with UCB and Bioprojet.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Unclear risk

Record linkage. Medical record reviewed, and participants fulfilling levels 1 to 3 of Brighton Collaboration definition included as cases. Recruited through 14 centres across France. It is possible that healthcare professionals were over‐represented.

CC ‐ control selection
All outcomes

Unclear risk

Hospital controls

CC ‐ comparability
All outcomes

Unclear risk

Cases and controls were matched only for age, sex, and geographical area.

CC ‐ exposure
All outcomes

Low risk

Vaccination records

Summary assessment

Unclear risk

Unclear risk of bias

bb DeStefano 2003

Methods

Case‐control study

Participants

Data from Vaccine Safety Datalink (large database of cases of disease following vaccination) in the USA

Interventions

Immunisation with influenza and other vaccines assessed by means of medical records.

Outcomes

Cases: physician diagnosis of multiple sclerosis or optic neuritis in medical record
Controls: up to 3 controls per case were selected from automated HMO member files, at least 1 year of HMO enrolment, matched on age (within 1 year) and gender

Notes

Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

From HMO registry

CC ‐ control selection
All outcomes

Low risk

From HMO registry

CC ‐ comparability
All outcomes

Unclear risk

Poor matching

CC ‐ exposure
All outcomes

Unclear risk

From registry and telephone interview

Summary assessment

Unclear risk

Unclear

bb Dieleman 2011a

Methods

Case‐control study

Participants

Cases (n = 145): Guillain‐Barré syndrome cases (defined according to the Brighton Collaboration definition) diagnosed in France between 2007 and 2010.
Controls (n = 1080): the dates for control recruitment were matched (by calendar month) to the index date of the associated case. Additional matching criteria included gender, age (65 years for cases aged 18 years or more and 61 years for cases younger than 18 years) and place of residence (southern or northern France).

Interventions

Exposure to influenza vaccine. Data about pandemic vaccine analysed separately.
Exposure to virus and occurrence of ILI also tested as risk factor.

Outcomes

Association between Guillain‐Barré syndrome and influenza vaccine exposure

Notes

The study has been financially supported by LA‐SER, GSK Biologicals, and Sanofi Pasteur.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Unclear risk

From different countries

CC ‐ control selection
All outcomes

Unclear risk

Not same population, insufficient description

CC ‐ comparability
All outcomes

Unclear risk

Matching

CC ‐ exposure
All outcomes

Unclear risk

Interview

Summary assessment

High risk

High risk of bias

bb Dieleman 2011b

Methods

Case‐control study

Participants

Cases (n = 145): Guillain‐Barré syndrome cases (defined according to the Brighton Collaboration definition) diagnosed in France between 2007 and 2010.
Controls (n = 1080): the dates for control recruitment were matched (by calendar month) to the index date of the associated case. Additional matching criteria included gender, age (65 years for cases aged 18 years or more and 61 years for cases younger than 18 years), and place of residence (southern or northern France).

Interventions

Exposure to influenza vaccine. Data about pandemic vaccine analysed separately.
Exposure to virus and occurrence of ILI also tested as risk factor.

Outcomes

Association between Guillain‐Barré syndrome and influenza vaccine exposure

Notes

The study has been financially supported by LA‐SER, GSK Biologicals, and Sanofi Pasteur.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Low

CC ‐ control selection
All outcomes

Unclear risk

Not same population

CC ‐ comparability
All outcomes

Unclear risk

Matching

CC ‐ exposure
All outcomes

Unclear risk

Interview

Summary assessment

Unclear risk

Unclear

bb Dieleman 2011c

Methods

Case‐control study

Participants

Cases (n = 145): Guillain‐Barré syndrome cases (defined according to the Brighton Collaboration definition) diagnosed in France between 2007 and 2010.
Controls (n = 1080): the dates for control recruitment were matched (by calendar month) to the index date of the associated case. Additional matching criteria included gender, age (65 years for cases aged 18 years or more and 61 years for cases younger than 18 years), and place of residence (southern or northern France).

Interventions

Exposure to influenza vaccine. Data about pandemic vaccine analysed separately.
Exposure to virus and occurrence of ILI also tested as risk factor.

Outcomes

Association between Guillain‐Barré syndrome and influenza vaccine exposure

Notes

The study has been financially supported by LA‐SER, GSK Biologicals, and Sanofi Pasteur.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Low

CC ‐ control selection
All outcomes

Unclear risk

Not same population

CC ‐ comparability
All outcomes

Unclear risk

Matching

CC ‐ exposure
All outcomes

Unclear risk

Interview

Summary assessment

Unclear risk

Unclear

bb Dieleman 2011d

Methods

Case‐control study

Participants

Cases (n = 145): Guillain‐Barré syndrome cases (defined according to the Brighton Collaboration definition) diagnosed in France between 2007 and 2010.
Controls (n = 1080): the dates for control recruitment were matched (by calendar month) to the index date of the associated case. Additional matching criteria included gender, age (65 years for cases aged 18 years or more and 61 years for cases younger than 18 years), and place of residence (southern or northern France).

Interventions

Exposure to influenza vaccine. Data about pandemic vaccine analysed separately.
Exposure to virus and occurrence of ILI also tested as risk factor.

Outcomes

Association between Guillain‐Barré syndrome and influenza vaccine exposure

Notes

The study has been financially supported by LA‐SER, GSK Biologicals, and Sanofi Pasteur.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Low

CC ‐ control selection
All outcomes

Unclear risk

Not same population

CC ‐ comparability
All outcomes

Unclear risk

Matching

CC ‐ exposure
All outcomes

Unclear risk

Interview

Summary assessment

Unclear risk

Unclear

bb Dieleman 2011e

Methods

Case‐control study

Participants

Cases (n = 145): Guillain‐Barré syndrome cases (defined according to the Brighton Collaboration definition) diagnosed in France between 2007 and 2010.
Controls (n = 1080): the dates for control recruitment were matched (by calendar month) to the index date of the associated case. Additional matching criteria included gender, age (65 years for cases aged 18 years or more and 61 years for cases younger than 18 years), and place of residence (southern or northern France).

Interventions

Exposure to influenza vaccine. Data about pandemic vaccine analysed separately.
Exposure to virus and occurrence of ILI also tested as risk factor.

Outcomes

Association between Guillain‐Barré syndrome and influenza vaccine exposure

Notes

The study has been financially supported by LA‐SER, GSK Biologicals, and Sanofi Pasteur.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Low

CC ‐ control selection
All outcomes

Unclear risk

Not same population

CC ‐ comparability
All outcomes

Unclear risk

Matching

CC ‐ exposure
All outcomes

Unclear risk

Interview

Summary assessment

Unclear risk

Unclear

bb Galeotti 2013

Methods

Case‐control study testing the association between influenza vaccination and Guillain‐Barré syndrome

Participants

Cases (n = 140): adults with Guillain‐Barré syndrome defined according to the Brighton Collaboration definition (levels 1 to 3) recruited at 121 neurological centres in 7 Italian regions and having symptoms onset between 1 October 2010 and 15 May 2011.

Controls (n = 308): were selected from among patients admitted to the emergency department of the same hospital as the cases for acute conditions unrelated to chronic diseases (e.g. trauma). Each control was individually matched to a case for admission date (i.e. the same date as the case or up to 30 days afterwards), sex, age (± 5 years), and region of residence.

Interventions

Exposure to influenza vaccination (date and brand of vaccine) was verified by contacting patients’ general practitioners by telephone. A neurologist (FG) closely verified and queried data quality.

Outcomes

Guillain‐Barré syndrome

Notes

The authors also performed data analysis with a controlled case series design, considering the 6 weeks following exposure as the risk time.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Consecutive series of cases

CC ‐ control selection
All outcomes

Low risk

Hospital control

CC ‐ comparability
All outcomes

Unclear risk

Matched analysis only for sex, age, region, admission date

CC ‐ exposure
All outcomes

Unclear risk

Unclear if interviewers were blinded to case‐control status

Summary assessment

Unclear risk

Unclear risk of bias

bb Garbe 2012

Methods

Case‐control surveillance study

Participants

Cases (n = 169): patients 18 years of age or older with a diagnosis of certain or probable immune thrombocytopenia. Of the included 169 cases, 130 were outpatients and 39 were inpatients.
Controls (n = 770): 731 outpatients and 39 inpatients selected from the same hospitals as the cases. The index date for outpatient controls was defined as the date of hospitalisation or the date of initiation of the control disease episode if this preceded hospitalisation. The index date for inpatient controls was the date of the interview.

Interventions

Exposure to influenza vaccination during the 28 days preceding the index date. Exposure to other vaccines and drugs was also considered.

Outcomes

Immune thrombocytopenia

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Hospital population

CC ‐ control selection
All outcomes

Low risk

Hospital control

CC ‐ comparability
All outcomes

Unclear risk

No matching

CC ‐ exposure
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

bb Grimaldi‐Bensouda 2011

Methods

Multicentre, case‐control study

Participants

Cases (n = 104): Guillain‐Barré syndrome cases (Brighton Collaboration definition, levels 1 to 3).
Controls (n = 1198): each case was matched to up to 25 controls on age (plus or minus 1 year), sex, index date, and country. Matched controls recruited in the Netherlands, Sweden, the UK, France, and Denmark.

Interventions

Exposure to monovalent pandemic H1N1 2009 to 2010 influenza vaccine during the 6 months preceding the index date. Vaccination data were obtained from vaccine registries (Denmark and France), from general practitioner records (UK and Netherlands), and from structured interviews (Sweden).

Outcomes

Guillain‐Barré syndrome

Notes

This study was funded by the European Centre for Disease Prevention and Control.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Neurological clinic registry

CC ‐ control selection
All outcomes

Unclear risk

From the same population using only general practitioner registry

CC ‐ comparability
All outcomes

Unclear risk

Poor matching

CC ‐ exposure
All outcomes

Unclear risk

Interview and record linkage

Summary assessment

Unclear risk

Unclear

bb Grimaldi‐Bensouda 2012

Methods

Case‐control study

Participants

Cases (n = 198) were people with a diagnosis of immune thrombocytopenia (American Society of Hematology diagnostic criteria) identified with the collaboration of 22 university and major regional hospitals in France participating in the Pharmacoepidemiological General Research on ITP (PGRx‐ITP) registry project.

Controls (n = 878) matched on age (2 years), sex, region of residence (northern or southern France), index date (date of first symptoms for the cases and date of consultation for the referents 2 months) from a random sample.

Interventions

Exposure to influenza vaccine. Assessed by structured interview and confirmed by vaccination records.

Outcomes

Immune thrombocytopenia

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Multicentre registry consecutive series of cases

CC ‐ control selection
All outcomes

Unclear risk

Same population using registry from a sample of GPs

CC ‐ comparability
All outcomes

Unclear risk

Matching 1:5

CC ‐ exposure
All outcomes

Unclear risk

Structured interview ‐ confirmation by GPs

Summary assessment

Unclear risk

Unclear

bb Hernan 2004

Methods

Case‐control study based on the General Practice Research Database (GPRD)

Participants

Cases (n = 163): patients with confirmed diagnosis of multiple sclerosis between 1 January 1993 and 31 December 2000.

Controls (n = 1604): subjects from the GPRD matched to the cases for age, sex, practice, date of joining the practice.

Interventions

Exposure to vaccinations (also influenza) as shown from medical records

Outcomes

Association between exposure to influenza vaccine and onset of multiple sclerosis

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Nested case‐control from GPRD registry

CC ‐ control selection
All outcomes

Low risk

GPRD registry

CC ‐ comparability
All outcomes

Low risk

Matched

CC ‐ exposure
All outcomes

Low risk

Registry

Summary assessment

Low risk

Low

bb MacIntyre 2013

Methods

Case‐control study investigating the protective effect of influenza vaccination against acute myocardial infarction

Participants

Cases (n = 275): patients aged ≥ 40 years of age admitted with an acute myocardial infarction, evolving or recent myocardial infarction to the cardiology unit during the influenza season. Eligible respondents were those able to provide samples within 72 hours of the acute myocardial infarction event, residing in Sydney, Australia, available for follow‐up, and provided informed consent. Cases reporting a previous cardiovascular event were eligible. A diagnosis of acute myocardial infarction was defined as a typical rise and gradual fall in troponin or more rapid rise and fall in creatine kinase‐MB biochemical markers of myocardial necrosis, with 1 or more of the following: ischaemic symptoms (chest or arm pain, nausea/vomiting, sweating, shortness of breath); development of pathological Q waves on ECG; ECG changes indicative of ischaemia (ST segment elevation or depression); coronary artery intervention; or pathological findings of an acute myocardial infarction. Participants were recruited into the study between 27 June and 20 October 2008; 18 May and 23 October 2009; and 21 June and 28 October 2010.

Controls (n = 284): controls were people aged ≥ 40 years of age attending the orthopaedic or ophthalmic outpatient clinics during the same time period. Respondents residing in Sydney, available for follow‐up, and able to provide informed consent were eligible. Controls were unmatched, except for the same age cut‐off and recruitment period, to ensure similar level of exposure to circulating influenza. Controls were excluded if they reported a history of acute myocardial infarction, transient ischaemic attack, or stroke in the previous 12 months. Stable angina was permissible if there had been no worsening of angina or acute myocardial infarction episodes or hospital admissions in the last year. Controls were recruited into the study between 30 June and 31 October 2008; 19 May and 26 October 2009; and 23 June and 29 October 2010.

Interventions

Influenza vaccination status was validated for current and previous influenza seasons from hospital and GP records, with GPs contacted via facsimile or telephone. If discrepancies arose between GP and self report, GP‐reported vaccination status was considered correct. Self reported vaccination status was considered sufficient in those individuals whose GP could not be contacted. Type and characteristics of the administered vaccines are not provided.

Outcomes

Notes

Funding source ‐ industry

This work was supported by a grant from GlaxoSmithKline, Belgium. Dr Iman Ridda and Dr Holly Seale are supported by Australian National Health and Medical Research Council Training Fellowships.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Consecutive series of cases (patients admitted to the cardiology unit during the influenza season)

CC ‐ control selection
All outcomes

Unclear risk

Community controls (patients attending orthopaedic or ophthalmic outpatient clinics during the same period without history of disease)

CC ‐ comparability
All outcomes

Unclear risk

Unmatched

CC ‐ exposure
All outcomes

Low risk

Vaccination certificate, GP records

Summary assessment

Unclear risk

Unclear risk of bias

bb Mastrangelo 2000

Methods

Case‐control study assessing the association between influenza vaccines and cutaneous melanoma

Participants

99 cases and 104 controls

Interventions

Influenza vaccine exposure is not described.

Outcomes

Notes

The authors report a protective effect of repeated influenza vaccination on the risk cutaneous melanoma (OR 0.43, 95% CI 0.19 to 1.00). The study is at high risk of bias due to the selective nature of cases (all patients in the authors' hospital), attrition bias (4 cases and 4 controls eliminated because of "failure to collaborate"), recall bias (up to 5 years' exposure data were based on patients' recollection), and ascertainment bias (non‐blinded exposure survey).
Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Low

CC ‐ control selection
All outcomes

Unclear risk

Insufficient information

CC ‐ comparability
All outcomes

Unclear risk

Insufficient information

CC ‐ exposure
All outcomes

Low risk

Low

Summary assessment

Unclear risk

Unclear

bb Mutsch 2004

Methods

1 case‐control study and case series based in the German‐speaking regions of Switzerland, which assessed the association between an intranasal inactivated virosomal influenza vaccine and Bell's palsy

Participants

250 cases that could be evaluated (from an original 773 cases identified) were matched to 722 controls for age and date of clinic visit. All participants were around age 50.

Interventions

Immunisation with influenza vaccine took place within 91 days before disease onset.

Outcomes

Bells' palsy

Notes

The study reports a massive increase in risk (adjusted OR 84, 95% CI 20.1 to 351.9) within 1 to 91 days since vaccination. Despite the many limitations of the study (case attrition: 187 cases could not be identified; ascertainment bias: physicians picked controls for their own cases; confounding by indication: different vaccine exposure rate between controls and the reference population), it is unlikely that such a large OR could have been significantly affected by systematic error. The authors called for larger pre‐licence safety trials, given the rarity of Bell's palsy. On the basis of this study the vaccine was withdrawn commercially.
Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

"All 4891 primary care physicians, ear, nose, and throat specialists, and neurologists in the study area were invited twice to report cases of Bell’s palsy first diagnosed between October 1, 2000, and April 30, 2001."

CC ‐ control selection
All outcomes

Low risk

Subsequently, the physicians who had reported cases of Bell’s palsy were asked to document the date of the visit and information pertinent to the study’s inclusion and exclusion criteria and to select from among their patients without Bell’s palsy, 3 controls sequentially from their registration log.

Trained study monitors contacted the physicians and reviewed the selection forms regularly to ensure consistency in the selection of controls. At this point, participating physicians had not been made aware of the exposure to be investigated (influenza vaccination).

CC ‐ comparability
All outcomes

Unclear risk

The controls were matched with the case patients according to age (within 5 years), date of the clinic visit (within 4 days), and physician.

CC ‐ exposure
All outcomes

Low risk

"Physicians were asked to document the dates of administration and the brand name and type of influenza vaccine (parenteral or intranasal) used during the study period. Other vaccine exposures during the study period and the preceding 2 months were also documented. Since in all 43 sentinel cases reported in the study area the onset of Bell’s palsy occurred within 91 days after intranasal vaccination, we defined the period of 1 to 91 days as the postexposure risk period."

Summary assessment

Unclear risk

Unclear

bb Payne 2006

Methods

Case‐control study assessing the association between influenza and other vaccines (data not extracted for this review) and optic neuritis.

"A matched case‐control study design was used with each optic neuritis case matched to 3 controls based on sex, deployment during the 18 weeks preceding the diagnosis date, and the military component in which the individual served (eg, active or reserve/National Guard). The protocol for this vaccine postmarketing surveillance investigation was approved by the Centers for Disease Control and Prevention (CDC) Institutional Review Board and reviewed by the Food and Drug Administration and Department of Defense"

Participants

US military personnel aged at least 18 years

Interventions

Cases (n = 1131): participants with a diagnosis of optic neuritis between 1 January 1998 and 31 December 2003. The following ICD‐9 codes were considered: 377.30‐32, 377.39.
Controls (n = 4524): participants were matched to the cases on the basis of sex, deployment during the 18 weeks before diagnosis, military component. The study was carried out using data from the Defense Medical Surveillance System (DMSS), a longitudinal surveillance database.

Outcomes

Date of case diagnosis was ascertained, and immunisation status (anthrax, smallpox, hepatitis B, influenza) verified by means of electronic records with respect to 3 time intervals: 6, 12, and 18 weeks before onset. For controls, vaccination status was determined for the 3 intervals before the index date. Results were focused on the 18‐week time interval.

Notes

Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

"We defined optic neuritis cases as those having a first‐time diagnosis of the following ICD‐9‐CM codes: optic neuritis, unspecified (377.30); optic papillitis (377.31); retrobulbar neuritis, acute (377.32); and optic neuritis, other (377.39) during the period between 1 January 1998 and 31 December 2003."

CC ‐ control selection
All outcomes

Low risk

Controls were selected if their DMSS diagnostic records indicated no history of an optic neuropathy, if they served in the military on the same date of diagnosis as their matched case, and if they had at least 18 weeks of military service preceding this index date.

CC ‐ comparability
All outcomes

Low risk

Matching

CC ‐ exposure
All outcomes

Low risk

"We ascertained the date of each case’s first diagnosis of optic neuritis and determined all vaccinations received during each of the following 3 prior study intervals from the electronic record; 6 weeks (42 days), 12 weeks (84 days) and 18 weeks (126 days). For each of the 3 matched controls, we determined all vaccinations during the 3 intervals predating their index date."

Summary assessment

Low risk

Low

bb Ray 2011

Methods

Case‐control study

Participants

Cases (n = 415): people with diagnosis of definite rheumatoid arthritis based on American College of Rheumatology criteria.

Controls (n = 1245): matched for age and number of medical visits before index date.

Interventions

Exposure to influenza vaccine. Different times intervals before symptom onset were considered (90, 180, 365, and 730 days). Vaccine exposure status was determined from Kaiser Immunization Tracking System and supplemented by chart reviews. Risk of association was, moreover, also determined for tetanus and hepatitis B vaccines.

Outcomes

Notes

This study was funded by the Centers for Disease Control and Prevention Vaccine Safety Datalink Project.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Included as cases the incident cases from the cohort analysis and additional new onset cases identified from the study population whose symptoms began during 1996.

CC ‐ control selection
All outcomes

Low risk

Same population

CC ‐ comparability
All outcomes

Unclear risk

Poor matching

CC ‐ exposure
All outcomes

Low risk

NCKPHP databases

Summary assessment

Unclear risk

Unclear

bb Rouleau 2014

Methods

Case–control study aiming to identify what exposure factors could be linked to allergic‐like events in the general adult population

Participants

Cases of allergic‐like events were identified starting from the Quebec Adverse Events Surveillance System reviewing allergic‐like events that occurred 5 to 8 months after the vaccination campaign (between May and July 2010) and that were classified as “anaphylaxis”, ORS, or “allergy”. Cases with these diagnoses were contacted and interviewed by trained nurses who used a standardised phone questionnaire to verify the diagnosis by applying the Brighton Collaboration Criteria for Anaphylaxis and the National Advisory Committee on Immunization for ORS (bilateral red eyes, and/or facial swelling, and/or respiratory symptoms (cough, wheeze, chest tightness, difficulty breathing, difficulty swallowing/throat tightness, hoarseness or sore throat) with onset ≤ 24 hours after influenza vaccination); cases that met neither definition were considered as allergic‐like events (immediate or delayed allergic‐like events, depending on whether symptoms occurred within 4 hours after vaccination or thereafter, i.e. immediate allergic‐like events or delayed allergic‐like events).

Controls were randomly selected from the Pandemic Influenza Vaccination Registry by age and sex matching of each case with 2 controls.

Trained nurses collected by interview data about demographics, personal and family medical conditions, obstetric history (gravida, para, abortus), use of medication within 48 hours of vaccination, the presence of an acute respiratory illness at the time of vaccination (e.g. fever, respiratory infection, or ILI), reported allergy to potential allergenic components of the vaccine (i.e. eggs, fish, shellfish, thimerosal, latex), regular alcohol use, and physical activity.

In all, 471 cases and 849 controls were identified. Of these 36 (6%) and 136 (16%) refused to participate, resulting in 435 cases (50 anaphylaxis, 177 ORS, 97 immediate allergic‐like events, and 111 delayed allergic‐like events) and 849 controls.

Interventions

A univariate analysis and multivariate logistic regression were performed with the aim of identifying potential risk (aeroallergens, drug allergy, food allergy, dermographism, hypothyroidism, family history, allergy, administration of drugs for obstructive airway disease, healthcare worker as profession, vaccinated in weeks) or protective factors (administration of anti‐inflammatory or mineral supplements, being physically active, consumption of alcohol). Separate unconditional regression models were built for each case definition and effect estimate (odds ratio) adjusted for sex and age group.

Outcomes

Exposure to vaccination with pandemic, monovalent, AS03‐adjuvanted H1N1 vaccine (Arepanrix, GSK) during the first 4 weeks of the immunisation campaign

Notes

Funding source ‐ government

Exposure to vaccination is considered within the first 4 weeks of campaign (i.e. not vaccinated versus vaccinated). Among the group of vaccinated within the 4 campaign weeks, healthcare workers were strongly represented (for this group there was also significant association with all outcomes), which could have introduced a certain recall/selection bias.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Review of surveillance register by applying case definition during phone interview. Drawn from a nationwide active surveillance register

CC ‐ control selection
All outcomes

Unclear risk

Randomly selected from a vaccination registry

CC ‐ comparability
All outcomes

Low risk

Possible confounders have been considered for analysis.

CC ‐ exposure
All outcomes

Unclear risk

Phone interview

Summary assessment

Unclear risk

Unclear risk of bias

bb Siscovick 2000

Methods

Study assessing the association between influenza vaccination the previous year and the risk of primary (i.e. occurring in people with no previous history of cardiac disease) cardiac arrest. Case‐control study on 360 cases and 418 controls

Participants

Cases: people who had experienced primary cardiac arrest, aged between 25 and 74 years.
Controls: healthy people selected randomly from the community, who were matched to the cases for age and sex.

