The how's and what's of vaccine reactogenicity

Caroline Hervé, Béatrice Laupèze, Giuseppe Del Giudice, Arnaud M Didierlaurent, Fernanda Tavares Da Silva, Caroline Hervé, Béatrice Laupèze, Giuseppe Del Giudice, Arnaud M Didierlaurent, Fernanda Tavares Da Silva

Abstract

Reactogenicity represents the physical manifestation of the inflammatory response to vaccination, and can include injection-site pain, redness, swelling or induration at the injection site, as well as systemic symptoms, such as fever, myalgia, or headache. The experience of symptoms following vaccination can lead to needle fear, long-term negative attitudes and non-compliant behaviours, which undermine the public health impact of vaccination. This review presents current knowledge on the potential causes of reactogenicity, and how host characteristics, vaccine administration and composition factors can influence the development and perception of reactogenicity. The intent is to provide an overview of reactogenicity after vaccination to help the vaccine community, including healthcare professionals, in maintaining confidence in vaccines by promoting vaccination, setting expectations for vaccinees about what might occur after vaccination and reducing anxiety by managing the vaccination setting.

Keywords: Signs and symptoms; Vaccines.

Conflict of interest statement

Competing interestsAll authors are employees of the GSK group of companies. A.D., G.D.G., B.L. and F.T.D.S. hold shares in the GSK group of companies. A.D. owns patents related to AS01 that are unrelated to the topic of this paper.

© The Author(s) 2019.

Figures

Fig. 1
Fig. 1
Summary of expected biological mechanisms underlying the development of reactogenicity symptoms. Vaccine antigens and immune enhancers (as adjuvants) injected into the muscle are recognised by the body as potential pathogens and/or danger signals. This recognition leads to the stimulation of local cells, followed by the recruitment of blood immune cells to the local site and the production of different soluble factors including vasodilators and cytokines, which may trigger the development of signs and symptoms of local inflammation (pain, redness and swelling). The passage of some of those factors in the bloodstream, as well as the production of other systemic factors by immune blood cells or distant organs (e.g. liver), may contribute to the development of general symptoms (fever, myalgia, headache etc) in the vaccinee. CRP C-reactive protein
Fig. 2
Fig. 2
Hypothesised link between the innate immune response induced by vaccination and reactogenicity. Upon vaccination, inflammation is triggered by innate immune activation of pattern-recognition receptors (PRR) including Toll-like receptors (TLRs) that recognize and bind antigens (green circle in skeletal muscle) and potential immune enhancers (purple circle in skeletal muscle) present in the vaccine formulation. Resident immune cells, mast cells, monocytes and macrophages are activated within minutes of injection and release soluble factors (proinflammatory cytokines, chemokines, effectors of the complement cascade) and vasodilators, that allow cell recruitment from blood but also lead to the development of redness and swelling symptoms. These newly recruited immune cells, mainly composed of blood-born neutrophils, monocytes and T lymphocytes, also contribute to pain sensation by releasing soluble factors, such as cytokines, prostaglandins or ATP, that can directly interact with local sensory receptors called nociceptors and cause pain through the fast neural route if the threshold is reached. Once produced, cytokines act both locally in autocrine and paracrine manners, and may act systemically at distant organs, leading to the production of C-reactive protein and other acute phase proteins by the liver. Several immune-to-brain signaling pathways may propagate an inflammatory response to the central nervous system after peripheral activation of the innate immune system (slow humoral route), leading to the development of fever and sickness behaviours
Fig. 3
Fig. 3
Summary of vaccination and host factors that can influence reactogenicity. As a general concept, all the conditions that can influence the immune, the endocrine or the hormonal systems of the host (intrinsic factors), all the conditions that can increase tissue stress (administration factors) and the components that activate innate immunity contained in the vaccines (vaccine factors) could theoretically impact the incidence and severity of local and general symptoms induced after vaccination
Fig. 4
Fig. 4
Practical recommendations to decrease side-effects that may occur at the time of injection: adapted from the World Health Organization recommendations. *There are no additional evidence-based, age-specific recommendations available for adolescents beyond the general measures recommended for all age groups