Interventions

Immunisation with influenza vaccine, assessed by means of questionnaires

Outcomes

Cardiac arrest

Notes

The authors concluded that vaccination is protective against primary cardiac arrest (OR 0.51, 95% CI 0.33 to 0.79). The difficulty of case ascertainment (77% of potential cases had no medical report and/or autopsy) and recall bias (spouses provided exposure data for 304 cases, while 56 survivor cases provided data jointly with their spouses) make the conclusions of this study unreliable. The reliability of this study is unclear due to a lack of detail on the circulation of influenza in the study areas in the 12 months preceding cardiac arrest (the causal hypothesis is based on the effects of influenza infection on the oxygen supply to the myocardium through lung infection and inflammation).

Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Cases of out‐of‐hospital PCA attended by paramedics in King County, Washington, from October 1988 to July 1994 were identified from paramedic incident reports. Primary cardiac arrest cases were defined by the occurrence of a sudden pulseless condition and the absence of evidence of a non‐cardiac condition as the cause of cardiac arrest.

CC ‐ control selection
All outcomes

High risk

Selected from the community using random digit dialling

CC ‐ comparability
All outcomes

Unclear risk

For each PCA case, 1 to 2 controls, matched for age (within 7 years) and sex

CC ‐ exposure
All outcomes

Unclear risk

"Data on the participants’ vaccination status were collected from both case and control spouses by using a standardised questionnaire. For each participant, information was collected on whether they had received an influenza vaccination during the previous 12 months and, if so, when the vaccination had been given. We did not collect information on whether they had received influenza vaccination during the years prior to that period."

Summary assessment

High risk

bb Zorzon 2003

Methods

Case‐control study

Participants

Cases (n = 140): people affected by MS as defined by the International Panel on MS Diagnosis.
Controls (n = 131): sex‐ and age‐matched to the cases.

Interventions

Exposure to influenza vaccination (unspecified). Exposure to many other factors was assessed by means of face‐to‐face structured questionnaires. Time of onset after exposure is probably not mentioned in the text.

Outcomes

Multiple sclerosis

Notes

"The study was supported by a grant of the University of Trieste, Italy: MPI 60%, 2001"

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Hospital population

CC ‐ control selection
All outcomes

High risk

Blood donor population

CC ‐ comparability
All outcomes

High risk

Poor matching

CC ‐ exposure
All outcomes

High risk

Interview

Summary assessment

High risk

High risk

cb Bardage 2011

Methods

Large, prospective, cohort study assessing the possible association between monovalent, pandemic, H1N1 flu vaccine Pandemrix (GSK) and neurological and/or autoimmune disease

Participants

The study population comprised 1,945,024 people and corresponds to all people registered in Stockholm County on 1 October 2009 who had lived in the region since 1 January 1998.

Interventions

Monovalent A (H1N1) pandemic vaccine Pandemrix (GlaxoSmithKline, Middlesex, UK) containing adjuvants AS03 and squalene.
H1N1 vaccination campaign was initially targeted at healthcare workers and groups considered to be at high risk of complications from influenza (children with multifunctional disorders; pregnant women; people with chronic heart or lung disease, diabetes mellitus, chronic liver failure, chronic renal failure, or immunosuppression; people with body mass index > 40; people with neuromuscular disease affecting breathing capacity).
For the campaign an apposite register was established (Vaccinera) in which information on the dates of a first and second dose of vaccine, batch number, medical contraindications against vaccination, and chronic conditions defining high‐risk patients were recorded.
The vaccination campaign began on 13 October 2009 with 2 phases. During the first 6 weeks (from 13 October through November 2009), participants with a high‐risk condition were preferentially vaccinated; the vaccination was then offered to the remainder of the population during the second phase (from December 2009 onwards).
In total, 1,024,019 participants received at least 1 vaccine dose (446,770 during phase I, 577,249 during phase II).

Outcomes

Data on vaccination (Vaccinera database) were linked to data on utilisation of inpatient and specialist health care (admissions to hospital and visits to specialist care in the county, dates, diagnoses, responsible medical departments, and length of hospital stay) contained in the common healthcare registers for Stockholm County Council (GVR) from 1 January 1998 to 31 August 2010.

Neurological and autoimmune diagnoses to consider for follow‐up were selected based on indication of the European Medicines Agency and defined by the ICD‐10 classification for hospital admissions and visits to specialist care:

  • Guillain‐Barré syndrome: G61

  • Multiple sclerosis (demyelinating disease): G35 (G36.0 + G37.9)

  • Bell's paralysis: G51

  • Narcolepsy: G47.4

  • Polyneuropathy, unspecified: G62.9

  • An/hypoaesthesia: R20.0 + R20.1

  • Paraesthesia: R20.2

  • Rheumatological disease: M05‐M06 + M08

  • Inflammatory bowel disease (Crohn's disease and ulcerative colitis): K50‐K51

  • Insulin‐dependent diabetes among individuals born in 1990 and later: E10

Entering diagnoses into the county healthcare database is part of the doctor’s routine diagnostic work and therefore depends on patients seeking health care. An active search for adverse events during the study period was not performed.
For each investigated pathology, the prevalent diagnoses were considered (i.e. those registered between 1 January 1998 and 30 September 2009) and the incident diagnoses (i.e. those during or after the pandemic period for unvaccinated people and after a first vaccination for vaccinated people between 1 October 2009 and 31 August 2010).
Since risk groups were prioritised for vaccination, risk estimates analysis data were stratified for the first and second phase of the vaccination campaign (the cut‐off point was 45 days from 1 October 2009), considering vaccination as a time‐varying covariate and also time since first vaccination (6 weeks).

Notes

Preliminary assessment (prevalence in vaccination phase I and II):

All but 1 (narcolepsy) of the investigated neurological and autoimmune disorders were significantly more prevalent in those vaccinated in the early phase of the campaign (first 45 days) than in the unvaccinated cohort. Comparing those vaccinated in the late phase (> 45 days) with the unvaccinated cohort, the prevalence of the investigated diseases was not statistically relevant, except for inflammatory bowel disease (prevalence OR 1.17, 95% CI 1.12 to 1.22), Guillain‐Barré syndrome (OR 0.79, 95% CI 0.67 to 0.95), and type 1 diabetes (OR 0.77, 95% CI 0.64 to 0.92, for those born in 1990 and later).

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

Selected group of users

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Drawn from the same community as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

High risk

Not assessed

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Record linkage

Summary assessment

High risk

“The study is a retrospective datalinkage cohort study, with unclear data quality”

cb Baxter 2012

Methods

Retrospective cohort study in which the incidence of medical attended events (MAEs) that occurred in people immunised with LAIV through several seasons was compared with that observed in 2 matched control groups (unvaccinated and immunised with inactivated vaccine). Data for the LAIV‐exposed population were also analysed with a self controlled case series method.

Participants

Participants were members of the Kaiser Permanente (KP) Health Plans in Northern California, Hawaii, and Colorado. Through KP immunisation registries, approximately 20,000 individuals 18 to 49 years of age who were immunised from the 2003 to 2004 to 2007 to 2008 influenza seasons with LAIV as part of routine clinical practice were identified.

Interventions

Intervention hemi‐cohort: Live attenuated influenza vaccine vaccine provided by MedImmune. Each annual formulation of the vaccines contained the strains recommended for inclusion by the US Public Health Service. Study participants with high‐risk underlying medical conditions such as cancer, organ transplantation, diabetes, endocrine and metabolic disorders, blood disorders, liver disorders, kidney disorders, and cardiopulmonary disorders were identified via automated extraction of healthcare databases and excluded from all analysis cohorts. A total of 21,340 participants 18 to 49 years of age were vaccinated with the Ann Arbor strain LAIV during the 5 study seasons.
Control hemi‐cohort 1: unvaccinated (n = 21,340). Participants were KP members who participated in the health plan during the same month as the reference LAIV recipients; for the unvaccinated population, the effective vaccination date was the date on which the matched LAIV recipient was vaccinated.
Control hemi‐cohort 2: trivalent inactivated vaccine purchased by KP for immunisation practices (n = 18,316). Participants were KP members vaccinated during the same month as the reference LAIV recipient.
Both controls were matched for region (Northern California, Hawaii, Colorado), birth date (within 1 year), sex, and prior healthcare utilisation (≤ 1 or > 1 clinic visits during the 180 days before vaccination) 1:1 to the participants of the intervention hemi‐cohort. For Northern California only, participants were also matched on their specific medical clinic. In the case that a match could not be found within a specific control group, the LAIV recipient was excluded from the cohort comparison.
For self controlled case series analysis, intervals of 3 and 21 days' postvaccination were compared with control intervals from 4 to 42 days' postvaccination (for the 3‐day risk interval) and 22 to 42 days' postvaccination (for a 0 to 21‐day risk interval).

Outcomes

Medical attended adverse events

Based on medical diagnoses found in KP database records and collected from outpatient clinics, emergency departments, and hospital admissions, MAEs occurred in 5 main categories and included events considered to be vaccine associated:

  1. Acute respiratory tract events: acute laryngitis, acute laryngotracheitis, acute respiratory failure, acute tracheitis, acute respiratory distress syndrome, asthma, bronchitis, cough, epiglottitis, influenza, influenza with pneumonia, mastoiditis, otitis media, pharyngitis, pneumococcal pneumonia, pneumonia, pulmonary congestion and hypostasis, shortness of breath, sinusitis, tachypnoea, tonsillitis, urinary tract infection, viral pneumonia. Follow‐up 42 days

  2. Acute gastrointestinal tract events: abdominal pain, acute gastritis, acute gastroenteritis, appendicitis, intestinal obstruction, intussusception, irritable bowel syndrome, mesenteric adenitis, nausea and vomiting, pancreatitis, paralytic ileus, perforation of intestine, peritonitis, persistent vomiting, small bowel obstruction, ulceration of intestine, and volvulus. Follow‐up 42 days

  3. Asthma and wheezing events: asthma/reactive airway disease, wheezing/shortness of breath. Follow‐up 180 days

  4. Systemic bacterial infections events: bacteraemia, bacterial meningitis, intracranial and intraspinal abscess, septicaemia, shock: unspecified, shock: endotoxic, and gram‐negative shock. Follow‐up 42 days

  5. Rare diagnoses: potentially related to wild‐type influenza infection: encephalitis/encephalopathy, Guillain‐Barré syndrome, meningitis, myocarditis, other paralytic syndromes, pericarditis, polymyositis, Reye syndrome, and viral meningitis. Follow‐up 42 days

Severe adverse events
Death, inpatient hospitalisation, persistent or significant disability or incapacity, congenital anomaly/birth defect (in the offspring of a participant), or any life‐threatening event. Follow‐up from 0 to 42 days' postvaccination

Notes

Sources of support: "This study was sponsored by MedImmune, LLC. Authors employed by MedImmune were involved in the study design, analysis, and interpretation of data, and in the preparation of the manuscript. Authors employed by Kaiser Permanente were involved in the study design, collection, analysis, and interpretation of data, and in the preparation of the manuscript. The Kaiser Permanente Vaccine Study Center was paid for their services in data collection and analysis but authors were not compensated for their work on this manuscript"

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Selected group of users

Participants were screened for underlying medical conditions and provided the appropriate vaccine based on the eligibility criteria in each vaccine’s package insert, physician discretion, and patient choice.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

No description of the derivation of the non‐exposed cohort

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Matched but not very relevant:

"TIV‐vaccinated and unvaccinated participants were matched to LAIV recipients on region (Northern California, Hawaii, Colorado), birth date (within one year), sex, and prior healthcare utilization. Prior utilization was calculated based on the number of clinic visits during the 180 days before vaccination and classified as low (≤ 1 visit) and high (> 1 visit) for matching. In Northern California, participants also were matched on their specific medical clinic, of which there were 48"

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Record linkage

Summary assessment

Unclear risk

Unclear

cb Kaplan 1982

Methods

Surveillance population‐based study conducted in the USA during the 1979 to 1980 and 1980 to 1981 influenza seasons. The study tested the association between influenza vaccination and Guillain‐Barré syndrome. Reports from each case were obtained from neurologists. All case reports were included. The follow‐up period was 1 September 1979 to 31 March 1980 and 1 September 1980 to 31 March 1981.

Participants

USA (minus Maryland), adult population, 18 years or older

Interventions

Seasonal parenteral vaccine

Outcomes

Cases of Guillain‐Barré syndrome. Vaccine‐associated cases were defined as those with onset within the 8‐week period after influenza vaccination.

Notes

Vaccination rates in the population were obtained from a national immunisation survey.
Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

High risk

High risk

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

High risk

High risk

PCS/RCS ‐ comparability
All outcomes

High risk

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

High risk

High risk

Summary assessment

High risk

High risk

cb Lasky 1998

Methods

Surveillance, population‐based study conducted in the USA (4 states: Illinois, Maryland, North Carolina, Washington) during the 1992 to 1993 and 1993 to 1994 influenza seasons. Discharge diagnoses databases were used to identify cases. Hospital charts were reviewed to confirm diagnosis. The follow‐up period was 1 September 1992 to 28 February 1993 and 1 September 1993 to 28 February 1994.

Participants

Approximately 21 million people, 18 years or older

Interventions

Seasonal parenteral vaccine

Outcomes

Cases of Guillain‐Barré syndrome. Vaccine‐associated cases were defined a priori as those with onset within the 6‐week period after influenza vaccination.

Notes

Results were stratified by age and adjusted by season and sex. Vaccination rates in population were estimated from a random‐digit dialling telephone survey.

Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

High risk

High risk

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

High risk

High risk

PCS/RCS ‐ comparability
All outcomes

High risk

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

High risk

High risk

Summary assessment

High risk

High risk

cb Moro 2013

Methods

Retrospective cohort study evaluating the association between the administration of monovalent pandemic inactivated vaccine H1N1 and severe adverse events

Participants

Participants were identified within several administrative and medical databases of the Italian region Emilia Romagna (about 4.4 million individuals). By data linkage participants immunised with Focetria in the 2009 to 2010 season (n = 103,642) were identified. From the unvaccinated population (n = 3,967,917) a matched unexposed cohort was selected using a propensity score.

Interventions

Immunisation with MF59‐adjuvanted, monovalent H1N1 vaccine Focetria (Novartis Vaccines and Diagnostics, Siena, Italy)

Outcomes

Guillain‐Barré syndrome, paralytic syndromes, encephalitis and encephalomyelitis, Bell’s palsy, demyelinating disease, convulsion, autoimmune hepatitis, vasculitis, immune thrombocytopenia

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear description of the vaccinated population

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Using administrative databases

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Propensity score

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Blind validation process throughout

Summary assessment

Unclear risk

Unclear

cb O'Flanagan 2014

Methods

Large retrospective, population‐based cohort study assessing the possible association between monovalent, pandemic, H1N1 flu vaccine Pandemrix (GSK) and narcolepsy

Participants

Virtually the whole population of Ireland is included in the study, which consists of 90,280 children and adolescents aged below 20 and 3,325,643 adults.

Interventions

Exposure to Pandemrix between October 2009 and March 2010. Information on vaccination was collected in 1 of 2 databases, depending on where vaccination was administered: vaccinations performed in general practitioner clinics were registered in the primary care reimbursement service (PCRS) database, and those performed in Health Service Executive mass vaccination clinics in the pandemic data management system (PDMS) database. The number of individuals vaccinated with Pandemrix was extracted from these databases by week of vaccination. The number of unvaccinated individuals was computed by subtracting the number of individuals vaccinated with any pandemic vaccine brand from the total number of individuals reported in the 2011 census.

Outcomes

Narcolepsy: cases have to fulfil the definition of levels 1 to 3 from Brighton Collaboration.

Level 1: Excessive daytime sleepiness AND/OR suspected cataplexy AND cerebrospinal fluid hypocretin‐1 deficiency.

Level 2: Excessive daytime sleepiness AND definite cataplexy AND level 1 or 2 Multiple Sleep Latency Test abnormalities (mean sleep latency < 8 minutes for adults and < 12 minutes for children < 16 years AND/OR at least 2 sleep‐onset REM periods).

Level 3: Excessive daytime sleepiness AND level 1 Multiple Sleep Latency Test abnormalities (mean sleep latency < 8 minutes for adults and < 12 minutes for children < 16 years AND at least 2 sleep‐onset REM periods).

Narcolepsy cases were identified by means of active case finding by contacting all sleep clinics, neurologists, paediatricians, GPs, psychiatrists, psychologists, and public health nurses in Ireland.

2 experts (1 adult and 1 paediatric neurologist who were blinded to the vaccination status of the cases) reviewed the clinical history of narcolepsy cases (medical records and clinical charts) to confirm the diagnosis and classify them using the internationally agreed Brighton Collaboration case definition for narcolepsy. Cases were included in the study if:

  • their date of first symptom of narcolepsy recorded in medical files occurred after 1 April 2009 and before 31 December 2010;

  • cases or guardians gave oral informed consent;

  • they were classified as level 1, 2, or 3 as per the Brighton case definition.

Prevalent cases with onset prior to April 2009 were excluded. The date of first contact with health care for narcolepsy symptoms as retrieved from GP notes and clinical records was used to estimate the onset of narcolepsy in primary analysis.

Notes

Funding source ‐ government

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

Virtually the whole Irish population is included.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same source

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Possible confounders have been taken into account.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Clinical information of possible cases were reviewed, and the correspondence to a standard case definition verified.

Summary assessment

Unclear risk

Low risk of bias

cb Persson 2014

Methods

Cohort study. Large prospective, register‐based cohort study assessing the possible association between monovalent, pandemic, H1N1 flu vaccine Pandemrix (GSK) and neurological or autoimmune disease, or both

Participants

The present study represents the extension of the cb Bardage 2011 study to the population of more Swedish regions, namely the healthcare regions of Skåne and Västra Götaland and the counties of Kalmar, Östergötland, Stockholm, Värmland, and Norrbotten. Included are 5,845,039 participants, corresponding to about 61% of the whole Swedish population in 2009.

Interventions

Exposure to Pandemrix between October 2009 and March 2010. Vaccinated participants were registered in vaccination centres and identified by means of a personal identification number (PIN, a 10‐digit number attributed to each newborn in Sweden) and linked to vaccination registries. Vaccination data are linked to the National Population Registry by use of the PIN: all individuals registered as vaccinated (n = 3,347,467) were exposed, whereas all remaining individuals were assumed not to be vaccinated (n = 2,497,572).

Personal identification number was also linked to the following databases to obtain further information about participants:

  • National Patient Register, Prescribed Drug Register, and Cancer Registry (National Board of Health and Welfare), to identify hospitalisations and non‐primary care outpatient visits to identify the outcomes under study;

  • Medical Birth Register (National Board of Health and Welfare), to identify pregnancy status at vaccination;

  • National Cause of Death Register (National Board of Health and Welfare), to define deaths during follow‐up.

Outcomes

Neurological and immuno‐related conditions

Outcomes were selected under consideration of previous influenza safety issues, of the results of the previous study carried out in the Stockholm region only, and identified in the registers by using ICD‐10 codes and data about medical drug prescription. Due to the fact that several of the investigated outcomes could have a slow and insidious onset, “prodromal” conditions were identified by linking information present in the registers (date of visits, drug prescriptions, etc.) considering the 5 years preceding the study. Participants who had diagnosis before study start were excluded from risk assessment.

As done in the previous study, risk estimates were stratified for “early” (vaccinated in the first 45 days from the beginning of the campaign) and “late” vaccination (vaccinated after at least 45 days from the beginning of the campaign), as medically “at risk” participants were considered to be priority group for influenza vaccination.

Stratification considering time since vaccination (within/more than 1 year; within 6 weeks/more than 6 weeks) was also carried out.

Association risk between vaccine exposure and outcomes was calculated by means of Cox regression using vaccination as time‐dependent variable (i.e. individuals contributed to the unexposed person‐time until vaccinated and to the exposed ones thereafter). Hazard risk estimates were adjusted for age (in 5‐year bands), gender and county, education and income, number of hospital admissions and ambulatory care visits, pregnancy status, and presence of diagnoses defined by ICD‐10 code.

Notes

Funding source: government

Vaccination status could not be confirmed for 16% to 22% of the Kalmar, Värmland, and Norrbotten participants (corresponding to roughly 2.3% of the whole vaccinated cohort), because PIN was not available in the database. These participants were considered as unvaccinated.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

The exposed hemi‐cohort consists of all people who received the vaccine within 6 Swedish regions.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same populations as the exposed cohort (all people who did not receive influenza vaccination)

PCS/RCS ‐ comparability
All outcomes

Low risk

Age, gender and county, education and income, number of hospital admissions and ambulatory care visits, pregnancy status, and presence of diagnoses defined by ICD‐10 code.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Medical records

Summary assessment

Low risk

Low risk of bias

cb Ray 2011

Methods

See bb Ray 2011. Study data were analysed using a cohort design.

Participants

Interventions

Outcomes

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

cb Shonberger 1979

Methods

Surveillance, population‐based study conducted in the USA during the 1976 to 1977 influenza season. The study tested the association between influenza vaccination and Guillain‐Barré syndrome. Neurologists were directly contacted; physician and hospital records were reviewed. Suspected cases were reported to the CDC directly by patients or medical personnel and were included only if accepted by a state health department. Follow‐up period was 1 October 1976 to 31 January 1977.

Participants

USA population

Interventions

Monovalent A/New Jersey/76 or bivalent A/New Jersey/76 and A/Victoria/75 parenteral vaccine

Outcomes

Cases of Guillain‐Barré syndrome

Notes

Results were stratified by age group and vaccine type. Vaccination rates in the population were obtained from a national immunisation survey.
Rare events (safety)

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

High risk

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

High risk

PCS/RCS ‐ comparability
All outcomes

Unclear risk

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

High risk

Summary assessment

Unclear risk

High risk

paa Ma 2014

Methods

Controlled clinical trial. The effect of pandemic influenza vaccine administration during pregnancy was assessed by comparing the occurrence and the characteristics of pregnancy outcomes and clinical course between vaccinated and non‐vaccinated women and assessing the effectiveness of vaccine administration in preventing ILI.

Participants

Healthy pregnant women between the age of 18 and 35 (n = 226) recruited in 4 adjacent villages of Xiangshui, Jiangsu Province, China. The pregnancies ranged from 5 weeks’ to 32 weeks’ gestation; 122 women received the H1N1 vaccine, whereas 104 formed the control group and did not receive any vaccination. Pregnant women in the control group had to reside in the same or adjacent village/community and have an age difference of < 3 years compared to the women in the vaccinated group, a gestational age of < 3 weeks, and the same numbers of pregnancies as those in the vaccinated group.

Interventions

Split‐virion nonadjuvanted influenza A(H1N1) vaccine (lot 200909008; Shanghai Institute of Biological Products). Each dose contained 15 µg of H1N1 antigen.

Outcomes

Pregnancy outcomes were recorded by the maternity and child healthcare organisations or midwifery agencies according to routine prenatal and delivery services in the pregnant women’s health records (filling out of a unified form on complications during pregnancy and pregnancy outcomes):

  • Spontaneous abortion

  • Artificial abortion

  • Postnatal death

  • Premature birth

  • Prolonged pregnancy

  • Low birth weight

  • Delivery mode (eutocia or Caesarean delivery)

  • Birth weight (< 3500 g or > 3500 g)

  • Apgar score at 1 min (7 to 8 or > 9)

Effectiveness outcomes

  • Influenza‐like illness was defined according to WHO guidelines, which include documented fever (at least 38.0 °C) and cough or sore throat. Participants were asked to contact the local vaccination site or the Xiangshui County Center for Disease Control and Prevention once influenza‐like symptoms appeared.

Notes

Funding source ‐ government

This study has been registered at ClinicalTrials.gov under registration no. NCT01842997.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described, only stated that participants were “divided” into 2 groups.

Allocation concealment (selection bias)

High risk

Absent

Blinding (performance bias and detection bias)
All outcomes

High risk

Not blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No loss to follow‐up

Summary assessment

High risk

High risk of bias

paa Madhi 2014

Methods

Randomised controlled trial carried out on pregnant women to assess the effectiveness of trivalent inactivated influenza vaccine administration during pregnancy against confirmed influenza in women and their newborn. The study was carried out during 2 subsequent epidemic seasons (2011 and 2012).

Participants

Pregnant women aged between 18 and 38 years and having an estimated gestation between 20 and 36 weeks who tested negative for HIV were recruited at 4 antenatal clinics of Soweto, South Africa, during 2 consecutive epidemic seasons (March to August 2011 and March to July 2012). In all, 2116 women entered the study: 1062 were allocated to receive vaccine, 1054 to placebo. In addition, 1026 infants born from vaccinated mothers and 1023 infants born from placebo recipients were enrolled.

Interventions

Women enrolled in the study were randomised 1:1 using a computer‐generated assignment and a block size of 30 and allocated to 1 of the following treatments:

  • Trivalent inactivated influenza vaccine (Vaxigrip, lot number G05831 in 2011 and H7221‐2 in 2012; Sanofi Pasteur) containing 15 μg each of A/California/7/2009 (A/(H1N1)pdm09), A/Victoria/210/2009 (A/H3N2), and a B/Brisbane/60/2008–like virus (B/Victoria), as recommended by WHO for the Southern Hemisphere in 2011 and 2012

  • Placebo consisting of sterile 0.9% saline solution

Both preparations were administered by study staff in the deltoid muscle in a 0.5 mL dose and were macroscopically indistinguishable.

Outcomes

Cases of ILI were identified through active surveillance. The following criteria were used to identify cases among mother and infants respectively:

  • ILI (mothers): fever ≥ 38 °C on oral measurement or history of chills, rigors, or feeling feverish; AND

    • presence of cough or sore throat or pharyngitis; OR

    • presence of myalgia, arthralgia, or headache; OR

    • presence of dyspnoea, breathing difficulty, or chest pain when breathing.

  • ILI (infants):

    • axillary temperature ≥ 37.8 °C or mother’s perception that the infant was feverish, or both, without evidence of a non‐respiratory localised source, coupled with at least 1 sign or symptom of acute respiratory infection within the past 72 hours; OR

    • at least 2 signs and/or symptoms of acute respiratory illness within the past 72 hours including: respiratory rate of ≥ 60 and ≥ 50 breaths per minute in infant 0 to 2 months and 2 to 6 months of age, respectively; difficulty breathing reported by the mother, cough, wheezing, runny or congested nose, cyanosis or oxygen saturation < 90% on room air, chest wall in‐drawing, grunting on expiration, and pus draining from either ear.

Influenza: women and infants with ILI, as well as those presenting or hospitalised at antenatal clinics for any respiratory illness, who underwent PCR test with a positive result for influenza viruses.

Events occurring within the timespans of 24 weeks' postpartum (for women) and the 24th week of age (for infants) have been considered for analysis.

Local and systemic reactions recorded on diary cards during the first week following immunisation

Notes

Funding source ‐ industry

Supported by grants from the Bill and Melinda Gates Foundation (OPP1002747), the National Institutes of Health, National Center for Advancing Translational Sciences Colorado Clinical and Translational Sciences Institute (UL1 TR000154, for REDCap), the South African Research Chairs Initiative of the Department of Science and Technology and National Research Foundation in Vaccine‐Preventable Diseases, and the Respiratory and Meningeal Pathogens Research Unit of the Medical Research Council.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation lists, in blocks of 30 (15 IIV3, 15 placebo) were generated with assignment of a 4‐digit study number being done in sequence of enrolment (SAS 9.1, SAS Institute Inc., Cary, NC, USA).

Allocation concealment (selection bias)

Low risk

Block size of 30 were allocated consecutively to the enrolling sites, after which the randomisation forms for that block were provided to the site in sealed, consecutively numbered envelopes with the pre‐printed study number and the alphabetical code for vaccine or placebo in the envelope.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Both administered preparations were macroscopically indistinguishable.

With the exception of the statistician and the pharmacist, study personnel and study participants were unaware of the group assignments.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

None

Summary assessment

Low risk

Low risk of bias

pba Benowitz 2010

Methods

Case‐control study assessing the effectiveness of influenza vaccination of pregnant women in preventing hospitalisation for influenza in their newborns. Study period ranged from October 2000 to April 2009.

Participants

Cases (n = 113): infants below 12 months hospitalised for influenza between October 2000 and April 2009 who tested positive for influenza with direct fluorescent antibody (DFA)

Controls (n = 192): participants hospitalised for influenza during the same time interval as the cases but negative with the DFA test. For each case 1 or 2 controls matched for birth date and date of hospitalisation were randomly selected.

Interventions

Immunisation with influenza vaccine during pregnancy (until 14 days before delivery)

Outcomes

DFA‐confirmed influenza

Notes

This study was supported by the National Center for Research Resources, a component of the National Institutes of Health.

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Infants hospitalised with DFA positive

CC ‐ control selection
All outcomes

Low risk

Infant hospitalised with DFA negative

CC ‐ comparability
All outcomes

Low risk

Matching

CC ‐ exposure
All outcomes

Unclear risk

Structured interview

Summary assessment

Unclear risk

Unclear

pba Poehling 2011

Methods

Case‐control study assessing the effectiveness of influenza vaccine administered during pregnancy in preventing influenza in newborns under 6 months

Participants

Children (n = 1510) aged below 6 months who were hospitalised for fever or acute respiratory illness, or both during 7 consecutive epidemic seasons (between 2002 and 2003 and 2008 and 2009). Those with positive laboratory confirmation of influenza were enrolled as cases (n = 151); those whose result was negative were enrolled as controls (n = 1359).

Interventions

Influenza vaccination during pregnancy

Outcomes

Influenza

Notes

This project was supported the Centers for Disease Control and Prevention, National Institute of Allergy and Infectious Diseases, and Wachovia Research Fund. 3 authors had received past funding from industry (of these 1 was on the MedImmune Advisory Board and another was a NexBio consultant).

Funding source ‐ mixed

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Laboratory confirmed

CC ‐ control selection
All outcomes

Low risk

Infants without laboratory‐confirmed influenza

CC ‐ comparability
All outcomes

Unclear risk

No matching, unclear information

CC ‐ exposure
All outcomes

Unclear risk

Structured interview

Summary assessment

Unclear risk

Unclear

pbb Irving 2013

Methods

Case‐control study investigating the association between influenza immunisation during pregnancy and spontaneous abortion

Participants

Cases (n = 243) were identified from among the members of 6 Vaccine Safety Datalink organisations. Diagnoses of spontaneous abortion (ICD‐9 code 634) and unspecified abortion (ICD‐9 codes 637) assigned during the 2005 to 2006 and 2006 to 2007 seasons were reviewed and different diagnoses excluded.

Controls (n = 243) were selected from among women who had confirmed intrauterine pregnancy and delivery after the 20th gestational week by frequency‐matching of last menstrual period (within 2 weeks) and healthcare organisation.

Interventions

Immunisation with influenza vaccine. Participants were considered exposed if they were immunised within 28 days before index date. Analysis considering whether vaccine exposure occurred during or before pregnancy was also performed.

Outcomes

Spontaneous abortion cases

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

CC ‐ case selection
All outcomes

Low risk

Consecutive series of cases from electronic databases

CC ‐ control selection
All outcomes

Low risk

From the same population

CC ‐ comparability
All outcomes

Unclear risk

Matched by LMP ‐ confounders

CC ‐ exposure
All outcomes

Unclear risk

Medical record

Summary assessment

Unclear risk

Unclear

pca Ahrens 2014

Methods

Retrospective cohort study. This study was performed by retrospective analysis of data within the Birth Defects Study, an ongoing case‐control study investigating the occurrence of neonatal malformations conducted by the Slone Epidemiology Center at Boston University. It includes hospitals serving the areas surrounding Philadelphia and San Diego, Rhode Island, southern New Hampshire, and parts of New York State and Massachusetts. Occurrence of preterm delivery and low birth weight were compared between non‐malformed (controls) infants born from vaccinated and unvaccinated mothers.

Participants

For the study seasons included in this analysis (from 2006–07 to 2009‐10), mothers of live‐born, non‐malformed infants were identified; 1619 were included in the study.

Interventions

Immunisation with trivalent inactivated vaccine during pregnancy. Within 6 months after delivery, a study nurse conducted a phone interview asking for information about immunisation (and other issues).

Women reporting influenza vaccination during pregnancy were asked to provide a release to allow study staff to obtain their vaccination records, but only 60% of the women complied with this request.

Reports of seasonal trivalent influenza vaccination were categorised according to the timing of receipt: any time during pregnancy (last menstruation period to day before delivery), first trimester (last menstruation period through 14 weeks), second trimester (greater than 15 through 28 weeks), and third trimester (greater than 29 weeks to day before delivery). Women who reported vaccination with pandemic H1N1 vaccine were excluded from the analysis. 334 women were immunised for all seasons considered in the study.

Outcomes

Gestational age at delivery and birth weight were obtained by self report from the mother during the interview. Gestational age was determined by calculating the difference between the last menstruation period and the day of delivery. If the self reported last menstruation period date differed by more than 7 days from the last menstruation period date calculated from the reported ultrasound‐determined due date, then the latter last menstruation period date was used to calculate gestational age. If the self reported last menstruation period date differed by 7 days or less from the last menstruation period calculated from the due date, we chose to use the self reported last menstruation period date because it was a date familiar to the mother and raised less confusion during the course of the interview.

  • Small for gestational age, defined as a weight < 10th percentile for gestational age, considering the sex‐specific distribution of birth weights of infants born in the US in 1999–2000

  • Preterm delivery, defined as live birth before 37 weeks' gestation

ILI: for the last season in study (2009‐10), having had ILI symptoms was also ascertained during the interview

Notes

Funding source ‐ government

At the time of manuscript preparation, Katherine Ahrens was a pre‐doctoral Boston University Reproductive, Perinatal and Pediatric Epidemiology trainee supported by the National Institutes of Health (Grant T32 HD052458). Data collection for this project has been funded by the Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services (Contract No. HHSO100201000038C); the Agency for Healthcare Research and Quality (Grant 1R18HS018463‐01); and the National Institutes of Health (Grants 1R01 HD059861 and 2 R01 HD46595). Drs Louik and Mitchell and Mr Kerr receive research support from Novartis Vaccines and Diagnostics (NVD) for an unrelated study of a meningitis vaccine. Dr Mitchell serves as a member of an advisory committee for a pregnancy registry for a multiple sclerosis agent conducted by Biogen Idec and as an unpaid consultant to NVD on matters unrelated to influenza vaccines. Dr Werler has provided consultation for Amgen, Bristol‐Meyers Squibb, and Abbott regarding their pregnancy registries for rheumatoid arthritis drugs. These companies do not manufacture influenza vaccines. Dr Ahrens has no conflicts to disclose.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Description is insufficient. Participants for this study are simply selected from control population of the Birth Defects Study.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Selected from the same population as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Not clear whether all possible confounding factors were considered

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Interview

Summary assessment

Unclear risk

Unclear risk of bias

pca Black 2004

Methods

Retrospective cohort study assessing the effectiveness of flu vaccination for the prevention of ILI or pneumonia in pregnant women and their newborns

Participants

  • All women with live births in Kaiser Permanente Northern California (KPNC) between the November and February of 5 subsequent seasons (1997 to 1998 to 2001 to 2002, n = 49,585), excluding cases lacking birth date information and women who were discharged after the end of the flu season.

  • All live births in KPNC that occurred during the same time periods as for the mothers (n = 48,639), again cases lacking gestational age or gender information and infants discharged after the end of the flu season were excluded.

Interventions

Immunisation with flu vaccine (no details about type and composition). Data about immunisation were obtained from the KPNC database. In all, 3707 out of the 49,585 pregnant women included in the study were vaccinated, whereas this was 3652 out of the 48,639 live births.

Outcomes

  • Hospitalisation for pneumonia or influenza: at least 1 inpatient stay during the same flu season as delivery or birth with a principal (first) diagnosis of either influenza or pneumonia. To identify these outcomes, the following ICD (9th revision) codes were used to identify inpatient cases: influenza 487 and pneumonia 480, 481, 482, 483, 484, 485, and 486.

  • Outpatient visits: at least 1 physician visit during the same flu season as delivery or birth with 1 of the following diagnoses: upper respiratory infection, pharyngitis, otitis media, asthma, bronchial asthma, viral infection, pneumonia, fever, cough or wheezing associated with respiratory illness.

This information was available from the KPNC databases, which include laboratory, hospitalisation, and outpatient utilisation information for their members.

The effect measure (hazard ratio and corresponding 95% confidence interval) was calculated for ILI visits (including and excluding asthma diagnoses) for the mother and hospitalisation for pneumonia or influenza, ILI visits (excluding otitis media), and otitis media visits in newborns.

  • Caesarean section.

  • Preterm delivery (< 37 weeks).

Notes

Government‐funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

From KPNC databases: the influenza vaccination status of women in the cohort was determined through review of the Kaiser Immunization Tracking System database.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

From KPNC databases

PCS/RCS ‐ comparability
All outcomes

High risk

No matching

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

KPNC maintains administrative databases that include laboratory, hospitalisation, and outpatient utilisation information for their members.

Summary assessment

High risk

High

pca Eick 2011

Methods

Prospective cohort study carried out in 6 hospitals located in the Navajo and White Mountain Apache reservation during 3 subsequent epidemic seasons (2002 to 2005)

Participants

Mother‐infant pairs recruited after delivery at Indian Health Service hospitals on the Navajo or White Mountain Apache reservation, either at the hospital or by home visit.

The study was conducted during 3 influenza seasons from November 2002 to September 2005.

The enrolment periods for each year were:

  • 1 December 2002 to 15 March 2003;

  • 1 November 2003 to 8 March 2004;

  • 1 November 2004 to 15 March 2005.

Inclusion was restricted to mothers who delivered a healthy infant at 36 weeks or later gestation during the enrolment periods. Eligible infants were aged 2 weeks or younger at enrolment. Overall, 1169 mother‐infant pairs were enrolled in the study (241 in 2002 to 2003; 574 in 2003 to 2004; and 354 in 2004 to 2005). Of these, 1160 had at least 1 serum sample and were included.

Interventions

Immunisation of the mother with influenza vaccine. Assessed by reviewing of medical record (also in order to obtain information about prenatal visits, illnesses, and birth information, in addition to administration and timing of influenza vaccine) or, if missing, by maternal report at enrolment.

The decision for influenza vaccination was made by the treating clinician and the pregnant woman; personnel had no role in this decision. Altogether 587 children were born from an unvaccinated mother and 573 from a vaccinated mother during the 3 study seasons.

Outcomes

Surveillance for all medically attended illnesses in enrolled infants was conducted at Indian Health Service and nearby private facilities through the influenza season, or until the child reached 6 months of age (whichever came first). It also included review of the clinic, emergency department, and inpatient paediatric ward logs. A nasopharyngeal aspirate specimen for viral culture was obtained from infants with ILI within 72 hours of the medical visit.

  • Medically attended ILI: defined as a medical visit with at least 1 of the following signs or symptoms reported: fever of 38.0 ºC or higher, diarrhoea, or respiratory symptoms (including cough, runny nose, or difficulty breathing).

  • Laboratory‐confirmed influenza: the first ILI episode with either:

    1. isolation of influenza virus from the nasopharyngeal aspirate specimen;

    2. a 4‐fold or greater rise in HI antibody in serum collected at 2 to 3 or 6 months compared with the previous serum specimen, indicating influenza virus infection during the time interval; or

    3. a positive rapid influenza diagnostic test result with a medical diagnosis of influenza.

Notes

Funding/support: "The study was funded by the National Vaccine Program Office, Department of Health and Human Services, the Office of Minority Women’s Health, Centers for Disease Control and Prevention, Aventis‐Pasteur, and Evans‐Powderject."

Funding source ‐ mixed

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

The study was carried out within Indian reservations.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Derived from the same community as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Reported for some parameters only: sex, presence of household smokers, having wood or coal stove in the house (more frequent among vaccinated), presence of other children in day care, infant breast fed (more frequent among vaccinated), gestational age, mean birth weight

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Active surveillance and testing for laboratory confirmation for symptomatic ILI cases

Summary assessment

Unclear risk

Unclear

pca France 2006

Methods

Retrospective cohort study based on Vaccine Safety Datalink, assessing the effect of influenza vaccination of pregnant women in preventing respiratory illness in newborns. 6 epidemic seasons were considered.

Participants

Infants who were born before or during the influenza season at 4 MCOs (Kaiser Permanente Colorado, Denver; Kaiser Permanente Northern California, Oakland; Kaiser Permanente Northwest, Portland, Oregon; and Group Health Cooperative, Seattle, Washington) between 1 October 1995 and 30 September 2001 were eligible for study inclusion.

Mother‐infant pairs were included in the final study population if:

  1. the mothers were aged 18 to 45 years and enrolled in the MCO for longer than 1 year;

  2. the infants’ gestational age was at least 30 weeks at birth;

  3. the infants were continuous MCO members for at least 14 days during the influenza season;

  4. the infants had a least 1 outpatient visit during the first 3 months of life.

Interventions

An infant was considered exposed if the mother was vaccinated against influenza during the pregnancy and there were at least 28 days from the vaccination date of the mother to the birth date of the infant. Infants of mothers vaccinated within 27 days of birth were excluded from the primary analysis.

Outcomes

Medically attended ARI

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

High risk

From MCO databases

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

High risk

From MCO databases

PCS/RCS ‐ comparability
All outcomes

High risk

Poor matching

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Datalink

Summary assessment

High risk

High risk

pca Hulka 1964

Methods

Prospective cohort study assessing the effectiveness of flu vaccination in pregnancy

Participants

Pregnant women (n = 544) recruited from the "hill" district of Pittsburgh

Interventions

  • Polyvalent flu vaccine containing 200 units of A2 antigen

  • Placebo

Two 1 mL doses were administered 1 month apart.

Outcomes

  • Adverse effects following immunisation (pain, malaise)

  • ILI

  • Days in bed

Assessed by means of questionnaires/phone interviews after epidemic

Notes

Effectiveness follow‐up was available for 59% and 100% of participants in the intervention and placebo arm, respectively.

Funding source ‐ mixed

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

High risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

High risk

Unclear

PCS/RCS ‐ comparability
All outcomes

High risk

Unclear ‐ high attrition

PCS/RCS ‐ assessment of outcome
All outcomes

High risk

Interview

Summary assessment

High risk

Unclear

pca Munoz 2005

Methods

Retrospective cohort study based on the electronic database of Kelsey‐Seybold Clinic (KSC), a large multispecialty clinic in the metropolitan area of Houston (USA). For the study 5 subsequent flu seasons were considered, from 1998 to 2003, taking into account the time between 1 July and 30 June of each year. Approximately 25 obstetricians and 60 paediatricians provided medical care in KSC locations, and about 2500 deliveries occurred every year during the time considered for the study.

Participants

Exposed cohort (n = 225): women who were immunised with inactivated influenza vaccine within 6 months before delivery and who had an uncomplicated singleton pregnancy, were healthy, had at least 1 prenatal care visit at KSC, and their offspring had at least 1 clinic visit at KSC in their first year of life.

Comparison (n = 826): for each vaccinated woman, a comparison group was selected by matching (KSC database) 3 to 5 women for maternal age at delivery, month of delivery, and type of insurance (with the exclusion of both Medicaid or self insurance due to small numbers in this clinic population), who had not received influenza vaccine during pregnancy.

Interventions

Influenza vaccines used during the study period were Aventis Pasteur or Wyeth products. For the control group the index date ("pseudo vaccination date") corresponds to the same number of days before delivery as the real vaccination date for a matching vaccinated woman.

Outcomes

Women

  • Acute respiratory illness: cases recorded at any time, during each flu season and during each epidemic peak of that season diagnosed with the following ICD‐9 codes: 079, 460‐466, 470‐478, 480‐487. The peak of influenza activity was the period during which the number of laboratory‐confirmed cases included at least 85% of influenza cases for that season.

  • Serious adverse events: hospitalisation (death, cause for hospitalisation, and permanently disabling conditions were also included) within 42 days from immunisation identified by ICD‐9 codes.

Medical diagnoses occurred between vaccination and delivery with an incidence ≥ 2% among vaccinated women.

Newborns

  • Diagnoses different from a “normal newborn infant” given at discharge and within 2 days from delivery.

  • Reason for at least 3 days hospitalisation within 1 week, between 8 and 180 days, and between 6 months and 1 year after delivery.

  • Diagnoses reported during ambulatory medical visits during the first 6 months of life.

In the last 2 categories URTI and respiratory infections are also included.

Notes

Little information about characteristics and comparability of the exposed and unexposed cohorts. Outcomes used to assess the effectiveness of vaccination are in some way 'surrogate' and include only hospitalisation and ambulatory diagnoses. The first 2 weeks after vaccination should have been excluded from follow‐up for the assessment of effectiveness in mothers.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Women were included in the study sample if they had received inactivated influenza vaccine within 6 months before delivery of an uncomplicated singleton pregnancy and were otherwise healthy, had at least 1 prenatal care visit at KSC, and their offspring had at least 1 clinic visit at KSC in their first year of life.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

A comparison group was selected by matching of maternal age at delivery, month of delivery, and type of insurance (patients with Medicaid or self insurance were excluded due to the small numbers in this clinic population). For each vaccinated woman, 3 to 5 (ratio 1:3.5) matching healthy women who met all the inclusion criteria but who had not received influenza vaccine during pregnancy were selected.

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Matching

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

The potential protective effect of the vaccine was estimated by recording the occurrence of ARIs in vaccinated women from the time of receipt of influenza vaccine to delivery and in unvaccinated women for the equivalent period of time. Specifically, the occurrence of ARIs during the peak of the influenza season was compared between the groups. Diagnostic codes for ARI included 079, 460‐466, 470‐478, 480‐487.

Summary assessment

Unclear risk

Unclear

pca Yamada 2012

Methods

Questionnaire‐based, retrospective cohort study performed at the 121 obstetrical facilities of Hokkaido (Japan)

Participants

All 121 obstetric facilities in Hokkaido were requested to deliver a 12‐item questionnaire to all postpartum women who gave birth between 1 December 2009 and 31 May 2010 during their stay in obstetric facilities. About 1/3 of the women who delivered in Hokkaido during this time answered the questionnaire (n = 7535).

Interventions

Influenza vaccination during pregnancy. Out of the 7535 women who answered the questionnaire, 4921 received pandemic influenza vaccine. Among them, 2212 were also reported to have been vaccinated with seasonal vaccine. A further 270 (considered as unvaccinated) received seasonal vaccine only.

Outcomes

Influenza. Definition was not provided. All information was collected by means of a questionnaire, on which items about admission to the intensive care unit, intubation or ventilation, and diagnosis of influenza encephalopathy were also present.

Notes

Strongly biased

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

High risk

By interview

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

High risk

By interview

PCS/RCS ‐ comparability
All outcomes

High risk

No matching

PCS/RCS ‐ assessment of outcome
All outcomes

High risk

By interview

Summary assessment

High risk

High

pcb Beau 2014

Methods

Retrospective cohort study. Pregnancies ending between 21 October 2009 (the start of the vaccination campaign) and 30 November 2010 and that had started before 31 January 2010 identified using databases EFEMERIS (database including pregnant women) and CNAMTS (vaccination database): 1645 women exposed to A/H1N1 vaccine during pregnancy and 3290 randomly selected who did not receive A/H1N1 (matched for month and year of the start of pregnancy)

Participants

Data about study population come from an extended database (EFEMERIS) collecting and linking information from 4 different sources:

  • CPAM (Caisse Primaire d’Assurance Maladie), the French Health Insurance System of Haute‐Garonne (South West France): it records all the reimbursed drugs prescribed and dispensed to patients under general state coverage (classified according to the World Health Organization’s Anatomical Therapeutic Chemical classification);

  • PMI (Protection Maternelle et Infantile), the Mother and Child Protection Center: it contains data about the health of both mother (maternal characteristics, some pathologies during pregnancy) and child (weight, size, Apgar score, neonatal pathologies, psychomotor development, congenital malformations) collected during the compulsory medical examinations at ages 8 days, 9 months and 2 years;

  • CDA (Centre de Diagnostic Anténatal), the Antenatal Diagnosis Center centralises data corresponding to all the occurrences of major and minor malformations in the maternities of the region where therapeutic termination has been considered (cause and date of termination);

  • PMSI (Programme de médicalisation des systèmes d’information), the French hospital medical information system provides the nature and date of termination (legal termination, stillbirth, and spontaneous abortion) from Toulouse University Hospital Center.