References

    1. Di Pasquale A, et al. Vaccine safety evaluation: practical aspects in assessing benefits and risks. Vaccine. 2016;34:6672–6680. doi: 10.1016/j.vaccine.2016.10.039.
    1. Bonhoeffer J, et al. The Brighton Collaboration: addressing the need for standardized case definitions of adverse events following immunization (AEFI) Vaccine. 2002;21:298–302. doi: 10.1016/S0264-410X(02)00449-8.
    1. Kohl KS, et al. The Brighton Collaboration: enhancing comparability of vaccine safety data. Pharmacoepidemiol. Drug Saf. 2003;12:335–340. doi: 10.1002/pds.851.
    1. Peltola H, Heinonen OP. Frequency of true adverse reactions to measles-mumps-rubella vaccine. A double-blind placebo-controlled trial in twins. Lancet. 1986;1:939–942. doi: 10.1016/S0140-6736(86)91044-5.
    1. Beutler B. Microbe sensing, positive feedback loops, and the pathogenesis of inflammatory diseases. Immunol. Rev. 2009;227:248–263. doi: 10.1111/j.1600-065X.2008.00733.x.
    1. Moser M, Leo O. Key concepts in immunology. Vaccine. 2010;28:C2–C13. doi: 10.1016/j.vaccine.2010.07.022.
    1. Ishii KJ, Koyama S, Nakagawa A, Coban C, Akira S. Host innate immune receptors and beyond: making sense of microbial infections. Cell host microbe. 2008;3:352–363. doi: 10.1016/j.chom.2008.05.003.
    1. Ren K, Dubner R. Interactions between the immune and nervous systems in pain. Nat. Med. 2010;16:1267–1276. doi: 10.1038/nm.2234.
    1. Stassijns J, Bollaerts K, Baay M, Verstraeten T. A systematic review and meta-analysis on the safety of newly adjuvanted vaccines among children. Vaccine. 2016;34:714–722. doi: 10.1016/j.vaccine.2015.12.024.
    1. Del Giudice, G., Rappuoli, R. & Didierlaurent, A. M. Correlates of adjuvanticity: a review on adjuvants in licensed vaccines. Semin. Immunol. 39, 14–21 (2018).
    1. Garcon, N., Leroux-Roels, G. & Cheng, W. F. Vaccine adjuvants. in Understanding Modern Vaccines: Perspectives in Vaccinology 89–113 (Elsevier, 2011) [Online].
    1. O’Hagan DT, Fox CB. New generation adjuvants—from empiricism to rational design. Vaccine. 2015;33:B14–B20. doi: 10.1016/j.vaccine.2015.01.088.
    1. O’Hagan DT, Ott GS, De Gregorio E, Seubert A. The mechanism of action of MF59—an innately attractive adjuvant formulation. Vaccine. 2012;30:4341–4348. doi: 10.1016/j.vaccine.2011.09.061.
    1. Mosca F, et al. Molecular and cellular signatures of human vaccine adjuvants. Proc. Natl Acad. Sci. USA. 2008;105:10501–10506. doi: 10.1073/pnas.0804699105.
    1. Didierlaurent AM, et al. Enhancement of adaptive immunity by the human vaccine adjuvant AS01 depends on activated dendritic cells. J. Immunol. 2014;193:1920–1930. doi: 10.4049/jimmunol.1400948.
    1. Calabro S, et al. Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine. 2011;29:1812–1823. doi: 10.1016/j.vaccine.2010.12.090.
    1. Morel S, et al. Adjuvant system AS03 containing α-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011;29:2461–2473. doi: 10.1016/j.vaccine.2011.01.011.
    1. Roth J, Harre EM, Rummel C, Gerstberger R, Hubschle T. Signaling the brain in systemic inflammation: role of sensory circumventricular organs. Front. Biosci. 2004;9:290–300. doi: 10.2741/1241.
    1. Rivest S. Molecular insights on the cerebral innate immune system. Brain. Behav. Immun. 2003;17:13–19. doi: 10.1016/S0889-1591(02)00055-7.
    1. Saper CB, Romanovsky AA, Scammell TE. Neural circuitry engaged by prostaglandins during the sickness syndrome. Nat. Neurosci. 2012;15:1088–1095. doi: 10.1038/nn.3159.
    1. Wilhelms DB, et al. Deletion of prostaglandin E2 synthesizing enzymes in brain endothelial cells attenuates inflammatory fever. J. Neurosci. 2014;34:11684–11690. doi: 10.1523/JNEUROSCI.1838-14.2014.
    1. Vasilache AM, Qian H, Blomqvist A. Immune challenge by intraperitoneal administration of lipopolysaccharide directs gene expression in distinct blood-brain barrier cells toward enhanced prostaglandin E(2) signaling. Brain. Behav. Immun. 2015;48:31–41. doi: 10.1016/j.bbi.2015.02.003.
    1. Burny W, et al. Different adjuvants induce common innate pathways that are associated with enhanced adaptive responses against a model antigen in humans. Front Immunol. 2017;8:943. doi: 10.3389/fimmu.2017.00943.
    1. Burny W, et al. Inflammatory parameters associated with systemic reactogenicity following vaccination with adjuvanted hepatitis B vaccines in humans. Vaccine. 2019;37:2004–2015. doi: 10.1016/j.vaccine.2019.02.015.
    1. Herrin DM, et al. Comparison of adaptive and innate immune responses induced by licensed vaccines for Human Papillomavirus. Hum. Vaccin. Immunother. 2014;10:3446–3454. doi: 10.4161/hv.34408.
    1. Sobolev O, et al. Adjuvanted influenza-H1N1 vaccination reveals lymphoid signatures of age-dependent early responses and of clinical adverse events. Nat. Immunol. 2016;17:204–213. doi: 10.1038/ni.3328.
    1. Khurana S, et al. Vaccines with MF59 adjuvant expand the antibody repertoire to target protective sites of pandemic avian H5N1 influenza virus. Sci. Transl. Med. 2010;2:15ra15. doi: 10.1126/scitranslmed.3000624.
    1. Lim PW, Garssen J, Sandalova E. Potential use of salivary markers for longitudinal monitoring of inflammatory immune responses to vaccination. Mediat. Inflamm. 2016;2016:6958293. doi: 10.1155/2016/6958293.
    1. Christian LM, Porter K, Karlsson E, Schultz-Cherry S, Iams JD. Serum proinflammatory cytokine responses to influenza virus vaccine among women during pregnancy versus non-pregnancy. Am. J. Reprod. Immunol. 2013;70:45–53. doi: 10.1111/aji.12117.
    1. Chan CY, et al. Early molecular correlates of adverse events following yellow fever vaccination. JCI Insight. 2017;2:96031. doi: 10.1172/jci.insight.96031.
    1. Huttner A, et al. A dose-dependent plasma signature of the safety and immunogenicity of the rVSV-Ebola vaccine in Europe and Africa. Sci. Transl. Med. 2017;9:eaaj1701. doi: 10.1126/scitranslmed.aaj1701.
    1. Mitchell TC, Casella CR. No pain no gain? Adjuvant effects of alum and monophosphoryl lipid A in pertussis and HPV vaccines. Curr. Opin. Immunol. 2017;47:17–25. doi: 10.1016/j.coi.2017.06.009.
    1. Lewis DJ, Lythgoe MP. Application of “Systems Vaccinology” to evaluate inflammation and reactogenicity of adjuvanted preventative vaccines. J. Immunol. Res. 2015;2015:909406. doi: 10.1155/2015/909406.
    1. El Yousfi M, et al. The inflammatory response to vaccination is altered in the elderly. Mech. Ageing Dev. 2005;126:874–881. doi: 10.1016/j.mad.2005.03.008.
    1. Cook IF. Sex differences in injection site reactions with human vaccines. Hum. Vaccin. 2009;5:441–449. doi: 10.4161/hv.8476.
    1. Klein SL, Jedlicka A, Pekosz A. The Xs and Y of immune responses to viral vaccines. Lancet Infect. Dis. 2010;10:338–349. doi: 10.1016/S1473-3099(10)70049-9.
    1. Pittman PR. Aluminum-containing vaccine associated adverse events: role of route of administration and gender. Vaccine. 2002;20:S48–S50. doi: 10.1016/S0264-410X(02)00172-X.
    1. Weber SK, Schlagenhauf P. Childhood vaccination associated adverse events by sex: a literature review. Travel Med. Infect. Dis. 2014;12:459–480. doi: 10.1016/j.tmaid.2014.01.008.
    1. Griffioen M, Halsey N. Gender differences in immediate hypersensitivity reactions to vaccines: a review of the literature. Public Health Nurs. 2014;31:206–214. doi: 10.1111/phn.12073.
    1. McCarthy MM, Nugent BM, Lenz KM. Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nat. Rev. Neurosci. 2017;18:471–484. doi: 10.1038/nrn.2017.61.
    1. Trigunaite A, Dimo J, Jorgensen TN. Suppressive effects of androgens on the immune system. Cell. Immunol. 2015;294:87–94. doi: 10.1016/j.cellimm.2015.02.004.
    1. Kovats S. Estrogen receptors regulate innate immune cells and signaling pathways. Cell. Immunol. 2015;294:63–69. doi: 10.1016/j.cellimm.2015.01.018.