EFEMERIS contains information about 58,171 mother‐outcome pairs with women who delivered in Haute‐Garonne between 1 July 2004 and 31 December 2010. For the present study, only pregnancies ending between 21 October 2009 (the start of the vaccination campaign) and 30 November 2010 (9 months after the vaccination centres had closed) and that had started before 31 January 2010 were considered (n = 12,120).

For each mother‐outcome pair exposed to A/H1N1 vaccine during pregnancy (n = 1645), 2 mother‐outcome pairs were randomly selected from among individuals in the study population who did not receive A/H1N1 vaccine during pregnancy but who were matched for month and year of the start of pregnancy (n = 3290).

Interventions

Exposure to monovalent pandemic H1N1 vaccine during pregnancy. Vaccination centres were required to report A/H1N1 vaccinations to the French National Health Insurance Fund for Salaried Workers (Caisse National de l’Assurance Maladie des Travailleurs Salariés; CNAMTS)

Exposure was considered at any time during pregnancy. For neonatal pathologies, a subset of babies born from mothers who were immunised within the 3rd pregnancy trimester was also considered.

Authors report that 93% of the exposed women received the non‐adjuvanted Panenza (Sanofi Pasteur), but did not provide any further details.

Outcomes

  • All‐cause pregnancy loss: any loss occurring during pregnancy (legal and therapeutic termination, spontaneous abortion, stillbirth, and ectopic pregnancy).

  • Preterm delivery: birth before 259 days of amenorrhoea/37 completed weeks.

  • Small for gestational age: any singleton with a birth weight < 2 standard deviations from the French reference weight mean, adjusted for gestational age and sex.

  • Neonatal pathologies (including respiratory distress, pneumothorax, neonatal jaundice, metabolic disorders, or sepsis): identified from the children’s health certificates established on the 8th day, as recorded by the physician during medical consultation; these records provided little detail about the conditions.

Notes

Funding source ‐ government

“The EFEMERIS database was funded by the Agence Nationale de Sécurité du Médicament et des produits de santé (ANSM), the CNAMTS, the Mutuelle Générale de l’Education Nationale, the Clinical Research Hospital Program (PHRC) and the Unions régionales des Caisses d’Assurance Maladie. Additional funding has been received from the ANSM for the influenza medication study”

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

The whole cohort of pregnant women between October 2009 and November 2010 was considered. Those who were vaccinated were the exposed cohort.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same source as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Low risk

Possible confounding factors have been taken into account in data analysis (adjustment).

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Secure records

Summary assessment

Low risk

Low risk of bias

pcb Cantu 2013

Methods

Retrospective cohort study. Pregnancy‐related outcomes were observed retrospectively among vaccinated and non‐vaccinated women who received prenatal care and delivered within Birmingham healthcare system in order to establish if vaccination could represent a risk factor.

Participants

Women with singleton pregnancy during 2009‐10 pandemic and 2010‐11 season who had prenatal visit between October and end of December of each season at 1 of the 6 prenatal clinics in Birmingham, Alabama (USA) without suspected foetal abnormality. Altogether, 1094 vaccinated and 2010 non‐vaccinated pregnant women were included.

Interventions

Vaccination with a pH1N1 virus containing vaccine (not further specified) at any time during pregnancy in pandemic season 2009/10 and in 2010 to 2011 epidemic. Participants immunised exclusively with TIV in 2009/10 season were excluded from the primary analysis.

Vaccination status was ascertained through perinatal record system and vaccination logs. Women who were immunised outside of healthcare system were included if they were able to provide their vaccination date.

Outcomes

The following outcomes were collected and recorded at the time of care at the centres.

  • Primary composite outcome: includes miscarriage, stillbirth, preterm birth < 37 weeks, and neonatal demise

  • Miscarriage: defined as delivery prior to 20 weeks

  • Preterm birth (< 37 weeks)

  • Birth weight < 2500 g

  • Neonatal demise (20 weeks)

  • Stillbirth: defined as delivery of a non‐viable foetus at or after 20 weeks

  • Pre‐eclampsia

  • Small for gestational age: foetal growth less than the 10th percentile

  • Neonatal intensive care unit admission

  • Length of maternal stay

  • Antiviral (oseltamivir) therapy

Notes

Funding source ‐ government

Results and effect estimates are provided for both seasons pooled.

Study population was limited to women with prenatal visit in the early flu season between 1 October and 31 December each year, when the vast majority of vaccines were given in order to assure that vaccinated and unvaccinated groups had similar exposure periods and avoid potential bias.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

Representative of a pregnant women population belonging to the Birmingham healthcare system

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same population as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Low risk

Possible confounders have been taken into account.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Secure record

Summary assessment

Low risk

Low risk of bias

pcb Chambers 2013

Methods

Prospective cohort study. The study has been carried out within the Organization of Teratology Information Specialists (OTIS) Research Group studies, whose aim is to evaluate the occurrence of pregnancy outcomes following an exposure to a medication or vaccine administered during pregnancy. OTIS services are located in academic institutions or hospitals throughout the US and Canada and provide counselling to about 70,000 callers annually who present with questions about the risks of exposures in pregnancy. Among these women, exposed and not exposed (comparison group) to the agent of interest are included in the study. In the present pH1N1 vaccine study, participants were enrolled between October 2009 and April 2012 and were exposed or not exposed to either the monovalent (2009–10 season) or trivalent (2009–12 seasons) pH1N1 vaccine in 1 of 3 influenza seasons. Information about exposure and outcomes of exposed and unexposed cohort were assessed by means of phone questionnaires.

Participants

841 pregnant women exposed to a pH1N‐containing vaccine

191 not exposed pregnant women

Interventions

Information about vaccine type, date of vaccination, and medical setting where immunisation took place are present in the OTIS questionnaires. Participants were further asked to verify provided information on their vaccination record (when available) or to give permission to contact the participant’s provider to obtain this information. In cases where the vaccine was given in a non‐traditional setting, an attempt was made to determine the specific product used and the date and location of vaccine administration.

The timing of vaccine exposure was divided into 4 categories:

  • the 2 weeks between last menstruation period and date of conception;

  • from conception to 13 weeks’ gestation;

  • > 13 to 26 weeks’ gestation;

  • > 26 weeks’ gestation.

In the 2009–10 season, some women received the 2009–10 seasonal vaccine (not containing the pH1N1 strain) prior to the pH1N1 monovalent vaccine becoming available, and were subsequently vaccinated with the monovalent pH1N1 vaccine. These women were classified as pH1N1 vaccine exposed; however, previous receipt of the non‐pandemic vaccine was considered a covariate.

The comparison group consisted of women who received no influenza vaccine of any type throughout their pregnancy. Vaccines were monovalent pH1N1 (unspecified) in 2009‐10, and pH1N1‐containing TIV in 2010‐11 and 2011‐12.

Outcomes

Outcomes were collected in the OTIS questionnaire (maternal interview) and medical records obtained from obstetrician, paediatrician, and delivery hospital. Ultrasound dating was used to correct gestational weeks as necessary using a standard algorithm, or if the LMP was unknown. The following definitions were used:

  • Spontaneous abortion: defined as spontaneous pregnancy loss at < 20 gestational weeks

  • Preterm delivery: delivery at < 37 completed gestational weeks

  • Small for gestational age: defined as < 10th centile for sex and gestational age in live‐born infants using standard US growth charts for full and preterm infants

  • Still birth

  • Termination

  • Major and minor birth defects

Analysis for the first 3 outcomes was performed considering timing of exposure. Crude and adjusted HH or OR estimate is provided, other than crude data.

Notes

Funding source ‐ government

This project has been funded in whole or in part with Federal funds from the Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, Department of Health and Human Services, under Contract No. HHS0100201000029C and the OTIS Collaborative Research Group.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

Quite a representative sample of pregnant women enrolled in the OTIS registers

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same source as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Possible confounding factors have been taken into account.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Secure records

Summary assessment

Low risk

Low risk of bias

pcb Cleary 2014

Methods

Cohort study. At the time of 2009‐10 pandemic, the monovalent pH1N1 was offered to pregnant women and other at‐risk groups by the Health Service Executive. Pregnancy outcomes were evaluated in women vaccinated during pregnancy and those not vaccinated during pregnancy.

Participants

Women who delivered at the Coombe Women and Infants University Hospital (Dublin) between December 2009 and September 2010 and who reported having been vaccinated (n = 2996).

The control consists of women who delivered during the same time interval at the same hospital but who reported not having received influenza vaccination (n = 3898).

A second historical control group includes all women who delivered during a time interval (December 2008 to September 2009) before the mass vaccination and the main wave of the 2009‐10 pandemic (n = 7044, not considered for the analysis).

Interventions

Vaccine exposure was ascertained by means of the delivery suite admission form, which contained the following questions:

  1. H1N1 vaccine this pregnancy (Y/N)

  2. When given (I; II, III trimester)

  3. Vaccine used? (Celvapan 1 to 2 doses, Pandremix, unknown)

Any exposure at any time during pregnancy is considered for analysis purposes.

Outcomes

Data on maternal characteristics, medical and obstetric history recorded at the antenatal booking interview, and perinatal outcomes recorded in the delivery suite and neonatal intensive care unit were extracted from electronic hospital records.

  • Preterm birth < 37 weeks

  • Spontaneous birth < 37 weeks

  • Very preterm birth < 32 weeks

  • Spontaneous birth < 32 weeks

  • Small for gestational age: birth weight determined to be less than the 10th centile customised for maternal weight, height, gestation, and infant sex, age

  • Apgar score < 3 at 1 min

  • Apgar score < 7 at 5 min

  • Admitted to neonatal unit

  • Congenital anomaly: ascertained from electronic records of any anomalies identified by midwifery or paediatric staff on the delivery suite or from congenital anomaly, body system or discharge diagnoses fields in the neonatal unit electronic discharge records

  • Perinatal death (within 7 days of life)

Notes

Funding source ‐ government

BC was funded by the charity Friends of the Coombe and the School of Pharmacy, Royal College of Surgeons in Ireland.

Exposure: about 56.5% of vaccinated women reported having received Celvapan (not adjuvanted), 23% Pandremix (AS03 adjuvanted), and 20% were unsure about the specific vaccine used.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

All women who delivered at the Coombe Women and Infants University Hospital between December 2009 and September 2010 and received influenza vaccine during pregnancy

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same source as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Low risk

Characteristics that differ significantly between exposed and not exposed group have been taken into account for effect measure calculation.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Hospital records

Summary assessment

Low risk

Low risk of bias

pcb Deinard 1981

Methods

Prospective cohort study assessing the safety of monovalent A/NJ/8/76 vaccine administration during pregnancy

Participants

Pregnant women enrolled at several obstetric clinics (Minneapolis) on the occasion of a prenatal visit (n = 706)

Interventions

Flu vaccine containing A/NewJersey/8/76 (split‐ or whole‐virus formulation) administered during the first, second, or third pregnancy trimester. Vaccine was administered to 189 women, whereas 517 acted as unvaccinated controls.

Outcomes

  • Local and systemic reactions observed and reported after vaccine administration (only the vaccinated assessed by questionnaire).

  • Pregnancy outcomes: maternal mortality, elective abortion, spontaneous abortion, stillbirth, premature live birth.

  • Infant outcomes: deaths, major or minor congenital anomalies, abnormalities during the first 8 days of life.

Notes

This study should have been performed without external/private/industry funding.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

High risk

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

High risk

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

High risk

Summary assessment

High risk

pcb Dodds 2012

Methods

Retrospective cohort study

Participants

Women who delivered a live or stillborn baby (> 500 g, singleton, > 20 weeks' gestation) at the IWK Clinical Centre (Halifax, Nova Scotia) between 1 April 2006 and 31 October 2009. In all, 9781 were included.

Interventions

TIV seasonal vaccine (not otherwise specified). Women delivering at the IWK were asked whether or not they had received the influenza vaccine. This information was then further verified by using a specific database. Out of the 9781 included participants, 1957 received the vaccine.

Outcomes

The information collected on influenza vaccination was linked to data from the Nova Scotia Atlee Perinatal Database (NSAPD, a population‐based database containing detailed clinical and demographic information for all deliveries in the province) to determine the characteristics of the cohort and the neonatal outcomes. The database includes live births and stillbirths born at a gestational age of at least 20 weeks or having a birth weight of at least 500 g, as well as extensive data on demographics, behaviour and lifestyle, labour and birth, and maternal and neonatal diseases and procedures. The following outcomes were considered:

  • Small for gestational age (≤ 10th percentile)

  • Low birth weight (≤ 2500 g)

  • Term low birth weight

  • Preterm birth (< 37 weeks)

Composite outcome

Notes

Funding source ‐ government

This study was funded by grants from the IWK Health Centre and from the Atlee Foundation of the Department of Obstetrics and Gynecology, Dalhousie University.

Data were provided pooled for all 3 seasons and cover the entire years. Data for the time between 1 April 2007 and 31 December 2007 were missed.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

About half of deliveries in the region occur in the study health centre.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

From the same source as exposed

PCS/RCS ‐ comparability
All outcomes

Unclear risk

It seems that not all possible confounding factors have been taken into account for calculation of adjusted estimate (only smoking habits Y/N). Only those modifying point estimate by > 5% were included.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Medical database records

Summary assessment

Unclear risk

Unclear risk of bias

pcb Fell 2012

Methods

Retrospective cohort assessing the safety of pandemic monovalent H1N1 vaccine in pregnant women, using Ontario’s birth record database

Participants

Women with singleton birth in 2009 to 2010 season (n = 55,570)

Interventions

Monovalent pandemic H1N1 influenza vaccine. In all, 23,340 pregnant women were also immunised with seasonal vaccine.

Outcomes

Frequency of neonatal outcomes in newborns:

  • Preterm birth (< 37 weeks or < 32 weeks)

  • Small for gestational age (below 10th or 3rd percentile)

  • 5‐minute Apgar score below 7

  • Foetal death

Notes

"This study was funded by the Canadian Institutes of Health Research (grant 218653)"

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Summary assessment

High risk

pcb Heikkinen 2012

Methods

Prospective cohort study assessing the safety of pandemic MF‐59 adjuvanted influenza vaccine (Focetria) during pregnancy

Participants

Pregnant women recruited in midwife practices and hospitals in the Netherlands (n = 4281), Argentina (n = 239), and Italy (n = 9). Altogether, 4508 pregnant women were included: 2295 were vaccinated and 2213 were not immunised. There were 4522 live births and 18 intrauterine deaths (2310 born from vaccinated and 2213 from unvaccinated mothers). 3 months' follow‐up data were available for 4385 babies.

Interventions

Monovalent pandemic H1N1, MF‐59 adjuvanted flu vaccine Focetria (Novartis Vaccine and Diagnostics, Cambridge, MA, USA). Among the 2295 vaccinated pregnant women, 1724 received 2 doses, 571 received 1 dose.

Outcomes

  • Gestational diabetes

  • Pre‐eclampsia

  • Spontaneous abortion

  • Stillbirth

  • Live birth

  • Low birth weight

  • Preterm birth

  • Neonatal death

  • Congenital malformation

Notes

"This study was supported by Novartis Vaccines and Diagnostics"

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Summary assessment

High risk

Unclear

pcb Håberg 2013

Methods

Cohort study assessing the risk of neonatal death following exposure to pandemic monovalent H1N1 influenza vaccine or influenza virus during pregnancy

Participants

A total of 113,331 pregnant women

Interventions

Immunisation with pandemic monovalent H1N1 adjuvanted influenza vaccine Pandemrix (GSK) or Celvapan (not adjuvanted)

Outcomes

Foetal death

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Data link

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Data link

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Multivariate model

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Data link

Summary assessment

Unclear risk

Unclear risk of bias

pcb Källén 2012

Methods

Retrospective cohort study assessing the effect on newborn outcomes of pandemic squalene adjuvanted H1N1 vaccine

Participants

The total number of vaccinated women was 18,612 having 18,844 infants (vaccination group, pandemic H1N1 Pandemrix). These women were compared with 136,914 women having 138,931 infants who gave birth after September 2009 and before the end of 2010 (non‐vaccinated group) and with 83,298 women having 84,484 infants who gave birth in the year 2009 before October (pre‐vaccination group).

Interventions

Pandemrix (GlaxoSmithKline; Brentford, Middlesex, UK) containing inactivated split influenza virus A/California/07/2009, squalene adjuvant and thiomersal preservative

Outcomes

  • Stillbirth

  • Preterm birth

  • Low birth weight

  • Small for gestational age

  • Congenital malformations

Notes

"No specific funding was obtained for this study"

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Unclear

Summary assessment

High risk

pcb Launay 2012

Methods

Prospective cohort study assessing the effect of immunisation with pandemic monovalent vaccine during pregnancy

Participants

Pregnant women (n = 877) between 12 and 35 weeks of gestation, aged at least 18 years, who were not vaccinated or infected

Interventions

Immunisation with pandemic monovalent influenza vaccine

Outcomes

Delivery before the 37th gestational week, birth weight, death before or during labour

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

Low

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Low

PCS/RCS ‐ comparability
All outcomes

Unclear risk

No information was given about possible confounders

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Unclear

Summary assessment

Unclear risk

Unclear

pcb Lin 2012

Methods

Retrospective cohort study

Participants

A total of 396 pregnant Taiwanese women were included in the study, of which 198 received influenza vaccine during pregnancy

Interventions

Monovalent H1N1 unadjuvanted, inactivated, split‐virus vaccine AdimFlu‐S (Adimmune Corporation; Taichung, Taiwan) containing 15 g of New York Medical College X‐179A reassortant of the A/California/7/2009 (H1N1)‐like strain in 0.5 mL dose

Outcomes

Systemic and local adverse events in vaccinated mothers

In newborns:

  • Hyperbilirubinaemia

  • Contact dermatitis

  • Upper respiratory tract infection

  • Seborrhoeic dermatitis

  • Respiratory distress

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Medical records

Summary assessment

High risk

pcb Louik 2013

Methods

Retrospective cohort study. The effect of immunisation with an influenza vaccine containing pH1N1 during pregnancy on preterm birth was assessed comparing vaccinated and non‐vaccinated women.

Participants

Study population belonged to those enrolled in a large surveillance‐based, case‐control study carried out by the Slone Epidemiology Center at Boston University. Children‐cases with major structural defects were identified from participating hospitals in the areas surrounding Philadelphia and San Diego as well as by means of birth defect registries in New York State and Massachusetts. Controls were normal‐formed infants randomly selected within the same study hospitals. For the purposes of the present study, only mothers of controls (without malformations) who delivered during the 2009‐10 and 2010‐11 seasons are included. Only mothers of singleton, live‐born infants, who were immunised not after the 37th gestation week, were included. Altogether, 951 women were included, 378 of whom received influenza vaccine.

Interventions

Exposure to a pH1N1‐containing vaccine during pregnancy within the seasons 2009‐10 and 2010‐11. Exposure was ascertained by means of a computer‐assisted phone interview administered 6 months after delivery and eventually verified by examining the vaccination records. A woman was considered exposed if she had received a pH1N1‐containing vaccination. Time of exposure was considered within 1st trimester (until 14th gestation week), 2nd trimester (gestation weeks 15 to 28), and 3rd trimester (from week 29 through delivery). Women whose reported time of exposure could not be attributed to 1 of the trimesters were excluded. Not‐exposed participants should have last menstrual date within the range of last menstrual date reported by exposed participants.

Outcomes

Preterm delivery: defined as delivery at gestational age less than 37 weeks

Notes

Funding source ‐ industry

Drs Louik, Chambers, Jones, Schatz, and Mitchell and Mr Kerr receive research support from Novartis Vaccines and Diagnostics (NVD) for an unrelated study of a meningitis vaccine. Dr Mitchell serves as a member of an advisory committee for a pregnancy registry for a multiple sclerosis agent conducted by Biogen‐Idec and as an unpaid consultant to NVD on matters unrelated to influenza vaccines. Drs Chambers and Jones receive support from GlaxoSmithKline Bio for an unrelated study of human papilloma virus vaccine. Drs Chambers and Jones receive support for unrelated research projects from various pharmaceutical companies: Abbott, Amgen, Bristol‐Myers Squibb, GlaxoSmithKline, Parr, Pfizer, Janssen, Roche Genentech, Sanofi Genzyme, Sandoz, and Teva. Dr Schatz has received research support for projects unrelated to the current study from Aerocrine, Genentech, GlaxoSmithKline, MedImmune, and Merck. Dr Schatz is also a research consultant on subjects unrelated to the current study for Amgen, Boston Scientific, and GlaxoSmithKline. Ms Pyo and Dr Ahrens have no conflicts to disclose.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Cohort consists of the control population of case‐control studies in which case population is represented by mothers of children born with major defects.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same source as the exposed one

PCS/RCS ‐ comparability
All outcomes

Low risk

Taken into account

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Secure records

Summary assessment

Unclear risk

Unclear risk of bias

pcb Ludvigsson 2013

Methods

Retrospective cohort study. Pregnancy outcomes were compared between women exposed to Pandemrix (monovalent H1N1 pandemic‐AS03‐adjuvanted influenza vaccine) during pregnancy and non‐exposed women.

Participants

All live‐born single infants in Stockholm County (conceived between February 2009 and January 2010, n = 21,087)

Interventions

Exposure to 1 dose of monovalent pH1N1‐AS03‐adjuvanted influenza vaccine (Pandemrix) at any time during the pregnancy before the 36th week. Records of vaccination are available from Vaccinera database (vaccination campaign was performed between October 2009 and April 2010, thus about 90% of the doses were administered before end of 2009). In total, 13,297 women were vaccinated against H1N1 during pregnancy before the 36th week. The non‐exposed hemi‐cohort consisted of women who did not receive influenza vaccination during pregnancy or who were immunised after the 36th week (n = 7790).

Different times of exposure during pregnancy (1st or 2nd ‐ 3rd trimester) were also considered for analysis.

Outcomes

Data about pregnancy outcomes were available in the Obstetrix database, in which information from the 1st antenatal visits (8 to 12 gestation weeks) until discharge are collected. For each participant a record is available that can be identified by means of a unique identifier (PIN), which permitted the link between Obstetrix data and those recorded in other archives (Vaccinera among others).

  • Birth weight < 2500 g

  • Gestational duration < 37 weeks

  • Small for gestational age

  • Apgar score at 5 min < 7

  • Caesarean section

Notes

Funding source ‐ government

This project was supported by grants from the Swedish Research Council (Medicine), and the Swedish Council for Working Life and Social Research (FAS). JFL was funded by the Swedish Research Council (Medicine), OS was funded by the Swedish Society of Medicine. LR was partially supported by grants from the Compagnia san Paolo/Firms and the Italian Association for Cancer Research.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

The whole birth cohort of infants conceived between February 2009 and January 2010 in Stockholm, Sweden, whose mother received the influenza vaccine during pregnancy

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

The whole birth cohort of infants conceived between February 2009 and January 2010 in Stockholm, Sweden, whose mother did not receive influenza vaccine during pregnancy

PCS/RCS ‐ comparability
All outcomes

Unclear risk

All possible confounders have been taken into account.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Secure records

Summary assessment

Unclear risk

Unclear risk of bias

pcb Nordin 2013

Methods

Retrospective cohort study based on data from Vaccine Safety Datalink

Participants

Pregnant women aged between 14 and 49 years (n = 223,898) identified in the Vaccine Safety Datalink, who were pregnant between 1 June 2002 and 31 July 2009

Interventions

Immunisation with inactivated trivalent influenza vaccine

Outcomes

Demyelinating diseases, neurological events, thrombocytopenia within 42 days after immunisation

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

KP registry

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

KP registry

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Matched analysis

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

KP registry

Summary assessment

Unclear risk

Unclear

pcb Nordin 2014

Methods

Retrospective cohort study. Data from 7 Vaccine Safety Datalink sites

Participants

In all, 57,554 women vaccinated and 57,554 matched women not vaccinated during pregnancy introduced in data analysis.

Interventions

Seasonal trivalent inactivated vaccine. Epidemic seasons 2004‐05, 2005‐06, 2006‐07, 2007‐08, 2008‐09 were considered.

Outcomes

  • Small for gestational age (< 10th or > 5th percentile)

  • Preterm delivery (< 37 weeks or < 34 weeks)

Notes

Founding source ‐ government

Data and estimate are pooled for all seasons. This is part of the population of the pcb Nordin 2013 study.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

KP registry

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

KP registry

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Matched analysis

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

KP registry

Summary assessment

Unclear risk

Unclear risk of bias

pcb Omer 2011

Methods

Retrospective cohort study based on data from the Georgia Pregnancy Risk Assessment Monitoring System (PRAMS)

Participants

In all, 4168 pregnant women were included during 2 consecutive epidemic seasons (2004 to 2005 and 2005 to 2006), of whom 578 received influenza vaccination.