    1. Mayr FB, et al. Ethnic differences in plasma levels of interleukin-8 (IL-8) and granulocyte colony stimulating factor (G-CSF) Transl. Res. 2007;149:10–14. doi: 10.1016/j.trsl.2006.06.003.
    1. Delaney NL, Esquenazi V, Lucas DP, Zachary AA, Leffell MS. TNF-alpha, TGF-beta, IL-10, IL-6, and INF-gamma alleles among African Americans and Cuban Americans. Report of the ASHI Minority Workshops: Part IV. Hum. Immunol. 2004;65:1413–1419. doi: 10.1016/j.humimm.2004.07.240.
    1. Stowe RP, Peek MK, Cutchin MP, Goodwin JS. Plasma cytokine levels in a population-based study: relation to age and ethnicity. J. Gerontol. A Biol. Sci. Med. Sci. 2010;65:429–433. doi: 10.1093/gerona/glp198.
    1. Rahim-Williams B, Riley JL, 3rd, Williams AK, Fillingim RB. A quantitative review of ethnic group differences in experimental pain response: do biology, psychology, and culture matter? Pain. Med. 2012;13:522–540. doi: 10.1111/j.1526-4637.2012.01336.x.
    1. Segerstrom SC, Miller GE. Psychological stress and the human immune system: a meta-analytic study of 30 years of inquiry. Psychol. Bull. 2004;130:601–630. doi: 10.1037/0033-2909.130.4.601.
    1. Marsland AL, Walsh C, Lockwood K, John-Henderson NA. The effects of acute psychological stress on circulating and stimulated inflammatory markers: a systematic review and meta-analysis. Brain. Behav. Immun. 2017;64:208–219. doi: 10.1016/j.bbi.2017.01.011.
    1. Levi F, Canon C, Dipalma M, Florentin I, Misset JL. When should the immune clock be reset? From circadian pharmacodynamics to temporally optimized drug delivery. Ann. N. Y. Acad. Sci. 1991;618:312–329. doi: 10.1111/j.1749-6632.1991.tb27251.x.
    1. Phillips AC, Gallagher S, Carroll D, Drayson M. Preliminary evidence that morning vaccination is associated with an enhanced antibody response in men. Psychophysiology. 2008;45:663–666. doi: 10.1111/j.1469-8986.2008.00662.x.
    1. Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogenesch JB. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl Acad. Sci. USA. 2014;111:16219–16224. doi: 10.1073/pnas.1408886111.
    1. Singer K, Lumeng CN. The initiation of metabolic inflammation in childhood obesity. J. Clin. Invest. 2017;127:65–73. doi: 10.1172/JCI88882.
    1. Esposito S, et al. Immunogenicity, safety and tolerability of inactivated trivalent influenza vaccine in overweight and obese children. Vaccine. 2016;34:56–60. doi: 10.1016/j.vaccine.2015.11.019.
    1. Petousis-Harris H. Vaccine injection technique and reactogenicity—evidence for practice. Vaccine. 2008;26:6299–6304. doi: 10.1016/j.vaccine.2008.08.052.
    1. Petousis-Harris H, et al. Factors associated with reported pain on injection and reactogenicity to an OMV meningococcal B vaccine in children and adolescents. Hum. Vaccin. Immunother. 2015;11:1875–1880. doi: 10.1080/21645515.2015.1016670.
    1. Vesikari T, Baer M, Willems P. Immunogenicity and safety of a second dose of measles-mumps-rubella-varicella vaccine in healthy children aged 5 to 6 years. Pediatr. Infect. Dis. J. 2007;26:153–158. doi: 10.1097/01.inf.0000250689.09396.21.
    1. Beck BR, Hatz CF, Loutan L, Steffen R. Immunogenicity of booster vaccination with a virosomal hepatitis A vaccine after primary immunization with an aluminum-adsorbed hepatitis A vaccine. J. Travel Med. 2004;11:201–206. doi: 10.2310/7060.2004.19002.
    1. Andrews NJ, et al. Predictors of immune response and reactogenicity to AS03B-adjuvanted split virion and non-adjuvanted whole virion H1N1 (2009) pandemic influenza vaccines. Vaccine. 2011;29:7913–7919. doi: 10.1016/j.vaccine.2011.08.076.
    1. Grabenstein JD, Manoff SB. Pneumococcal polysaccharide 23-valent vaccine: long-term persistence of circulating antibody and immunogenicity and safety after revaccination in adults. Vaccine. 2012;30:4435–4444. doi: 10.1016/j.vaccine.2012.04.052.
    1. Rennels MB. Extensive swelling reactions occurring after booster doses of diphtheria-tetanus-acellular pertussis vaccines. Semin. Pediatr. Infect. Dis. 2003;14:196–198. doi: 10.1016/S1045-1870(03)00033-5.