Interventions

Influenza vaccination during pregnancy

Outcomes

Small for gestational age and preterm births. Periods with different viral circulation were considered in the analysis.

Notes

"The study was partially funded through the Emory University, Global Health Institute Faculty of Distinction Fund award (recipient: SBO). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript"

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ assessment of outcome
All outcomes

High risk

Interview

Summary assessment

Unclear risk

Unclear

pcb Oppermann 2012

Methods

Prospective cohort study based on data from the Institute for Clinical Teratology and Drug Risk Assessment in Pregnancy (D) carried out during the 2009 to 2010 pandemic

Participants

Pregnant women who received consultation regarding reproductive safety of medical products, planned pregnancy, and lactation from the Institute for Clinical Teratology and Drug Risk Assessment. Out of the initial population (n = 16,788), 323 participants received influenza vaccine and completed the follow‐up. A randomly selected group of 1329 non‐vaccinated women formed the control group.

Interventions

  • Non‐adjuvanted split‐virion vaccine CSL H1N1 Pandemic Influenza Vaccine (CSL Biotherapies) approved by the responsible national authority (Paul‐Ehrlich‐Institut) in November 2009 exclusively for the vaccination of pregnant women (216/323).

  • MF59‐adjuvanted monovalent H1N1 vaccine (2/32).

  • Pandemrix (GlaxoSmithKline) AS03‐adjuvanted monovalent split‐virion influenza vaccine (90/323).

  • Unknown vaccine (15/323).

Outcomes

Abortion, preterm birth, malformations

Notes

"This study was supported by the German Federal Institute for Vaccines and Biomedicines (Paul‐Ehrlich‐Institut), Langen, Germany"

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Low

Summary assessment

Unclear risk

Unclear

pcb Pasternak 2012

Methods

Retrospective cohort study assessing the safety of pandemic H1N1 vaccination

Participants

Danish women who were pregnant during the time interval between November 2009 and September 2010 (n = 58,585). Of these, 7062 received influenza vaccine.

Interventions

Monovalent, inactivated, AS03‐adjuvanted split‐virion influenza A (H1N1) pdm09 vaccine (Pandemrix, Glaxosmithkline Biologicals)

Outcomes

Abortion cases (retained or spontaneous)

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Summary assessment

Unclear risk

Unclear

pcb Richards 2013

Methods

Retrospective cohort study assessing the effect of pandemic H1N1 immunisation during pregnancy on neonatal outcomes

Participants

Eligible pregnant women were identified by means of electronic medical records from Kaiser Permanente (KP) managed care organisation sites in Georgia and mid‐Atlantic states. A total of 3327 third‐trimester live births to 3236 mothers between 25 May 2009 and 17 April 2010 were included.

Interventions

Immunisation with H1N1 pandemic vaccine

Outcomes

Preterm birth (27 to 36 weeks), low birth weight

Notes

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

KP registry

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

KP registry

PCS/RCS ‐ comparability
All outcomes

High risk

Possible residual confounding

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Low

Summary assessment

Unclear risk

Unclear

pcb Rubinstein 2013

Methods

Cohort study

Participants

Women with live‐born or stillborn infants of at least 22 weeks or weighing at least 500 g between September 2010 and May 2011 in 49 hospitals of the Public Health Sector in major Argentinian cities were invited to participate in the study.

Interventions

MF‐59 adjuvanted pandemic H1N1 vaccine (Focetria). Information about exposure status and vaccination were obtained from documentation and official registry: vaccination chart, hospital vaccine registries, or centralised registry from the Ministry of Health if available.

The non‐exposed group consisted of non‐vaccinated pregnant women. In total, 7293 vaccinated and 23,195 not vaccinated pregnant women were included.

Outcomes

After participants had signed informed consent form, data from the medical record regarding maternal characteristics, evolution of the index pregnancy and delivery, and status of the newborn were extracted. Participants then completed a brief survey to complement the information. Authors collected data on both mothers and newborns up to day 7 after delivery. For cases discharged before day 7, participants were contacted by telephone at day 7 to check their health status. During the data collection period, a supervisory team visited the participating hospitals weekly to collect and check completion of the forms and to check the quality of the information by reviewing the birth record and clinical record. 2 field supervisors reviewed all forms before information was entered into the database. The following outcomes were considered:

  • Preterm delivery (< 37 weeks): defined as a newborn with a gestational age of less than 37 weeks according to physical examination at birth.

  • Low birth weight (< 2500 g): newborn weighing less than 2500 g.

  • Very low birth weight (< 1500 g): newborn weighing less than 1500 g.

  • Congenital malformations: defined as a newborn presenting with alterations in anatomical development occurring during intrauterine life and diagnosed during gestation or by physical examination within the first 7 days after birth.

  • Early neonatal mortality: defined as death of a newborn within 7 days after birth, foetal mortality as intrauterine death of the foetus in a pregnancy over 22 weeks, and perinatal mortality as early neonatal mortality plus foetal mortality.

  • Low Apgar scores at 5 minutes: defined as a newborn presenting with an Apgar score of less than 7 at 5 minutes.

  • Admission to neonatal intensive care: defined as admission of a newborn to the intensive care unit for a period longer than 48 hours.

Notes

Funding source ‐ industry

This study was funded by an independent research grant from Novartis Argentina SA (Internal Protocol No V111_17TP. 2010). The investigators designed and conducted the study; performed the analysis and interpretation of the data; and are responsible for the results, conclusions, and recommendations.

Sensitivity analysis was performed by excluding those participants for whom vaccination records were not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

The study was performed in 49 public hospitals where about 113,000 deliveries occur annually (about 15% of overall annual live births in Argentina). This is a somewhat representative sample of newborn populations.

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

Drawn from the same population as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Low risk

Possible confounding factors have been taken into account.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Hospital medical records

Summary assessment

Low risk

Low risk of bias

pcb Sheffield 2012

Methods

Retrospective cohort study assessing the safety of seasonal influenza vaccination administered during pregnancy, covering 5 subsequent epidemic seasons (from 2003 to 2004 to 2007 to 2008)

Participants

Women who delivered and received prenatal care at the Southwestern Medical Center of University of Texas and Parkland Health & Hospital System, Dallas, Texas. In all, 8690 were vaccinated and 76,153 acted as unvaccinated controls.

Interventions

Seasonal influenza vaccination was offered to pregnant women from October through March in each season.

Outcomes

  • Estimated gestational age

  • Birth weight

  • Major malformations*

  • Stillbirth*

  • NICU admission*

  • Neonatal death

  • Neonatal pneumonia*

  • Hyperbilirubinaemia

*For these outcomes the authors provided effect estimates considering the trimester of administration.

Notes

This study should have been performed without external/private/industry funding.

Government funded

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Unclear

Summary assessment

High risk

pcb Toback 2012

Methods

Retrospective cohort study testing the safety of live attenuated influenza vaccine when administered during pregnancy

Participants

Pregnant women (n = 834,999) identified by means of a safety database (LifeLink Health Plan Claims Database, Norwalk, USA) between October 2003 and September 2009. Of these, 138 received immunisation with live attenuated influenza vaccine during their pregnancy.

Interventions

Live attenuated influenza vaccine

Outcomes

Hospitalisation and emergency department visits within 42 days after immunisation

Notes

"This research was funded by MedImmune, LLC, Gaithersburg, MD. As part of a consulting agreement with RTI Health Solutions, MedImmune provided funding to support protocol development, data collection, analysis, and manuscript development activities associated with this manuscript. Editorial assistance in formatting the manuscript for submission was provided by Sue Myers, MSc, and Gerard P. Johnson, PhD, of Complete Healthcare Communications, Inc. (Chadds Ford, PA) and was funded by MedImmune, LLC"

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Unclear risk

Unclear

PCS/RCS ‐ comparability
All outcomes

High risk

PCS/RCS ‐ assessment of outcome
All outcomes

Unclear risk

Unclear

Summary assessment

High risk

pcb Trotta 2014

Methods

Retrospective cohort study

Participants

Women residing in the Lombardy region, aged at least 12 and up to 55 years (n = 86,171), whose delivery took place between 23 and 45 weeks of gestation between 1 October 2009 and 30 September 2010, in public or private institutions as well as at home, identified through the regional birth registry (stillbirths were included if the gestational age exceeded 180 days)

Interventions

MF‐59 adjuvanted pandemic H1N1 vaccine. 6426 women received the vaccine during pregnancy.

Outcomes

Pregnancy complication

  • pre‐eclampsia/eclampsia, gestational diabetes

  • in‐hospital maternal death (deaths during labour or delivery occurring in a healthcare institution)

  • admission to intensive care unit

  • type of delivery

Perinatal death

  • stillbirth (delivery of a dead foetus after 180 days of amenorrhoea)

  • in‐hospital neonatal death

Neonatal outcomes

  • small for gestational age neonates (< 10th centile)

  • admission to NICU

  • neonatal reanimation

  • composite outcome: presence of any of the following: clinical information/diagnoses: very low 5‐minute Apgar score (≤ 3), acute respiratory distress syndrome, asphyxia, intraventricular haemorrhage, and acute necrotising enterocolitis

  • congenital malformations including: nervous system, eye, ear, face, and neck, congenital heart defects, respiratory, orofacial clefts, digestive system, abdominal wall defects, urinary, genital, limb, others. Diagnosis with a code compatible to to ICD‐9 in either the medical birth registry or the hospital discharge, according to EUROCAT guideline

Notes

Funding source ‐ government

Funding: Only public employees of the national or regional health authorities were involved in conceiving, planning, and conducting the study; no additional funding was received.

Risk of bias

Bias

Authors' judgement

Support for judgement

PCS/RCS ‐ selection exposed cohort
All outcomes

Low risk

All mothers with singleton pregnancy between 1 October 2009 and 30 September 2010 in the Italian region of Lombardia

PCS/RCS ‐ selection non‐exposed cohort
All outcomes

Low risk

From the same population as the exposed cohort

PCS/RCS ‐ comparability
All outcomes

Low risk

Possible confounders have been taken into account.

PCS/RCS ‐ assessment of outcome
All outcomes

Low risk

Hospital records

Summary assessment

Low risk

Low risk of bias

AE = adverse event
ARI = acute respiratory illness
ATP = according to protocol
CCA = chicken erythrocyte agglutination
CCI = culture‐confirmed influenza illness
CCIV = cell culture‐derived inactivated flu vaccine
CDC = Centers for Disease Control and Prevention
CI = confidence interval
DFA = direct fluorescent antibody
ECG = electrocardiogram
FEF = forced expiratory flow
FEV1 = forced expiratory volume in one second
FVC = forced vital capacity
GBS = Guillain‐Barré syndrome
GMT = geometrical mean titre
GP = general practitioner
GSK = GlaxoSmithKline
HA = haemagglutinin
HA0 = full‐length uncleaved haemagglutinin
HI = haemagglutination inhibition
HMO = health maintenance organisation
ICD = International Classification of Diseases
IgA = immunoglobulin A
ILI = influenza‐like illness
ITI = intention‐to‐immunise
ITT = intention‐to‐treat
IM = intramuscular
IN = intranasal
IU = international units
KP = Kaiser Permanente
KSC = Kelsey‐Seybold Clinic
LAIV = live attenuated influenza vaccine
LCI = laboratory‐confirmed influenza
LMP = last menstrual period
MAE = medical attended event
MCO = managed care organisation
MDCK = Madin‐Darby canine kidney cells
MS = multiple sclerosis
NCKPHP = Northern California Kaiser Permanente Health Plan
NICU = neonatal intensive care unit
OMP = outer membrane protein
OR = odds ratio
ORS = oculo‐respiratory syndrome
PCA = primary cardiac arrest
PCR = polymerase chain reaction
PCS/RCS = prospective/retrospective cohort study
pfu = plaque‐forming units
PP = per‐protocol
RCT = randomised controlled trial
rHA0 = recombinant uncleaved haemagglutinin glycoprotein
RhMK = rhesus macaque kidney cells
RT‐PCR = reverse transcription polymerase chain reaction
SAE = serious adverse event
SAS = statistical analysis systems
TIV = trivalent inactivated vaccine
URTI = upper respiratory tract infection
VMCCI = vaccine‐matched, culture‐confirmed influenza
WDL = working days lost
WHO = World Health Organization
WRL = Wellcome Research Laboratories (Beckenham, Kent)

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

ab Wacheck 2010

Experimental vaccine; dose escalation study

ab López‐Macías 2011a

Experimental vaccine; no outcomes of interest

ab López‐Macías 2011b

Experimental vaccine; no outcomes of interest

ab Mallory 2010

No outcomes of interest

ab Plennevaux 2010

No outcomes of interest

ab Precioso 2011

No outcomes of interest

ab Treanor 2010

Experimental vaccine

ab Turley 2011

Experimental vaccine; no outcomes of interest

Al‐Dabbagh 2013

No outcomes of interest, differences in cytokine levels between ORS cases and controls after vaccination

Ambrosch 1976

Data tables and figure missing

Ambrose 2012

No original data

Andersson 2015

Comment on cb Persson 2014 study

Aoki 1986

Randomised controlled trial, single‐blind. Outcomes were clinical cases and adverse effects. Follow‐up data were not reported by arm.

Arnou 2010

Intradermal administration (3 different lots of the same vaccine) versus intramuscular administration. Serologic response and AE at day 21. No adequate placebo/no intervention control

Atmar 1995

No outcomes of interest

Atmar 2011

Absence of an adequate control

Atsmon 2012

Experimental vaccine; no outcomes of interest

Ausseil 1999

No design (average days of sick leave in vaccinated and non‐vaccinated participants during 1996 and 1997 from staff of an international banking institution)

Banzhoff 2001

No design (cohort), no safety outcomes

Baxter 2010

No design: cohort study for effectiveness

Baxter 2011

A 'head‐to‐head' trial: "FluBlok (purified HA proteins manufactured in expresSF+® insect cells under serum free conditions using a baculovirus expression system (BEVS). Uncleaved HA produced by this method is referred to as rHA0. Vaccine formulation consisted of 135g total HA protein (45g each) as determined by single radial immunodiffusion assay (SRID) and included rHA0 derived from the following influenza strains A/Solomon Islands/03/2006 (H1N1), A/Wisconsin/67/2005 (H3N2), and B/Malaysia/2506/2004 VS. The same CDC‐derived vaccine seed viruses were used for the licensed trivalent inactivated vaccine (TIV; Fluzone [2007–2008 formulation; Sanofi Pasteur, Swiftwater, PA), which contained 15g of each HA [45g total])"

Baxter 2012

No design: controlled case series

Baxter 2013

Self controlled time series study

Belongia 2009

Case‐control study, no harm assessment

Belshe 2001

No original data

Benke 2004

Questionnaire survey; non‐comparative analysis

Beran 2013

Absence of an adequate control group (quadrivalent versus trivalent inactivated vaccine; low versus normal adjuvant content)

Betts 1977b

Trial with swine vaccine (Hsw1N1, A/New Jersey/76)

Beyer 1996

Review

Carlson 1979

No adequate control, no outcome of interest

Cate 1977

Trial with swine vaccine (Hsw1N1, A/New Jersey/76)

Chavant 2013

Absence of a control group; study population consists of vaccinated pregnant women only

Chichester 2012

Experimental vaccine; no outcomes of interest

Chlibek 2002

Not a randomised controlled trial

Choe 2011a

No design: cross‐sectional study

Choe 2011b

No design: case series

Choe 2011c

No design: case series

Chou 2007

Case report

Clover 1991

Randomised controlled trial. More than 75% of the study population was out of the age range stated in the protocol.

Confavreux 2001

Participants are MS cases.

Conlin 2013

Inadequate comparison and study design: cohort study with pandemic versus seasonal (not exposed) vaccines in women and newborns

Couch 2012

Experimental vaccine; no outcomes of interest

Das Gupta 2002

Does not contain effectiveness data

Davidson 2011

Inadequate comparison: all enrolled participants received LAIV, then were randomised to either placebo or Lactobacillus rhamnosus GG

Davies 1972

Cohort with efficacy outcomes. Experimental and control group were selected separately.

Davies 1973

Not randomised. Participants volunteered for immunisation, and comparison was made with a randomly selected non‐immunised control group.

De Serres 2003a

No comparison, absence of adequate control group

De Serres 2003b

No control

De Serres 2004

Population at risk of further ORS episodes.

De Wals 2012

No design: self controlled case series for association between H1N1 and GBS

Dolin 1977

Trial with swine vaccine (Hsw1N1, A/New Jersey/76)

Dominguez 2012

No design: case‐control study assessing effectiveness in general population

Duffy 2014

Case‐centred study

Eames 2012

No design: effectiveness cohort study in general population

Edmonson 1970

Influenza B vaccine was used as control.

Eick‐Cost 2012

No design: case‐control study assessing effectiveness in general population

El'shina 1998

Major inconsistencies in the study text

Englund 1993

Inadequate comparison (tetanus toxoid vaccine)

Finklea 1969

Randomised controlled trial, double‐blind. 2 bivalent inactivated influenza vaccines with the same viral composition, differing in purification procedures, were compared.
Outcomes were clinical cases and adverse effects.
Raw data about clinical cases were not reported by arm.
Circulating virus showed significant antigenic differences from the A2 vaccine strain.

Fisher 2012

No outcomes of interest (antibody titres only)

Foy 1981

Absence of adequate control

Frank 1981

No usable safety data (scores)

Freestone 1976

Conference proceedings

Gerstoft 2001

Not a randomised controlled trial

Greenbaum 2002

No outcome of interest

Greene 2013

Case‐centred study

Gross 1999

Outcome measures outside inclusion criteria.

Grotto 1998

Not a randomised controlled trial

Gruber 1994

Randomised controlled trial conducted in the USA on 41 cystic fibrosis (CF) patients and 89 family members, recruited through a clinic. Participants were randomly assigned in a double‐blinded fashion by family to receive either intranasal, live, cold‐adapted influenza A vaccine or the recommended intramuscular trivalent inactivated influenza vaccine.

The study lasted 3 years (from 1989 to 1991). Participants were immunised each fall, staying in the same assigned vaccine group. The live vaccine arm counted 20 CF and 33 family members; the trivalent vaccine arm 21 and 56, respectively.

69 participants (17 CF patients and 52 family members) dropped out. The reasons were stated in the article.

The live vaccine was the same throughout the period: A/Kawasaki/9/86 (H1N1) 107.3 pfu, A/Los Angeles/2/87 107.3 pfu.

The viral strains used in the inactivated vaccines were:

  • 1989 to 1990: A/Taiwan/1/86 (H1N1), A/Shanghai/11/87 (H3N2), B/Yagamata/16/88, 15 mg/dose of each

  • 1990 to 1991: A/Taiwan/1/86 (H1N1), A/Shanghai/16/89 (H3N2), B/Yagamata/16/88, 15 mg/dose of each

  • 1991 to 1992: A/Taiwan/1/86 (H1N1), A/Beijing/353/89 (H3N2), B/Panama/45/90, 15 mg/dose of each

Live vaccine recipients also received monovalent inactivated influenza B vaccine (identical to that contained in the trivalent vaccine) as an intramuscular placebo. Allantoic fluid was the placebo for aerosol administration.

Data were extracted and loaded for family members only.

Outcomes were clinical and laboratory‐confirmed cases, working days lost, admissions, deaths, and adverse effects.

Clinical cases were classified as "respiratory illness" or "febrile respiratory illness". Laboratory‐confirmed cases were defined by an influenza virus isolation from a throat swab.

Adverse effects were defined as temperature > 38 °C, rhinorrhoea, sore throat, cough, increasing sputum, redness, swelling, chills. Results are expressed as % of participant‐days with symptoms.

Participants were followed throughout the period. Owing to the dropouts, the vaccinated were counted as participant‐years: 54 in the live vaccine arm; 56 in the trivalent vaccine arm.

The influenza illness surveillance period for study participants was defined as the interval from the date of the first influenza isolate from the population under routine surveillance to 2 weeks after the last isolate for each year.

Viral strains circulating during the outbreaks were:

  • 1989 to 1990: A/Shanghai/11/87 (H3N2)

  • 1990 to 1991: A/Beijing/353/89 (H3N2), B/Panama/45/90‐like

  • 1991 to 1992: A/Beijing/353/89 (H3N2)

We excluded this trial because it was not placebo controlled, and the authors did not specify if the strains used to develop cold‐adapted and inactivated vaccines were antigenically comparable or not.

Gwini 2011

No design: self controlled case series

Haber 2004

Analysis of temporal trends of GBS 1990 to 2003, comparison with temporal trends of non‐GBS adverse event reports from the Vaccine Adverse Event Reporting System (VAERS)

Haigh 1973

Not randomised: all the volunteers were immunised on a single day, and the intention to allocate participants randomly was not strictly adhered to

Halperin 2002

Outcome measures outside inclusion criteria.

Hambidge 2011

Participants affected by sickle cell crisis.

Heinonen 1973

Control consists of another vaccine.

Hellenbrand 2012

No design: case‐control study assessing effectiveness in general population

Hobson 1970

Polyvalent influenza vaccine was used as control.

Hobson 1973

Randomised controlled trial. Clinical outcomes were side effects only.

Hoskins 1973

Influenza B vaccine was used as control.

Hoskins 1976

Not placebo or 'do nothing' controlled

Hoskins 1979

No control group

Howell 1967

Not prospective: appears to be an historical cohort

Huang 2011

Comparison is not adequate (vaccine versus vaccine).

Hurwitz 1983

Report of GBS surveillance 1978 to 1979, non‐comparative study

Jackson 2011

No adequate control (the same vaccine prepared with different antigenic concentrations was administered to each group)

Janjua 2012

No design: case‐control study assessing effectiveness in general population

Jianping 1999

Not a randomised controlled trial

Jimenez‐Jorge 2012

No design: case‐control study assessing effectiveness in general population

Keitel 2001

Efficacy outcome measures outside inclusion criteria. The safety data are presented in a non‐analysable way.

Kelly 2012

No design: case‐control study assessing effectiveness in general population

Khazeni 2009

Review and cost‐effectiveness analysis

Kiderman 2001

Tables and text show inconsistencies that do not allow data extraction.

Kim 2012

Surveillance for adverse events

Kissling 2012

No design: case‐control study assessing effectiveness in general population

Kunz 1977

No adequate control

Langley 2004

Review

Lavallee 2014

Review about stroke and vaccination in elderly people

Lee 2011

No design: self controlled case series

Leeb 2011

No design: case series

Leroux‐Roels 2010a

Absence of an adequate control, serological outcomes only

Leroux‐Roels 2010b

Absence of an adequate control, serological outcomes only

Liem 1973

Reported the results of 9 placebo‐controlled clinical trials and 2 field studies, involving a total of about 10,000 participants, carried out in several countries to assess the efficacy of killed influenza spray vaccines. Studies were conducted during the years 1969 to 1971.
Allocation of the participants to the arms of the trials was done according to a predetermined randomisation scheme. 8 of the studies were double‐blind. The field studies were not randomised. The attack rate for influenza among the population study was very low, and in 2 of the trials the vaccination procedure started too late, when the outbreak was ongoing. The attack rates, based exclusively on the serologically confirmed cases, are only reported by a graph and deriving the crude data is impossible.

Lind 2014

Surrogate exposure assessment (antibody level)

Liu 2012

Study to identify variables associated with uptake of influenza vaccination during pregnancy

Louik 2013

Methods for assessing flu vaccine exposure during pregnancy

Mackenzie 1975

No design: allocation is arbitrary, and groups with different characteristics were formed

Mackenzie 2012

Non‐comparative design

Mair 1974

Influenza B vaccine was used as control.

Maynard 1968

Influenza B vaccine was used as control.

McCarthy 2004

Review

Mendelman 2001

Does not report original results

Merelli 2000

Review

Meyers 2003a

Review

Meyers 2003b

Review

Micheletti 2011

Total number of AEs observed after administration of each vaccine type

Monto 2000

Not a randomised controlled trial

Montplaisir 2014

Study population outside age range.

Moro 2011

Non‐comparative study

Morris 1975

Design is unclear: no standard random allocation. Only 25 out of 30 participants seem to have been immunised, but in the method description 30 were considered for exposure to natural influenza A/Scotland/840/74. 1 of these was excluded prior due to tonsillitis.

Mostow 1977

Outcomes were safety only. Absence of adequate control

Muennig 2001

Not a randomised controlled trial

Murray 1979

Not adequate comparison (pregnant versus non‐pregnant women)

Nazareth 2013

Absence of control group, non‐comparative

Nichol 1996

Same data as Nichol 1995 (included)

Nichol 1999b

Review

Nichol 2001

Not a randomised controlled trial

Nichol 2003

Contains data from previous studies

Nichol 2004

Re‐analysis of Nichol 1999 (included)

Omon 2011

Non‐comparative study

Petrie 2011

No new data: reports data from already published and included studies (aa Ohmit 2006, aa Ohmit 2008, aa Monto 2009)

Phillips 2013

Absence of adequate control group

Phonrat 2013

No outcomes of interest

Pleguezuelos 2012

Experimental vaccine; no outcomes of interest

Puig‐Barbera 2012

No design: case‐control study assessing effectiveness in general population (also children and elderly)

Puleston 2010

Not outcomes of interest

Pyhala 2001

Not a randomised controlled trial

Reynales 2012

Safety survey after Celtura (H1N1) administration. Absence of control group

Rimmelzwaan 2000

Outcome measures outside inclusion criteria.

Rocchi 1979c

Very poor reporting, unclear definition, no description of methods

Rowhani‐Rahbar 2012

Participants are children

Ruben 1972

Absence of adequate control

Ruben 1973

Both arms contained the same vaccine strains.

Safranek 1991

Reassessment of Schonberger 1979 (included)

Sarateanu 1980

Absence of adequate control

Scheifele 2013

No outcomes of interest

Schonberger 1981

Review of the evidence of the aetiology of GBS, no original data presented

Schwartz 1996

Report about Nichol 1995 (included)

Simpson 2012

No design: cohort and case‐control study assessing effectiveness in general population

Sipilä 2015

Ecological study

Skowronski 2002

Non‐comparative (survey)

Skowronski 2003

Population at risk of further ORS episodes

Smith 1977a

Reports a small part of the Hoskins trial. It compared illness occurring among a group of vaccinated boys against non‐vaccinated controls that had no part in the trial.

Smith 1977b

Trial with swine vaccine (Hsw1N1, A/New Jersey/76)

Song 2011

1 trial is a 'head‐to‐head' trial (Gc501 versus Fluarix) with serological outcomes only; the other trial (safety) has no control.

Souayah 2011

Compares the incidence of GBS cases after tetravalent human papillomavirus vaccine with that observed after pneumococcal and flu vaccine administration

Spencer 1975

Authors did not report crude data on the clinical outcomes.

Spencer 1979

Reporting does not make clear the methods used to allocate participants and to conceal allocation. Clinical outcome data are not reported.

Steinhoff 2012

Inadequate control (23v pneumococcal vaccine administered to the control group). Re‐analysis of Zaman 2008 data (excluded)

Sumaya 1979

No outcomes of interest

Talaat 2010

Data on AEs are not provided in a useful form (bar graphs or cumulatively in the text).

Tavares 2011

Non‐comparative

Taylor 1969

No outcomes of interest, rhinovirus vaccine as control

Taylor 2012

Experimental vaccine; no outcomes of interest

Thompson 2014

Test‐positive case‐control study

Tokars 2012

No design: controlled case series

Treanor 2001

Outcome measures outside inclusion criteria.

Treanor 2002

Outcome measures outside inclusion criteria.

Treanor 2012

No design: case‐control study

Tsai 2010

Non‐comparative

Tsatsaris 2011

Same vaccine administered in different pregnancy weeks (inadequate comparison).

Tyrrell 1970

We were unable to include the 3 studies reported in this paper for the following reasons.

  1. No design, no comparison, no outcomes.

  2. Probable controlled clinical trial, but participants' ages likely out of range (schools).

  3. No design, even if an unvaccinated control group for school 3 and for the employees of the Imperial Chemical Industries is present.

Vesikari 2012

Safety data after dose I (seasonal versus placebo) are not extracted (bar graph).

Warren‐Gash 2013

Outside target age; all participants were older than 60 years

Warshauer 1976

Not randomised. Data reporting was not complete.

Wilde 1999

Pneumococcal vaccine was used as control.

Williams 1973

No placebo or 'do nothing' control

Williams 2011

No design: case series

Wise 2012

No design

Wood 1999

Not a randomised controlled trial

Wood 2000

Not a randomised controlled trial

Xu 2012

No original data presented

Yang 2012

No safety data

Yeager 1999

Non‐comparative study: absence of a control arm

Yih 2012

No design: controlled case series

Zaman 2008

Inadequate control (23v pneumococcal vaccine administered to the control group)

AE = adverse event
GBS = Guillain‐Barré syndrome
LAIV = live attenuated influenza vaccine
MS = multiple sclerosis
ORS = oculo‐respiratory syndrome
pfu = plaque‐forming units

Data and analyses

Open in table viewer
Comparison 1. Inactivated parenteral influenza vaccine versus placebo or 'do nothing'

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza Show forest plot

25

71221

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.36, 0.47]

Analysis 1.1

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

1.1 WHO recommended ‐ matching vaccine

15

46444

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.34, 0.49]

1.2 WHO recommended ‐ vaccine matching absent or unknown

7

15068

Risk Ratio (M‐H, Random, 95% CI)

0.45 [0.34, 0.59]

1.3 Monovalent not WHO recommended ‐ vaccine matching

2

9675

Risk Ratio (M‐H, Random, 95% CI)

0.22 [0.10, 0.52]

1.4 Monovalent not WHO recommended ‐ vaccine matching ‐ high dose

1

34

Risk Ratio (M‐H, Random, 95% CI)

0.11 [0.00, 2.49]

2 Influenza‐like illness Show forest plot

16

25795

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.75, 0.95]

Analysis 1.2

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.

2.1 WHO recommended ‐ matching vaccine

7

4760

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.77, 0.91]

2.2 WHO recommended ‐ vaccine matching absent or unknown

7

20942

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.69, 1.18]

2.3 Monovalent not WHO recommended ‐ vaccine matching

1

59

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.28, 3.70]

2.4 Monovalent not WHO recommended ‐ vaccine matching ‐ high dose

1

34

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.09, 2.30]

3 Physician visits Show forest plot

2

2308

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.40, 1.89]

Analysis 1.3

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 3 Physician visits.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 3 Physician visits.

3.1 WHO recommended ‐ matching vaccine

1

1178

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.37, 0.91]

3.2 WHO recommended ‐ vaccine matching absent or unknown

1

1130

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.90, 1.83]

4 Days ill Show forest plot

3

3133

Mean Difference (IV, Random, 95% CI)

‐0.21 [‐0.98, 0.56]

Analysis 1.4

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 4 Days ill.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 4 Days ill.

4.1 WHO recommended ‐ matching vaccine

2

2003

Mean Difference (IV, Random, 95% CI)

‐0.58 [‐0.85, ‐0.32]

4.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

0.66 [0.16, 1.16]

5 Times any drugs were prescribed Show forest plot

2

2308

Mean Difference (IV, Random, 95% CI)

‐0.01 [‐0.03, 0.01]

Analysis 1.5

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 5 Times any drugs were prescribed.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 5 Times any drugs were prescribed.

5.1 WHO recommended ‐ matching vaccine

1

1178

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.04, ‐0.00]

5.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.00, 0.00]

6 Times antibiotic was prescribed Show forest plot

2

2308

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.03, ‐0.01]

Analysis 1.6

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 6 Times antibiotic was prescribed.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 6 Times antibiotic was prescribed.

6.1 WHO recommended ‐ matching vaccine

1

1178

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.03, ‐0.01]

6.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

‐0.01 [‐0.03, 0.01]

7 Working days lost Show forest plot

4

3726

Mean Difference (IV, Random, 95% CI)

‐0.04 [‐0.14, 0.06]

Analysis 1.7

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 7 Working days lost.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 7 Working days lost.

7.1 WHO recommended ‐ matching vaccine

3

2596

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐0.19, 0.02]

7.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

0.09 [0.00, 0.18]

8 Hospitalisations Show forest plot

3

11924

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.85, 1.08]

Analysis 1.8

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 8 Hospitalisations.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 8 Hospitalisations.

8.1 WHO recommended ‐ matching vaccine

1

1178

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

8.2 WHO recommended ‐ vaccine matching absent or unknown

1

1130

Risk Ratio (M‐H, Random, 95% CI)

2.89 [0.12, 70.68]

8.3 Monovalent not WHO recommended ‐ vaccine matching

1

9616

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.85, 1.08]

9 Clinical cases (clinically defined without clear definition) Show forest plot

3

4259

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.72, 1.05]

Analysis 1.9

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 9 Clinical cases (clinically defined without clear definition).

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 9 Clinical cases (clinically defined without clear definition).

9.1 WHO recommended ‐ matching vaccine

2

2056

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.64, 1.25]

9.2 WHO recommended ‐ vaccine matching absent or unknown

1

2203

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.69, 0.99]

10 Local harms Show forest plot

20

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 10 Local harms.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 10 Local harms.

10.1 Local ‐ tenderness/soreness

20

35655

Risk Ratio (M‐H, Random, 95% CI)

3.13 [2.44, 4.02]

10.2 Local ‐ erythema

9

29499

Risk Ratio (M‐H, Random, 95% CI)

2.59 [1.77, 3.78]

10.3 Local ‐ induration

3

7786

Risk Ratio (M‐H, Random, 95% CI)

4.28 [1.25, 14.67]

10.4 Local ‐ arm stiffness

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.54, 4.83]

10.5 Local ‐ combined endpoint (any or highest symptom)

11

12307

Risk Ratio (M‐H, Random, 95% CI)

2.44 [1.82, 3.28]

11 Systemic harms Show forest plot

17

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.11

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 11 Systemic harms.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 11 Systemic harms.

11.1 Systemic ‐ myalgia

11

35008

Risk Ratio (M‐H, Random, 95% CI)

1.74 [1.41, 2.14]

11.2 Systemic ‐ fever

13

23850

Risk Ratio (M‐H, Random, 95% CI)

1.55 [1.26, 1.91]

11.3 Systemic ‐ headache

14

35999

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.99, 1.30]

11.4 Systemic ‐ fatigue or indisposition

12

35788

Risk Ratio (M‐H, Random, 95% CI)

1.19 [1.05, 1.36]

11.5 Systemic ‐ nausea/vomiting

4

6315

Risk Ratio (M‐H, Random, 95% CI)

1.80 [0.65, 5.04]

11.6 Systemic ‐ malaise

3

26111

Risk Ratio (M‐H, Random, 95% CI)

1.51 [1.18, 1.92]

11.7 Systemic ‐ combined endpoint (any or highest symptom)

6

2128

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.87, 1.53]

Open in table viewer
Comparison 2. Live aerosol influenza vaccine versus placebo or 'do nothing'

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza Show forest plot

9

11579

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.35, 0.62]

Analysis 2.1

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

1.1 WHO recommended ‐ matching vaccine

4

6584

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.37, 0.82]

1.2 WHO recommended ‐ vaccine matching absent or unknown

3

4568

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.27, 0.68]

1.3 Non WHO recommended ‐ vaccine matching absent or unknown

2

427

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.08, 0.56]

2 Influenza‐like illness Show forest plot

6

12688

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.84, 0.96]

Analysis 2.2

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.

2.1 WHO recommended ‐ matching vaccine

2

4254

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.76, 1.12]

2.2 WHO recommended ‐ vaccine matching absent or unknown

3

8150

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.82, 0.97]

2.3 Non WHO recommended ‐ vaccine matching absent or unknown

1

284

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.73, 1.16]

3 Influenza cases (clinically defined without clear definition) Show forest plot

3

23900

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.71, 1.11]

Analysis 2.3

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Influenza cases (clinically defined without clear definition).

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Influenza cases (clinically defined without clear definition).

3.1 WHO recommended ‐ matching vaccine

1

1931

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.49, 0.80]

3.2 WHO recommended ‐ vaccine matching absent or unknown

1

2082

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.88, 1.25]

3.3 Non WHO recommended ‐ vaccine matching absent or unknown

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.92, 1.05]

4 Local harms Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 4 Local harms.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 4 Local harms.

4.1 Local ‐ upper respiratory infection symptoms

6

496

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.22, 2.27]

4.2 Local ‐ cough

6

2401

Risk Ratio (M‐H, Random, 95% CI)

1.51 [1.08, 2.10]

4.3 Local ‐ coryza

2

4782

Risk Ratio (M‐H, Random, 95% CI)

1.56 [1.26, 1.94]

4.4 Local ‐ sore throat

7

6940

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.49, 1.86]

4.5 Local ‐ hoarseness

1

306

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.51, 2.83]

4.6 Local ‐ combined endpoint (any or highest symptom)

3

4921

Risk Ratio (M‐H, Random, 95% CI)

1.56 [1.31, 1.87]

5 Systemic harms Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 5 Systemic harms.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 5 Systemic harms.

5.1 Systemic ‐ myalgia

4

1318

Risk Ratio (M‐H, Random, 95% CI)

2.47 [1.26, 4.85]

5.2 Systemic ‐ fever

4

1318

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.54, 1.92]

5.3 Systemic ‐ fatigue or indisposition

3

1018

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.93, 2.07]

5.4 Systemic ‐ headache

2

975

Risk Ratio (M‐H, Random, 95% CI)

1.54 [1.09, 2.18]

5.5 Systemic ‐ combined endpoint (any or highest symptom)

5

1018

Risk Ratio (M‐H, Random, 95% CI)

1.40 [0.82, 2.38]

Open in table viewer
Comparison 3. Inactivated aerosol influenza vaccine versus placebo or 'do nothing'

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza Show forest plot

1

1348

Odds Ratio (M‐H, Random, 95% CI)

0.38 [0.14, 1.02]

Analysis 3.1

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

1.1 WHO recommended ‐ vaccine matching absent or unknown

1

1348

Odds Ratio (M‐H, Random, 95% CI)

0.38 [0.14, 1.02]

1.2 WHO recommended ‐ matching vaccine

0

0

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Local harms Show forest plot

3

1578

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.71, 1.27]

Analysis 3.2

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Local harms.

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Local harms.

2.1 Local ‐ sore throat

3

1500

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.54, 1.33]

2.2 Local ‐ combined endpoint (any or highest symptom)

1

78

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.71, 1.48]

3 Systemic harms Show forest plot

3

1880

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.71, 1.62]

Analysis 3.3

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Systemic harms.

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Systemic harms.

3.1 Systemic ‐ myalgia

2

151

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.36, 2.25]

3.2 Systemic ‐ fatigue or indisposition

2

151

Risk Ratio (M‐H, Random, 95% CI)

1.40 [0.52, 3.75]

3.3 Systemic ‐ headache

2

151

Risk Ratio (M‐H, Random, 95% CI)

1.52 [0.85, 2.72]

3.4 Systemic ‐ fever

1

1349

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.03, 7.80]

3.5 Systemic ‐ combined endpoint (any or highest symptom)

1

78

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.12, 1.04]

Open in table viewer
Comparison 4. Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza in mothers Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.1

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 1 Influenza in mothers.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 1 Influenza in mothers.

1.1 TIV containing pH1N1

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Influenza‐like illness in mothers Show forest plot

2

2342

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.20, 1.95]

Analysis 4.2

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 2 Influenza‐like illness in mothers.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 2 Influenza‐like illness in mothers.

2.1 TIV containing pH1N1

1

2116

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.79, 1.16]

2.2 Monovalent pH1N1

1

226

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.08, 1.02]

3 Influenza in newborn Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.3

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 3 Influenza in newborn.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 3 Influenza in newborn.

3.1 TIV containing pH1N1

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Influenza‐like illness in newborn Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.4

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 4 Influenza‐like illness in newborn.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 4 Influenza‐like illness in newborn.

4.1 TIV containing pH1N1

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 5. Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal inactivated vaccine effectiveness in mothers ‐ pregnant women Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.1

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 1 Seasonal inactivated vaccine effectiveness in mothers ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 1 Seasonal inactivated vaccine effectiveness in mothers ‐ pregnant women.

1.1 H1N1 ‐ vaccine ‐ effectiveness ILI (unadjusted data)

1

7328

Risk Ratio (M‐H, Random, 95% CI)

0.11 [0.06, 0.21]

1.2 Seasonal ‐ vaccine ‐ effectiveness ILI ‐ (unadjusted data)

3

50507

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.24, 1.18]

2 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women Show forest plot

2

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 5.2

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 2 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 2 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.

2.1 Seasonal vaccine effectiveness ILI (HR adjusted data)

2

Hazard Ratio (Random, 95% CI)

0.96 [0.90, 1.03]

3 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women Show forest plot

1

Risk Ratio (Random, 95% CI)

Subtotals only

Analysis 5.3

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 3 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 3 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.

3.1 Seasonal vaccine effectiveness ILI (RR adjusted data)

1

Risk Ratio (Random, 95% CI)

0.92 [0.73, 1.16]

3.2 Seasonal vaccine efficacy influenza ‐ laboratory‐confirmed

1

Risk Ratio (Random, 95% CI)

0.59 [0.37, 0.94]

4 H1N1 vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women Show forest plot

15

Odds Ratio (Random, 95% CI)

Subtotals only

Analysis 5.4

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 4 H1N1 vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 4 H1N1 vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.

4.1 Abortion (OR adjusted data)

5

Odds Ratio (Random, 95% CI)

0.75 [0.62, 0.90]

4.2 Abortion (HR adjusted data)

3

Odds Ratio (Random, 95% CI)

0.81 [0.63, 1.04]

4.3 Congenital malformation (OR adjusted data)

6

Odds Ratio (Random, 95% CI)

1.11 [0.99, 1.23]

4.4 Prematurity (< 37 weeks) (OR unadjusted data)

11

Odds Ratio (Random, 95% CI)

0.76 [0.67, 0.85]

4.5 Prematurity (< 37 weeks) (OR adjusted data)

7

Odds Ratio (Random, 95% CI)

0.84 [0.76, 0.93]

4.6 Prematurity (< 37 weeks) (HR adjusted data)

2

Odds Ratio (Random, 95% CI)

1.11 [0.46, 2.68]

4.7 Prematurity (< 37 weeks) vaccination in I trimester OR adjusted data

2

Odds Ratio (Random, 95% CI)

1.08 [0.92, 1.28]

4.8 Prematurity (< 37 weeks) vaccination in II/III trimester OR adjusted data

2

Odds Ratio (Random, 95% CI)

0.96 [0.87, 1.06]

4.9 Neonatal death (OR adjusted data)

2

Odds Ratio (Random, 95% CI)

1.09 [0.40, 2.95]

5 Seasonal vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women Show forest plot

7

Odds Ratio (Random, 95% CI)

Subtotals only

Analysis 5.5

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 5 Seasonal vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 5 Seasonal vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.

5.1 Abortion (OR unadjusted data)

1

Odds Ratio (Random, 95% CI)

0.60 [0.41, 0.86]

5.2 Congenital malformation (OR unadjusted data)

2

Odds Ratio (Random, 95% CI)

0.55 [0.08, 3.73]

5.3 Prematurity (OR unadjusted data)

6

Odds Ratio (Random, 95% CI)

0.95 [0.82, 1.10]

5.4 Prematurity (OR adjusted data)

2

Odds Ratio (Random, 95% CI)

0.93 [0.82, 1.06]

5.5 Neonatal death (OR unadjusted data)

1

Odds Ratio (Random, 95% CI)

0.55 [0.35, 0.88]

6 Seasonal vaccine containing H1N1 Show forest plot

2

Risk Ratio (Random, 95% CI)

Subtotals only

Analysis 5.6

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 6 Seasonal vaccine containing H1N1.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 6 Seasonal vaccine containing H1N1.

6.1 Prematurity (37 weeks) vaccination in I trimester HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.63 [0.76, 3.47]

6.2 Prematurity (< 37 weeks) vaccination in II trimester HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.48 [0.21, 10.64]

6.3 Prematurity (< 37 weeks) vaccination in III trimester HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.37 [0.44, 4.25]

6.4 Prematurity (< 37 weeks) vaccination at any time during pregnancy HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.75 [0.57, 5.44]

Open in table viewer
Comparison 6. Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Effectiveness in newborns ‐ pregnant women (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

0.24 [0.04, 1.40]

Analysis 6.1

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 1 Effectiveness in newborns ‐ pregnant women (adjusted data).

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 1 Effectiveness in newborns ‐ pregnant women (adjusted data).

1.1 Seasonal vaccine ‐ effectiveness ‐ ILI ‐ pregnant women

2

Odds Ratio (Random, 95% CI)

0.24 [0.04, 1.40]

2 Seasonal vaccine safety ‐ pregnancy‐related outcomes (adjusted data) Show forest plot

1

Odds Ratio (Random, 95% CI)

0.80 [0.36, 1.78]

Analysis 6.2

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 2 Seasonal vaccine safety ‐ pregnancy‐related outcomes (adjusted data).

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 2 Seasonal vaccine safety ‐ pregnancy‐related outcomes (adjusted data).

2.1 Abortion

1

Odds Ratio (Random, 95% CI)

0.80 [0.36, 1.78]

Open in table viewer
Comparison 7. Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal influenza vaccination and Guillain‐Barré syndrome Show forest plot

3

Risk Ratio (Random, 95% CI)

1.28 [0.85, 1.93]

Analysis 7.1

Comparison 7 Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies, Outcome 1 Seasonal influenza vaccination and Guillain‐Barré syndrome.

Comparison 7 Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies, Outcome 1 Seasonal influenza vaccination and Guillain‐Barré syndrome.

1.1 General population (adjusted data)

2

Risk Ratio (Random, 95% CI)

1.29 [0.83, 2.02]

1.2 Pregnant women (unadjusted data)

1

Risk Ratio (Random, 95% CI)

0.65 [0.03, 15.95]

Open in table viewer
Comparison 8. Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 2009 to 2010 A/H1N1 ‐ general population (unadjusted data) Show forest plot

6

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.1

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 1 2009 to 2010 A/H1N1 ‐ general population (unadjusted data).

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 1 2009 to 2010 A/H1N1 ‐ general population (unadjusted data).

1.1 < 7 weeks

6

1528

Odds Ratio (M‐H, Random, 95% CI)

2.22 [1.14, 4.31]

1.2 At any time

6

1656

Odds Ratio (M‐H, Random, 95% CI)

1.69 [0.87, 3.29]

2 2009 to 2010 A/H1N1 ‐ general population (adjusted data) Show forest plot

4

Odds Ratio (Random, 95% CI)

0.83 [0.39, 1.75]

Analysis 8.2

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 2 2009 to 2010 A/H1N1 ‐ general population (adjusted data).

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 2 2009 to 2010 A/H1N1 ‐ general population (adjusted data).

2.1 < 7 weeks

4

Odds Ratio (Random, 95% CI)

0.92 [0.35, 2.40]

2.2 > 6 weeks (i.e. at any time)

3

Odds Ratio (Random, 95% CI)

0.71 [0.22, 2.32]

3 Seasonal influenza vaccination general population (adjusted data) Show forest plot

1

Odds Ratio (Random, 95% CI)

1.38 [0.18, 10.43]

Analysis 8.3

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 3 Seasonal influenza vaccination general population (adjusted data).

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 3 Seasonal influenza vaccination general population (adjusted data).

Open in table viewer
Comparison 9. Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza vaccination (seasonal) ‐ demyelinating diseases (unadjusted data) Show forest plot

1

223898

Odds Ratio (M‐H, Random, 95% CI)

0.16 [0.02, 1.25]

Analysis 9.1

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 1 Influenza vaccination (seasonal) ‐ demyelinating diseases (unadjusted data).

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 1 Influenza vaccination (seasonal) ‐ demyelinating diseases (unadjusted data).

1.1 General population

0

0

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Pregnant women

1

223898

Odds Ratio (M‐H, Random, 95% CI)

0.16 [0.02, 1.25]

2 Influenza vaccination (H1N1) ‐ demyelinating diseases (unadjusted) Show forest plot

1

144252

Risk Ratio (M‐H, Random, 95% CI)

2.06 [0.51, 8.22]

Analysis 9.2

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 2 Influenza vaccination (H1N1) ‐ demyelinating diseases (unadjusted).

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 2 Influenza vaccination (H1N1) ‐ demyelinating diseases (unadjusted).

Open in table viewer
Comparison 10. Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza vaccination (seasonal) ‐ general population ‐ demyelinating diseases (unadjusted data) Show forest plot

4

8009

Odds Ratio (M‐H, Random, 95% CI)

0.96 [0.79, 1.17]

Analysis 10.1

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 1 Influenza vaccination (seasonal) ‐ general population ‐ demyelinating diseases (unadjusted data).

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 1 Influenza vaccination (seasonal) ‐ general population ‐ demyelinating diseases (unadjusted data).

2 Influenza vaccination (seasonal) ‐ general population ‐ multiple sclerosis (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

0.76 [0.54, 1.08]

Analysis 10.2

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 2 Influenza vaccination (seasonal) ‐ general population ‐ multiple sclerosis (adjusted data).

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 2 Influenza vaccination (seasonal) ‐ general population ‐ multiple sclerosis (adjusted data).

3 Influenza vaccination (seasonal) ‐ general population ‐ optic neuritis (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

1.03 [0.82, 1.30]

Analysis 10.3

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 3 Influenza vaccination (seasonal) ‐ general population ‐ optic neuritis (adjusted data).

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 3 Influenza vaccination (seasonal) ‐ general population ‐ optic neuritis (adjusted data).

Open in table viewer
Comparison 11. Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal influenza vaccine ‐ HR (adjusted data) Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 11.1

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 1 Seasonal influenza vaccine ‐ HR (adjusted data).

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 1 Seasonal influenza vaccine ‐ HR (adjusted data).

1.1 General population

0

Hazard Ratio (Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Pregnant women

1

Hazard Ratio (Random, 95% CI)

0.90 [0.68, 1.19]

2 Seasonal influenza vaccine (unadjusted data) Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 11.2

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 2 Seasonal influenza vaccine (unadjusted data).

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 2 Seasonal influenza vaccine (unadjusted data).

2.1 General population

0

0

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Pregnant women

1

223898

Odds Ratio (M‐H, Random, 95% CI)

0.92 [0.70, 1.20]

Open in table viewer
Comparison 12. Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal influenza vaccine ‐ general population (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

Subtotals only

Analysis 12.1

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 1 Seasonal influenza vaccine ‐ general population (adjusted data).

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 1 Seasonal influenza vaccine ‐ general population (adjusted data).

1.1 < 2 months

2

Odds Ratio (Random, 95% CI)

1.87 [0.43, 8.06]

1.2 < 6 months

1

Odds Ratio (Random, 95% CI)

0.90 [0.55, 1.47]

1.3 < 12 months

1

Odds Ratio (Random, 95% CI)

0.70 [0.47, 1.04]

2 Seasonal influenza vaccine ‐ general population (unadjusted data) Show forest plot

2

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 12.2

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 2 Seasonal influenza vaccine ‐ general population (unadjusted data).

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 2 Seasonal influenza vaccine ‐ general population (unadjusted data).

2.1 < 2 months

2

1926

Odds Ratio (M‐H, Random, 95% CI)

1.72 [0.48, 6.15]

2.2 < 6 months

1

1065

Odds Ratio (M‐H, Random, 95% CI)

0.92 [0.59, 1.43]

2.3 < 12 months

1

1066

Odds Ratio (M‐H, Random, 95% CI)

0.72 [0.50, 1.05]

Open in table viewer
Comparison 13. 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

3

3065

Risk Ratio (M‐H, Random, 95% CI)

0.71 [0.57, 0.88]

Analysis 13.1

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

1.1 Standard recommended parenteral ‐ non‐matching ‐ 1 dose

3

2715

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.57, 0.95]

1.2 Standard recommended parenteral ‐ non‐matching ‐ 2 doses

1

350

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.44, 0.98]

2 Influenza Show forest plot

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.87]

Analysis 13.2

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.

2.1 Standard recommended parenteral ‐ non‐matching

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.87]

3 Hospitalisations Show forest plot

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.41, 1.68]

Analysis 13.3

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.

3.1 Standard recommended parenteral ‐ non‐matching

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.41, 1.68]

4 Pneumonia Show forest plot

1

2072

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.14, 7.17]

Analysis 13.4

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.

4.1 Standard recommended parenteral ‐ non‐matching

1

2072

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.14, 7.17]

Open in table viewer
Comparison 14. 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

4

4580

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.25, 0.48]

Analysis 14.1

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

1.1 WHO recommended parenteral ‐ matching vaccine ‐ 1 dose

4

4226

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.23, 0.53]

1.2 WHO recommended parenteral ‐ matching vaccine ‐ 2 doses

1

354

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.22, 0.57]

2 Influenza Show forest plot

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.07 [0.02, 0.31]

Analysis 14.2

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.

2.1 WHO recommended parenteral ‐ matching vaccine

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.07 [0.02, 0.31]

3 Hospitalisations Show forest plot

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.13, 0.94]

Analysis 14.3

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.

3.1 WHO recommended parenteral ‐ matching vaccine

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.13, 0.94]

4 Pneumonia Show forest plot

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.05, 6.51]

Analysis 14.4

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.

4.1 WHO recommended parenteral ‐ matching vaccine

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.05, 6.51]

5 Working days lost Show forest plot

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

Analysis 14.5

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 5 Working days lost.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 5 Working days lost.

5.1 WHO recommended parenteral ‐ matching vaccine

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

6 Days ill Show forest plot

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

Analysis 14.6

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 6 Days ill.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 6 Days ill.

6.1 WHO recommended ‐ matching vaccine

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

Open in table viewer
Comparison 15. 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

2

1000

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.46, 0.95]

Analysis 15.1

Comparison 15 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 15 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

1.1 Inactivated polyvalent aerosol vaccine versus placebo ‐ non‐matching ‐ 1 dose

2

644

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.32, 1.27]

1.2 Inactivated polyvalent aerosol vaccine versus placebo ‐ non‐matching ‐ 2 doses

1

356

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.44, 0.97]

Open in table viewer
Comparison 16. 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

2

1009

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.32, 0.91]

Analysis 16.1

Comparison 16 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 16 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

1.1 Inactivated monovalent aerosol vaccine versus placebo ‐ matching ‐ 1 dose

2

650

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.17, 1.41]

1.2 Inactivated monovalent aerosol vaccine versus placebo ‐ matching ‐ 2 doses

1

359

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.38, 0.86]

Open in table viewer
Comparison 17. 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza cases (clinically defined without clear definition) Show forest plot

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.92, 1.05]

Analysis 17.1

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 1 Influenza cases (clinically defined without clear definition).

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 1 Influenza cases (clinically defined without clear definition).

1.1 Non‐matching

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.92, 1.05]

2 Complications (bronchitis, otitis, pneumonia) Show forest plot

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.03, 2.24]

Analysis 17.2

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 2 Complications (bronchitis, otitis, pneumonia).

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 2 Complications (bronchitis, otitis, pneumonia).

2.1 Non‐matching

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.03, 2.24]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.
Figuras y tablas -
Analysis 1.1

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.
Figuras y tablas -
Analysis 1.2

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 3 Physician visits.
Figuras y tablas -
Analysis 1.3

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 3 Physician visits.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 4 Days ill.
Figuras y tablas -
Analysis 1.4

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 4 Days ill.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 5 Times any drugs were prescribed.
Figuras y tablas -
Analysis 1.5

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 5 Times any drugs were prescribed.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 6 Times antibiotic was prescribed.
Figuras y tablas -
Analysis 1.6

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 6 Times antibiotic was prescribed.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 7 Working days lost.
Figuras y tablas -
Analysis 1.7

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 7 Working days lost.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 8 Hospitalisations.
Figuras y tablas -
Analysis 1.8

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 8 Hospitalisations.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 9 Clinical cases (clinically defined without clear definition).
Figuras y tablas -
Analysis 1.9

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 9 Clinical cases (clinically defined without clear definition).

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 10 Local harms.
Figuras y tablas -
Analysis 1.10

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 10 Local harms.

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 11 Systemic harms.
Figuras y tablas -
Analysis 1.11

Comparison 1 Inactivated parenteral influenza vaccine versus placebo or 'do nothing', Outcome 11 Systemic harms.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.
Figuras y tablas -
Analysis 2.1

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.
Figuras y tablas -
Analysis 2.2

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Influenza‐like illness.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Influenza cases (clinically defined without clear definition).
Figuras y tablas -
Analysis 2.3

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Influenza cases (clinically defined without clear definition).

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 4 Local harms.
Figuras y tablas -
Analysis 2.4

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 4 Local harms.

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 5 Systemic harms.
Figuras y tablas -
Analysis 2.5

Comparison 2 Live aerosol influenza vaccine versus placebo or 'do nothing', Outcome 5 Systemic harms.

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.
Figuras y tablas -
Analysis 3.1

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 1 Influenza.

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Local harms.
Figuras y tablas -
Analysis 3.2

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 2 Local harms.

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Systemic harms.
Figuras y tablas -
Analysis 3.3

Comparison 3 Inactivated aerosol influenza vaccine versus placebo or 'do nothing', Outcome 3 Systemic harms.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 1 Influenza in mothers.
Figuras y tablas -
Analysis 4.1

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 1 Influenza in mothers.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 2 Influenza‐like illness in mothers.
Figuras y tablas -
Analysis 4.2

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 2 Influenza‐like illness in mothers.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 3 Influenza in newborn.
Figuras y tablas -
Analysis 4.3

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 3 Influenza in newborn.

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 4 Influenza‐like illness in newborn.
Figuras y tablas -
Analysis 4.4

Comparison 4 Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy, Outcome 4 Influenza‐like illness in newborn.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 1 Seasonal inactivated vaccine effectiveness in mothers ‐ pregnant women.
Figuras y tablas -
Analysis 5.1

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 1 Seasonal inactivated vaccine effectiveness in mothers ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 2 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.
Figuras y tablas -
Analysis 5.2

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 2 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 3 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.
Figuras y tablas -
Analysis 5.3

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 3 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 4 H1N1 vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.
Figuras y tablas -
Analysis 5.4

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 4 H1N1 vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 5 Seasonal vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.
Figuras y tablas -
Analysis 5.5

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 5 Seasonal vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women.

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 6 Seasonal vaccine containing H1N1.
Figuras y tablas -
Analysis 5.6

Comparison 5 Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies, Outcome 6 Seasonal vaccine containing H1N1.

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 1 Effectiveness in newborns ‐ pregnant women (adjusted data).
Figuras y tablas -
Analysis 6.1

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 1 Effectiveness in newborns ‐ pregnant women (adjusted data).

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 2 Seasonal vaccine safety ‐ pregnancy‐related outcomes (adjusted data).
Figuras y tablas -
Analysis 6.2

Comparison 6 Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies, Outcome 2 Seasonal vaccine safety ‐ pregnancy‐related outcomes (adjusted data).

Comparison 7 Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies, Outcome 1 Seasonal influenza vaccination and Guillain‐Barré syndrome.
Figuras y tablas -
Analysis 7.1

Comparison 7 Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies, Outcome 1 Seasonal influenza vaccination and Guillain‐Barré syndrome.

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 1 2009 to 2010 A/H1N1 ‐ general population (unadjusted data).
Figuras y tablas -
Analysis 8.1

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 1 2009 to 2010 A/H1N1 ‐ general population (unadjusted data).

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 2 2009 to 2010 A/H1N1 ‐ general population (adjusted data).
Figuras y tablas -
Analysis 8.2

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 2 2009 to 2010 A/H1N1 ‐ general population (adjusted data).

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 3 Seasonal influenza vaccination general population (adjusted data).
Figuras y tablas -
Analysis 8.3

Comparison 8 Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies, Outcome 3 Seasonal influenza vaccination general population (adjusted data).

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 1 Influenza vaccination (seasonal) ‐ demyelinating diseases (unadjusted data).
Figuras y tablas -
Analysis 9.1

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 1 Influenza vaccination (seasonal) ‐ demyelinating diseases (unadjusted data).

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 2 Influenza vaccination (H1N1) ‐ demyelinating diseases (unadjusted).
Figuras y tablas -
Analysis 9.2

Comparison 9 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies, Outcome 2 Influenza vaccination (H1N1) ‐ demyelinating diseases (unadjusted).

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 1 Influenza vaccination (seasonal) ‐ general population ‐ demyelinating diseases (unadjusted data).
Figuras y tablas -
Analysis 10.1

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 1 Influenza vaccination (seasonal) ‐ general population ‐ demyelinating diseases (unadjusted data).

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 2 Influenza vaccination (seasonal) ‐ general population ‐ multiple sclerosis (adjusted data).
Figuras y tablas -
Analysis 10.2

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 2 Influenza vaccination (seasonal) ‐ general population ‐ multiple sclerosis (adjusted data).

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 3 Influenza vaccination (seasonal) ‐ general population ‐ optic neuritis (adjusted data).
Figuras y tablas -
Analysis 10.3

Comparison 10 Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies, Outcome 3 Influenza vaccination (seasonal) ‐ general population ‐ optic neuritis (adjusted data).

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 1 Seasonal influenza vaccine ‐ HR (adjusted data).
Figuras y tablas -
Analysis 11.1

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 1 Seasonal influenza vaccine ‐ HR (adjusted data).

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 2 Seasonal influenza vaccine (unadjusted data).
Figuras y tablas -
Analysis 11.2

Comparison 11 Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies, Outcome 2 Seasonal influenza vaccine (unadjusted data).

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 1 Seasonal influenza vaccine ‐ general population (adjusted data).
Figuras y tablas -
Analysis 12.1

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 1 Seasonal influenza vaccine ‐ general population (adjusted data).

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 2 Seasonal influenza vaccine ‐ general population (unadjusted data).
Figuras y tablas -
Analysis 12.2

Comparison 12 Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies, Outcome 2 Seasonal influenza vaccine ‐ general population (unadjusted data).

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.
Figuras y tablas -
Analysis 13.1

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.
Figuras y tablas -
Analysis 13.2

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.
Figuras y tablas -
Analysis 13.3

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.
Figuras y tablas -
Analysis 13.4

Comparison 13 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.
Figuras y tablas -
Analysis 14.1

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.
Figuras y tablas -
Analysis 14.2

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 2 Influenza.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.
Figuras y tablas -
Analysis 14.3

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 3 Hospitalisations.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.
Figuras y tablas -
Analysis 14.4

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 4 Pneumonia.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 5 Working days lost.
Figuras y tablas -
Analysis 14.5

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 5 Working days lost.

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 6 Days ill.
Figuras y tablas -
Analysis 14.6

Comparison 14 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo, Outcome 6 Days ill.

Comparison 15 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.
Figuras y tablas -
Analysis 15.1

Comparison 15 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 16 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.
Figuras y tablas -
Analysis 16.1

Comparison 16 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo, Outcome 1 Influenza‐like illness.

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 1 Influenza cases (clinically defined without clear definition).
Figuras y tablas -
Analysis 17.1

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 1 Influenza cases (clinically defined without clear definition).

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 2 Complications (bronchitis, otitis, pneumonia).
Figuras y tablas -
Analysis 17.2

Comparison 17 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo, Outcome 2 Complications (bronchitis, otitis, pneumonia).

Summary of findings for the main comparison. Inactivated parenteral influenza vaccine compared to placebo or 'do nothing' for preventing influenza in healthy adults

Inactivated parenteral influenza vaccine compared to placebo or 'do nothing' for preventing influenza in healthy adults

Patient or population: healthy adults
Setting: community‐based studies in North America, South America, and Europe (1969 to 2009)
Intervention: inactivated parenteral influenza vaccine
Comparison: placebo or 'do nothing'

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo or 'do nothing'

Risk with inactivated parenteral influenza vaccine

Influenza
assessed by laboratory confirmation

Timing of assessment: most studies tested vaccines over a single influenza season

Study population1

RR 0.41
(0.36 to 0.47)

71,221
(25 RCTs)

⊕⊕⊕⊝
MODERATE 2 3

23 per 1000

9 per 1000
(8 to 11)

Influenza‐like illness
assessed by subjective report

Timing of assessment: most studies tested vaccines over a single influenza season

Low1

RR 0.84
(0.75 to 0.95)

25,795
(16 RCTs)

⊕⊕⊕⊝
MODERATE 2 4

40 per 1000

34 per 1000
(30 to 38)

Moderate

215 per 1000

181 per 1000
(161 to 205)

High

910 per 1000

764 per 1000
(683 to 864)

Hospitalisations

Timing of assessment: single influenza season

Study population1

RR 0.96
(0.85 to 1.08)

11,924
(3 RCTs)

⊕⊕⊝⊝
LOW 5 6

147 per 1000

141 per 1000
(125 to 158)

Time off work

Timing of assessment: single influenza season

Study population1

NA

3726

(4 RCTs)

⊕⊕⊝⊝
LOW 7 8

Average number of days lost per person ranged from 0.2 to 2 days over the season.

Average reduction in working days lost following vaccination was 0.04 days fewer (0.14 fewer to 0.06 days more)

Fever

assessed by subjective report

Timing of assessment: single influenza season

Study population1

RR 1.55
(1.26 to 1.91)

23,850
(13 RCTs)

⊕⊕⊕⊕
HIGH

15 per 1000

23 per 1000
(19 to 28)

Nausea or vomiting
assessed by subjective report

Timing of assessment: single influenza season

Study population1

RR 1.80
(0.65 to 5.04)

6315
(4 RCTs)

⊕⊕⊝⊝
LOW 6 7

37 per 1000

66 per 1000
(24 to 185)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; NA: not applicable; RCT: randomised controlled trial; RR: risk ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: We are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Control group risk calculated as the sum of events over total sample size from the control groups. For the outcome of influenza‐like illness, control group risk was stratified as low, moderate (or median), and high due to variation in risk groups across the studies. For the remaining outcomes, the control group risk was taken as aggregate.
2Sensitivity analysis by excluding studies with two or more domains at unclear risk of bias did not meaningfully alter the direction, size, or precision of effect. We are confident that bias is unlikely to exaggerate the intervention effect because the absolute reduction in influenza and relative reduction in the risk of influenza‐like illness are small with vaccination.
3Downgraded one level due to serious indirectness. Uncertainty over definition, surveillance and testing of influenza in older trials.
4Downgraded one level for serious inconsistency. There is discordance between the direction and size of effects across the studies. Different definitions of influenza‐like illness across the studies could explain why there is variation in the event rates across the control arms.
5Downgraded one level due to serious risk of bias. Meta‐analysis heavily influenced by a large study with high risk of bias across several domains.
6Downgraded one level due to serious imprecision. Confidence interval includes meaningful reduction and increase in effect.
7Downgraded one level due to serious risk of bias. Effect is influenced by studies judged to be at unclear risk of bias.
8Downgraded one level due to serious inconsistency. Direction and magnitude of effect differed across the studies (I2 = 82%). Wide confidence interval reflects the range of study effect sizes.

Figuras y tablas -
Summary of findings for the main comparison. Inactivated parenteral influenza vaccine compared to placebo or 'do nothing' for preventing influenza in healthy adults
Table 1. Studies included in the various versions of this review and their impact on our conclusions

Review version (searches date)

Number of included trials (RCTs/CCTs)

Number of included observational studies

Estimates of effect (RCTs/CCTs only)

Conclusions (1‐2 lines from abstract)

Version 1

Demicheli 1999

(6 July 1999)

20

0

Clinical influenza

TIV = 24% (95% CI 15% to 32%)

LAIV = 13% (95% CI 5% to 20%)

IAV = 40% (95% CI 13% to 59%)

Laboratory‐confirmed influenza

TIV = 68% (95% CI 49% to 79%)

LAIV = 48% (95% CI 24% to 64%)

IAV = no evidence

Influenza vaccines are effective in reducing serologically confirmed cases of influenza A. However, they are not as effective in reducing cases of clinical influenza. The use of WHO recommended vaccines appears to enhance their effectiveness in practice.

Version 2
Demicheli 2004

(24 May 2004)

25

0

Clinical influenza

TIV = 25% (95% CI 13% to 35%)

LAIV = 15% (95% CI 8% to 21%)

IAV = 40% (95% CI 13% to 59%)

Laboratory‐confirmed influenza

TIV = 70% (95% CI 56% to 80%)

LAIV = 48% (95% CI 24% to 64%)

IAV = no evidence

Influenza vaccines are effective in reducing serologically confirmed cases of influenza. However, they are not as effective in reducing cases of clinical influenza and number of working days lost. Universal immunisation of healthy adults is not supported by the results of this review.

Version 3
Jefferson 2007

(16 February 2007)

38

10

(for harms only)

ILI

TIV = 30% (95% CI 17% to 41%)

LAIV = n.s.

IAV = n.s.

Influenza

TIV = 80% (95% CI 56% to 81%)

LAIV = 56% (95% CI 19% to 76%)

IAV = no evidence

Influenza vaccines are effective in reducing cases of influenza, especially when the content accurately predicts circulating types and circulation is high. However, they are less effective in reducing cases of influenza‐like illness and have a modest impact on working days lost. There is insufficient evidence to assess their impact on complications. Whole‐virion monovalent vaccines may perform best in a pandemic.

Version 4
Jefferson 2010

(15 June 2010)

40

10

(for harms only)

ILI

TIV = 30% (95% CI 17% to 41%)

LAIV = n.s.

IAV = n.s.

Influenza

TIV = 73% (95% CI 54% to 84%)

LAIV = 56% (95% CI 19% to 76%)

IAV = no evidence

Influenza vaccines have a modest effect in reducing influenza symptoms and working days lost. There is no evidence that they affect complications, such as pneumonia, or transmission.

Version 5
Jefferson 2014

(4 March 2014)

48

42

ILI

TIV = 17% (95% CI 11% to 23%)

LAIV = n.s.

IAV = n.s.

Influenza

TIV = 63% (95% CI 55% to 69%)

LAIV = 45% (95% CI 18% to 63%)

IAV = n.s.

Influenza vaccines have a very modest effect in reducing influenza symptoms and working days lost in the general population, including pregnant women. No evidence of association between influenza vaccination and serious adverse events was found in the comparative studies considered in the review.

CCT: controlled clinical trial
CI: confidence interval
IAV: inactivated aerosol vaccines
ILI: influenza‐like illness
LAIV: live attenuated vaccines
n.s.: not statistically significant
RCT: randomised controlled trial
TIV: trivalent inactivated vaccines
WHO: World Health Organization

Versions 1 and 2

Effect estimates are from Comparison 02 (At least one vaccine recommended for that year versus placebo or other vaccine).

A clinically defined case was assumed as any case definition based on symptoms without further specification.

A clinically defined case (specific definition) was defined as:

  • 'flu‐like illness' according to a predefined list of symptoms (including the Centers for Disease Control and Prevention case definition for surveillance);

  • 'upper respiratory illness' according to a predefined list of symptoms.

When more than one definition was given for the same trial, data related to the more specific definition were included.

In Analysis 2.1 from versions 1 and 2, studies with both definitions are included.

Evidence about effectiveness of aerosol inactivated vaccine comes only from studies carried out during the 1968‐69 pandemic. From version 3 onwards, specific comparisons have been added.

Versions 3, 4, 5

Recommended vaccine matching circulating strains.

Version 5

Out of the 42 included observational studies, 8 assessed efficacy or effectiveness of vaccine, or both, when administered during pregnancy (6 cohort and 2 case‐control studies).

Version 6 (current)

In two new RCTs included in this version, vaccination was performed during pregnancy.

Regarding efficacy/effectiveness of TIV administered in general population, estimates assessed by applying random‐effects model were 16% (95% CI 9% to 23%) against ILI and 62% (95% CI 52% to 69%) against influenza, respectively.

In a previous interim unpublished update before the decision to stabilise the review was made, a further 16 observational studies were included: 3 case‐control and 2 cohort studies assessing the safety of influenza vaccine administration in general population, 10 cohort studies assessing the safety of influenza vaccine administration during pregnancy, and one cohort study assessing efficacy/effectiveness of the vaccine administration during pregnancy. In this 2016 updated review, we included a total of 160 studies (137 data sets), while we no longer updated searches for observational comparative studies.

Figuras y tablas -
Table 1. Studies included in the various versions of this review and their impact on our conclusions
Table 2. Risk of bias in included studies

Study design

High risk

Low risk

Unclear risk

Total

Case‐control

3

2

18

23

Cohort

14

8

18

40

RCT/CCT

7

12

55

74

Total

24

22

91

137

CCT: controlled clinical trial
RCT: randomised controlled trial

This table displays the overall methodological quality assessment of the included studies described in the text and represented in extended form (with all items of the tools) in Figure 1.

Figuras y tablas -
Table 2. Risk of bias in included studies
Table 3. Funding source of included studies

Study design

Government, institutional, or public

Industry

Mixed

Total

Case‐control

14

2

2

18

Cohort

33

5

2

40

RCT/CCT

32

15

5

52

Total

79

22

9

110

CCT: controlled clinical trial
RCT: randomised controlled trial

Figuras y tablas -
Table 3. Funding source of included studies
Table 4. Sensitivity analysis for 'Summary of findings' table outcomes

Outcome (analysis)

All studies (primary analysis)

Studies at low risk of bias (sensitivity analysis)

Influenza (Analysis 1.1)

RR 0.41 (0.36 to 0.47)

RR 0.34 (0.25 to 0.45)

Influenza‐like illness (Analysis 1.2)

RR 0.84 (0.75 to 0.95)

RR 0.82 (0.69 to 0.98)

Hospitalisations (Analysis 1.8)

RR 0.96 (0.85 to 1.08)

RR 2.89 (0.12 to 70.68)

Fever (Analysis 1.11.2)

RR 1.55 (1.26 to 1.91)

RR 1.59 (1 to 2.53)

Nausea/vomiting (Analysis 1.11.5)

RR 1.80 (0.65 to 5.04)

RR 7.05 (1.61 to 30.87)

RR: risk ratio

Figuras y tablas -
Table 4. Sensitivity analysis for 'Summary of findings' table outcomes
Comparison 1. Inactivated parenteral influenza vaccine versus placebo or 'do nothing'

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza Show forest plot

25

71221

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.36, 0.47]

1.1 WHO recommended ‐ matching vaccine

15

46444

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.34, 0.49]

1.2 WHO recommended ‐ vaccine matching absent or unknown

7

15068

Risk Ratio (M‐H, Random, 95% CI)

0.45 [0.34, 0.59]

1.3 Monovalent not WHO recommended ‐ vaccine matching

2

9675

Risk Ratio (M‐H, Random, 95% CI)

0.22 [0.10, 0.52]

1.4 Monovalent not WHO recommended ‐ vaccine matching ‐ high dose

1

34

Risk Ratio (M‐H, Random, 95% CI)

0.11 [0.00, 2.49]

2 Influenza‐like illness Show forest plot

16

25795

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.75, 0.95]

2.1 WHO recommended ‐ matching vaccine

7

4760

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.77, 0.91]

2.2 WHO recommended ‐ vaccine matching absent or unknown

7

20942

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.69, 1.18]

2.3 Monovalent not WHO recommended ‐ vaccine matching

1

59

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.28, 3.70]

2.4 Monovalent not WHO recommended ‐ vaccine matching ‐ high dose

1

34

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.09, 2.30]

3 Physician visits Show forest plot

2

2308

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.40, 1.89]

3.1 WHO recommended ‐ matching vaccine

1

1178

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.37, 0.91]

3.2 WHO recommended ‐ vaccine matching absent or unknown

1

1130

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.90, 1.83]

4 Days ill Show forest plot

3

3133

Mean Difference (IV, Random, 95% CI)

‐0.21 [‐0.98, 0.56]

4.1 WHO recommended ‐ matching vaccine

2

2003

Mean Difference (IV, Random, 95% CI)

‐0.58 [‐0.85, ‐0.32]

4.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

0.66 [0.16, 1.16]

5 Times any drugs were prescribed Show forest plot

2

2308

Mean Difference (IV, Random, 95% CI)

‐0.01 [‐0.03, 0.01]

5.1 WHO recommended ‐ matching vaccine

1

1178

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.04, ‐0.00]

5.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.00, 0.00]

6 Times antibiotic was prescribed Show forest plot

2

2308

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.03, ‐0.01]

6.1 WHO recommended ‐ matching vaccine

1

1178

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.03, ‐0.01]

6.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

‐0.01 [‐0.03, 0.01]

7 Working days lost Show forest plot

4

3726

Mean Difference (IV, Random, 95% CI)

‐0.04 [‐0.14, 0.06]

7.1 WHO recommended ‐ matching vaccine

3

2596

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐0.19, 0.02]

7.2 WHO recommended ‐ matching absent or unknown

1

1130

Mean Difference (IV, Random, 95% CI)

0.09 [0.00, 0.18]

8 Hospitalisations Show forest plot

3

11924

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.85, 1.08]

8.1 WHO recommended ‐ matching vaccine

1

1178

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

8.2 WHO recommended ‐ vaccine matching absent or unknown

1

1130

Risk Ratio (M‐H, Random, 95% CI)

2.89 [0.12, 70.68]

8.3 Monovalent not WHO recommended ‐ vaccine matching

1

9616

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.85, 1.08]

9 Clinical cases (clinically defined without clear definition) Show forest plot

3

4259

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.72, 1.05]

9.1 WHO recommended ‐ matching vaccine

2

2056

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.64, 1.25]

9.2 WHO recommended ‐ vaccine matching absent or unknown

1

2203

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.69, 0.99]

10 Local harms Show forest plot

20

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

10.1 Local ‐ tenderness/soreness

20

35655

Risk Ratio (M‐H, Random, 95% CI)

3.13 [2.44, 4.02]

10.2 Local ‐ erythema

9

29499

Risk Ratio (M‐H, Random, 95% CI)

2.59 [1.77, 3.78]

10.3 Local ‐ induration

3

7786

Risk Ratio (M‐H, Random, 95% CI)

4.28 [1.25, 14.67]

10.4 Local ‐ arm stiffness

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.54, 4.83]

10.5 Local ‐ combined endpoint (any or highest symptom)

11

12307

Risk Ratio (M‐H, Random, 95% CI)

2.44 [1.82, 3.28]

11 Systemic harms Show forest plot

17

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

11.1 Systemic ‐ myalgia

11

35008

Risk Ratio (M‐H, Random, 95% CI)

1.74 [1.41, 2.14]

11.2 Systemic ‐ fever

13

23850

Risk Ratio (M‐H, Random, 95% CI)

1.55 [1.26, 1.91]

11.3 Systemic ‐ headache

14

35999

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.99, 1.30]

11.4 Systemic ‐ fatigue or indisposition

12

35788

Risk Ratio (M‐H, Random, 95% CI)

1.19 [1.05, 1.36]

11.5 Systemic ‐ nausea/vomiting

4

6315

Risk Ratio (M‐H, Random, 95% CI)

1.80 [0.65, 5.04]

11.6 Systemic ‐ malaise

3

26111

Risk Ratio (M‐H, Random, 95% CI)

1.51 [1.18, 1.92]

11.7 Systemic ‐ combined endpoint (any or highest symptom)

6

2128

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.87, 1.53]

Figuras y tablas -
Comparison 1. Inactivated parenteral influenza vaccine versus placebo or 'do nothing'
Comparison 2. Live aerosol influenza vaccine versus placebo or 'do nothing'

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza Show forest plot

9

11579

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.35, 0.62]

1.1 WHO recommended ‐ matching vaccine

4

6584

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.37, 0.82]

1.2 WHO recommended ‐ vaccine matching absent or unknown

3

4568

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.27, 0.68]

1.3 Non WHO recommended ‐ vaccine matching absent or unknown

2

427

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.08, 0.56]

2 Influenza‐like illness Show forest plot

6

12688

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.84, 0.96]

2.1 WHO recommended ‐ matching vaccine

2

4254

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.76, 1.12]

2.2 WHO recommended ‐ vaccine matching absent or unknown

3

8150

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.82, 0.97]

2.3 Non WHO recommended ‐ vaccine matching absent or unknown

1

284

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.73, 1.16]

3 Influenza cases (clinically defined without clear definition) Show forest plot

3

23900

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.71, 1.11]

3.1 WHO recommended ‐ matching vaccine

1

1931

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.49, 0.80]

3.2 WHO recommended ‐ vaccine matching absent or unknown

1

2082

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.88, 1.25]

3.3 Non WHO recommended ‐ vaccine matching absent or unknown

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.92, 1.05]

4 Local harms Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Local ‐ upper respiratory infection symptoms

6

496

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.22, 2.27]

4.2 Local ‐ cough

6

2401

Risk Ratio (M‐H, Random, 95% CI)

1.51 [1.08, 2.10]

4.3 Local ‐ coryza

2

4782

Risk Ratio (M‐H, Random, 95% CI)

1.56 [1.26, 1.94]

4.4 Local ‐ sore throat

7

6940

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.49, 1.86]

4.5 Local ‐ hoarseness

1

306

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.51, 2.83]

4.6 Local ‐ combined endpoint (any or highest symptom)

3

4921

Risk Ratio (M‐H, Random, 95% CI)

1.56 [1.31, 1.87]

5 Systemic harms Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Systemic ‐ myalgia

4

1318

Risk Ratio (M‐H, Random, 95% CI)

2.47 [1.26, 4.85]

5.2 Systemic ‐ fever

4

1318

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.54, 1.92]

5.3 Systemic ‐ fatigue or indisposition

3

1018

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.93, 2.07]

5.4 Systemic ‐ headache

2

975

Risk Ratio (M‐H, Random, 95% CI)

1.54 [1.09, 2.18]

5.5 Systemic ‐ combined endpoint (any or highest symptom)

5

1018

Risk Ratio (M‐H, Random, 95% CI)

1.40 [0.82, 2.38]

Figuras y tablas -
Comparison 2. Live aerosol influenza vaccine versus placebo or 'do nothing'
Comparison 3. Inactivated aerosol influenza vaccine versus placebo or 'do nothing'

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza Show forest plot

1

1348

Odds Ratio (M‐H, Random, 95% CI)

0.38 [0.14, 1.02]

1.1 WHO recommended ‐ vaccine matching absent or unknown

1

1348

Odds Ratio (M‐H, Random, 95% CI)

0.38 [0.14, 1.02]

1.2 WHO recommended ‐ matching vaccine

0

0

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Local harms Show forest plot

3

1578

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.71, 1.27]

2.1 Local ‐ sore throat

3

1500

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.54, 1.33]

2.2 Local ‐ combined endpoint (any or highest symptom)

1

78

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.71, 1.48]

3 Systemic harms Show forest plot

3

1880

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.71, 1.62]

3.1 Systemic ‐ myalgia

2

151

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.36, 2.25]

3.2 Systemic ‐ fatigue or indisposition

2

151

Risk Ratio (M‐H, Random, 95% CI)

1.40 [0.52, 3.75]

3.3 Systemic ‐ headache

2

151

Risk Ratio (M‐H, Random, 95% CI)

1.52 [0.85, 2.72]

3.4 Systemic ‐ fever

1

1349

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.03, 7.80]

3.5 Systemic ‐ combined endpoint (any or highest symptom)

1

78

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.12, 1.04]

Figuras y tablas -
Comparison 3. Inactivated aerosol influenza vaccine versus placebo or 'do nothing'
Comparison 4. Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza in mothers Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

1.1 TIV containing pH1N1

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Influenza‐like illness in mothers Show forest plot

2

2342

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.20, 1.95]

2.1 TIV containing pH1N1

1

2116

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.79, 1.16]

2.2 Monovalent pH1N1

1

226

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.08, 1.02]

3 Influenza in newborn Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 TIV containing pH1N1

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Influenza‐like illness in newborn Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4.1 TIV containing pH1N1

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 4. Inactivated parenteral influenza vaccine versus placebo or 'do nothing' administered during pregnancy
Comparison 5. Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal inactivated vaccine effectiveness in mothers ‐ pregnant women Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 H1N1 ‐ vaccine ‐ effectiveness ILI (unadjusted data)

1

7328

Risk Ratio (M‐H, Random, 95% CI)

0.11 [0.06, 0.21]

1.2 Seasonal ‐ vaccine ‐ effectiveness ILI ‐ (unadjusted data)

3

50507

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.24, 1.18]

2 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women Show forest plot

2

Hazard Ratio (Random, 95% CI)

Subtotals only

2.1 Seasonal vaccine effectiveness ILI (HR adjusted data)

2

Hazard Ratio (Random, 95% CI)

0.96 [0.90, 1.03]

3 Seasonal inactivated vaccine effectiveness in newborns ‐ pregnant women Show forest plot

1

Risk Ratio (Random, 95% CI)

Subtotals only

3.1 Seasonal vaccine effectiveness ILI (RR adjusted data)

1

Risk Ratio (Random, 95% CI)

0.92 [0.73, 1.16]

3.2 Seasonal vaccine efficacy influenza ‐ laboratory‐confirmed

1

Risk Ratio (Random, 95% CI)

0.59 [0.37, 0.94]

4 H1N1 vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women Show forest plot

15

Odds Ratio (Random, 95% CI)

Subtotals only

4.1 Abortion (OR adjusted data)

5

Odds Ratio (Random, 95% CI)

0.75 [0.62, 0.90]

4.2 Abortion (HR adjusted data)

3

Odds Ratio (Random, 95% CI)

0.81 [0.63, 1.04]

4.3 Congenital malformation (OR adjusted data)

6

Odds Ratio (Random, 95% CI)

1.11 [0.99, 1.23]

4.4 Prematurity (< 37 weeks) (OR unadjusted data)

11

Odds Ratio (Random, 95% CI)

0.76 [0.67, 0.85]

4.5 Prematurity (< 37 weeks) (OR adjusted data)

7

Odds Ratio (Random, 95% CI)

0.84 [0.76, 0.93]

4.6 Prematurity (< 37 weeks) (HR adjusted data)

2

Odds Ratio (Random, 95% CI)

1.11 [0.46, 2.68]

4.7 Prematurity (< 37 weeks) vaccination in I trimester OR adjusted data

2

Odds Ratio (Random, 95% CI)

1.08 [0.92, 1.28]

4.8 Prematurity (< 37 weeks) vaccination in II/III trimester OR adjusted data

2

Odds Ratio (Random, 95% CI)

0.96 [0.87, 1.06]

4.9 Neonatal death (OR adjusted data)

2

Odds Ratio (Random, 95% CI)

1.09 [0.40, 2.95]

5 Seasonal vaccine ‐ safety ‐ pregnancy‐related outcomes ‐ pregnant women Show forest plot

7

Odds Ratio (Random, 95% CI)

Subtotals only

5.1 Abortion (OR unadjusted data)

1

Odds Ratio (Random, 95% CI)

0.60 [0.41, 0.86]

5.2 Congenital malformation (OR unadjusted data)

2

Odds Ratio (Random, 95% CI)

0.55 [0.08, 3.73]

5.3 Prematurity (OR unadjusted data)

6

Odds Ratio (Random, 95% CI)

0.95 [0.82, 1.10]

5.4 Prematurity (OR adjusted data)

2

Odds Ratio (Random, 95% CI)

0.93 [0.82, 1.06]

5.5 Neonatal death (OR unadjusted data)

1

Odds Ratio (Random, 95% CI)

0.55 [0.35, 0.88]

6 Seasonal vaccine containing H1N1 Show forest plot

2

Risk Ratio (Random, 95% CI)

Subtotals only

6.1 Prematurity (37 weeks) vaccination in I trimester HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.63 [0.76, 3.47]

6.2 Prematurity (< 37 weeks) vaccination in II trimester HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.48 [0.21, 10.64]

6.3 Prematurity (< 37 weeks) vaccination in III trimester HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.37 [0.44, 4.25]

6.4 Prematurity (< 37 weeks) vaccination at any time during pregnancy HR adjusted data

2

Risk Ratio (Random, 95% CI)

1.75 [0.57, 5.44]

Figuras y tablas -
Comparison 5. Inactivated parenteral influenza vaccine versus placebo ‐ cohort studies
Comparison 6. Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Effectiveness in newborns ‐ pregnant women (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

0.24 [0.04, 1.40]

1.1 Seasonal vaccine ‐ effectiveness ‐ ILI ‐ pregnant women

2

Odds Ratio (Random, 95% CI)

0.24 [0.04, 1.40]

2 Seasonal vaccine safety ‐ pregnancy‐related outcomes (adjusted data) Show forest plot

1

Odds Ratio (Random, 95% CI)

0.80 [0.36, 1.78]

2.1 Abortion

1

Odds Ratio (Random, 95% CI)

0.80 [0.36, 1.78]

Figuras y tablas -
Comparison 6. Inactivated parenteral influenza vaccine versus placebo ‐ case‐control studies
Comparison 7. Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal influenza vaccination and Guillain‐Barré syndrome Show forest plot

3

Risk Ratio (Random, 95% CI)

1.28 [0.85, 1.93]

1.1 General population (adjusted data)

2

Risk Ratio (Random, 95% CI)

1.29 [0.83, 2.02]

1.2 Pregnant women (unadjusted data)

1

Risk Ratio (Random, 95% CI)

0.65 [0.03, 15.95]

Figuras y tablas -
Comparison 7. Serious adverse events: Guillain‐Barré syndrome ‐ cohort studies
Comparison 8. Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 2009 to 2010 A/H1N1 ‐ general population (unadjusted data) Show forest plot

6

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 < 7 weeks

6

1528

Odds Ratio (M‐H, Random, 95% CI)

2.22 [1.14, 4.31]

1.2 At any time

6

1656

Odds Ratio (M‐H, Random, 95% CI)

1.69 [0.87, 3.29]

2 2009 to 2010 A/H1N1 ‐ general population (adjusted data) Show forest plot

4

Odds Ratio (Random, 95% CI)

0.83 [0.39, 1.75]

2.1 < 7 weeks

4

Odds Ratio (Random, 95% CI)

0.92 [0.35, 2.40]

2.2 > 6 weeks (i.e. at any time)

3

Odds Ratio (Random, 95% CI)

0.71 [0.22, 2.32]

3 Seasonal influenza vaccination general population (adjusted data) Show forest plot

1

Odds Ratio (Random, 95% CI)

1.38 [0.18, 10.43]

Figuras y tablas -
Comparison 8. Serious adverse events: Guillain‐Barré syndrome ‐ case‐control studies
Comparison 9. Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza vaccination (seasonal) ‐ demyelinating diseases (unadjusted data) Show forest plot

1

223898

Odds Ratio (M‐H, Random, 95% CI)

0.16 [0.02, 1.25]

1.1 General population

0

0

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Pregnant women

1

223898

Odds Ratio (M‐H, Random, 95% CI)

0.16 [0.02, 1.25]

2 Influenza vaccination (H1N1) ‐ demyelinating diseases (unadjusted) Show forest plot

1

144252

Risk Ratio (M‐H, Random, 95% CI)

2.06 [0.51, 8.22]

Figuras y tablas -
Comparison 9. Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ cohort studies
Comparison 10. Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza vaccination (seasonal) ‐ general population ‐ demyelinating diseases (unadjusted data) Show forest plot

4

8009

Odds Ratio (M‐H, Random, 95% CI)

0.96 [0.79, 1.17]

2 Influenza vaccination (seasonal) ‐ general population ‐ multiple sclerosis (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

0.76 [0.54, 1.08]

3 Influenza vaccination (seasonal) ‐ general population ‐ optic neuritis (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

1.03 [0.82, 1.30]

Figuras y tablas -
Comparison 10. Serious adverse events: demyelinating diseases (multiple sclerosis, optic neuritis) ‐ case‐control studies
Comparison 11. Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal influenza vaccine ‐ HR (adjusted data) Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

1.1 General population

0

Hazard Ratio (Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Pregnant women

1

Hazard Ratio (Random, 95% CI)

0.90 [0.68, 1.19]

2 Seasonal influenza vaccine (unadjusted data) Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 General population

0

0

Odds Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Pregnant women

1

223898

Odds Ratio (M‐H, Random, 95% CI)

0.92 [0.70, 1.20]

Figuras y tablas -
Comparison 11. Serious adverse events: immune thrombocytopenic purpura ‐ cohort studies
Comparison 12. Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Seasonal influenza vaccine ‐ general population (adjusted data) Show forest plot

2

Odds Ratio (Random, 95% CI)

Subtotals only

1.1 < 2 months

2

Odds Ratio (Random, 95% CI)

1.87 [0.43, 8.06]

1.2 < 6 months

1

Odds Ratio (Random, 95% CI)

0.90 [0.55, 1.47]

1.3 < 12 months

1

Odds Ratio (Random, 95% CI)

0.70 [0.47, 1.04]

2 Seasonal influenza vaccine ‐ general population (unadjusted data) Show forest plot

2

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 < 2 months

2

1926

Odds Ratio (M‐H, Random, 95% CI)

1.72 [0.48, 6.15]

2.2 < 6 months

1

1065

Odds Ratio (M‐H, Random, 95% CI)

0.92 [0.59, 1.43]

2.3 < 12 months

1

1066

Odds Ratio (M‐H, Random, 95% CI)

0.72 [0.50, 1.05]

Figuras y tablas -
Comparison 12. Serious adverse events: immune thrombocytopenic purpura ‐ case‐control studies
Comparison 13. 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

3

3065

Risk Ratio (M‐H, Random, 95% CI)

0.71 [0.57, 0.88]

1.1 Standard recommended parenteral ‐ non‐matching ‐ 1 dose

3

2715

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.57, 0.95]

1.2 Standard recommended parenteral ‐ non‐matching ‐ 2 doses

1

350

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.44, 0.98]

2 Influenza Show forest plot

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.87]

2.1 Standard recommended parenteral ‐ non‐matching

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.87]

3 Hospitalisations Show forest plot

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.41, 1.68]

3.1 Standard recommended parenteral ‐ non‐matching

1

2072

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.41, 1.68]

4 Pneumonia Show forest plot

1

2072

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.14, 7.17]

4.1 Standard recommended parenteral ‐ non‐matching

1

2072

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.14, 7.17]

Figuras y tablas -
Comparison 13. 1968 to 1969 pandemic: inactivated polyvalent parenteral influenza vaccine versus placebo
Comparison 14. 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

4

4580

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.25, 0.48]

1.1 WHO recommended parenteral ‐ matching vaccine ‐ 1 dose

4

4226

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.23, 0.53]

1.2 WHO recommended parenteral ‐ matching vaccine ‐ 2 doses

1

354

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.22, 0.57]

2 Influenza Show forest plot

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.07 [0.02, 0.31]

2.1 WHO recommended parenteral ‐ matching vaccine

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.07 [0.02, 0.31]

3 Hospitalisations Show forest plot

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.13, 0.94]

3.1 WHO recommended parenteral ‐ matching vaccine

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.13, 0.94]

4 Pneumonia Show forest plot

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.05, 6.51]

4.1 WHO recommended parenteral ‐ matching vaccine

1

1923

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.05, 6.51]

5 Working days lost Show forest plot

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

5.1 WHO recommended parenteral ‐ matching vaccine

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

6 Days ill Show forest plot

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

6.1 WHO recommended ‐ matching vaccine

1

1667

Mean Difference (IV, Random, 95% CI)

‐0.45 [‐0.60, ‐0.30]

Figuras y tablas -
Comparison 14. 1968 to 1969 pandemic: inactivated monovalent parenteral influenza vaccine versus placebo
Comparison 15. 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

2

1000

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.46, 0.95]

1.1 Inactivated polyvalent aerosol vaccine versus placebo ‐ non‐matching ‐ 1 dose

2

644

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.32, 1.27]

1.2 Inactivated polyvalent aerosol vaccine versus placebo ‐ non‐matching ‐ 2 doses

1

356

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.44, 0.97]

Figuras y tablas -
Comparison 15. 1968 to 1969 pandemic: inactivated polyvalent aerosol influenza vaccine versus placebo
Comparison 16. 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza‐like illness Show forest plot

2

1009

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.32, 0.91]

1.1 Inactivated monovalent aerosol vaccine versus placebo ‐ matching ‐ 1 dose

2

650

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.17, 1.41]

1.2 Inactivated monovalent aerosol vaccine versus placebo ‐ matching ‐ 2 doses

1

359

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.38, 0.86]

Figuras y tablas -
Comparison 16. 1968 to 1969 pandemic: inactivated monovalent aerosol influenza vaccine versus placebo
Comparison 17. 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Influenza cases (clinically defined without clear definition) Show forest plot

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.92, 1.05]

1.1 Non‐matching

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.92, 1.05]

2 Complications (bronchitis, otitis, pneumonia) Show forest plot

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.03, 2.24]

2.1 Non‐matching

1

19887

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.03, 2.24]

Figuras y tablas -
Comparison 17. 1968 to 1969 pandemic: live aerosol influenza vaccine versus placebo