    1. Ngai AL, et al. Safety and immunogenicity of one vs. two injections of Oka/Merck varicella vaccine in healthy children. Pediatr. Infect. Dis. J. 1996;15:49–54. doi: 10.1097/00006454-199601000-00011.
    1. Dagan R, et al. Safety and immunogenicity of a new formulation of an inactivated hepatitis A vaccine. Vaccine. 1999;17:1919–1925. doi: 10.1016/S0264-410X(98)00461-7.
    1. Castillo de Febres O, et al. Safety, immunogenicity and antibody persistence of an inactivated hepatitis A vaccine in 4 to 15 year old children. Vaccine. 1999;18:656–664. doi: 10.1016/S0264-410X(99)00272-8.
    1. WHO. Tetanus vaccines: WHO position paper - February 2017. Wkly. Epidemiol. Rec. 92, 53–76 (2017).
    1. Barbaud A, Deschildre A, Waton J, Raison-Peyron N, Trechot P. Hypersensitivity and vaccines: an update. Eur. J. Dermatol. 2013;23:135–141.
    1. Hennessen W, Mauler R, Gruschkau H, Lehmann HG. Reactogenicity to primary and repeated vaccination with influenza split virus vaccine. Dev. Biol. Stand. 1977;39:289–293.
    1. Van Buynder PG, et al. Increased reactions to pediatric influenza vaccination following concomitant pneumococcal vaccination. Influenza Other Respir. Virus. 2013;7:184–190. doi: 10.1111/j.1750-2659.2012.00364.x.
    1. Govaert TM, et al. Adverse reactions to influenza vaccine in elderly people: randomised double blind placebo controlled trial. BMJ. 1993;307:988–990. doi: 10.1136/bmj.307.6910.988.
    1. Okada C, et al. Reactogenicity of trivalent inactivated influenza vaccine in young children: Pronounced reactions by previous successive vaccinations. Vaccine. 2015;33:3586–3591. doi: 10.1016/j.vaccine.2015.05.040.
    1. Zafack JG, et al. Risk of recurrence of adverse events following immunization: a systematic review. Pediatrics. 2017;140:e20163707. doi: 10.1542/peds.2016-3707.
    1. Herzog C. Influence of parenteral administration routes and additional factors on vaccine safety and immunogenicity: a review of recent literature. Expert Rev. Vaccin. 2014;13:399–415. doi: 10.1586/14760584.2014.883285.
    1. Frosner G, Steffen R, Herzog C. Virosomal hepatitis a vaccine: comparing intradermal and subcutaneous with intramuscular administration. J. Travel Med. 2009;16:413–419. doi: 10.1111/j.1708-8305.2009.00351.x.
    1. Beirne, P. V., et al. Needle size for vaccination procedures in children and adolescents. Cochrane Database Syst. Rev. CD010720 (2015). 10.1002/14651858.CD010720.pub2.
    1. Gillet Y, Habermehl P, Thomas S, Eymin C, Fiquet A. Immunogenicity and safety of concomitant administration of a measles, mumps and rubella vaccine (M-M-RvaxPro) and a varicella vaccine (VARIVAX) by intramuscular or subcutaneous routes at separate injection sites: a randomised clinical trial. BMC Med. 2009;7:16. doi: 10.1186/1741-7015-7-16.
    1. Gelinck LB, et al. Intradermal influenza vaccination in immunocompromized patients is immunogenic and feasible. Vaccine. 2009;27:2469–2474. doi: 10.1016/j.vaccine.2009.02.053.
    1. Riese P, Sakthivel P, Trittel S, Guzman CA. Intranasal formulations: promising strategy to deliver vaccines. Expert Opin. Drug Deliv. 2014;11:1619–1634. doi: 10.1517/17425247.2014.931936.
    1. FLUMIST® QUADRIVALENT. Prescribing Information. (2019).
    1. Shahid N, Daniell H. Plant-based oral vaccines against zoonotic and non-zoonotic diseases. Plant Biotechnol. J. 2016;14:2079–2099. doi: 10.1111/pbi.12604.
    1. Ita K. Transdermal delivery of vaccines—recent progress and critical issues. Biomed. Pharmacother. 2016;83:1080–1088. doi: 10.1016/j.biopha.2016.08.026.
    1. Taddio A, et al. Physical interventions and injection techniques for reducing injection pain during routine childhood immunizations: systematic review of randomized controlled trials and quasi-randomized controlled trials. Clin. Ther. 2009;31:S48–S76. doi: 10.1016/j.clinthera.2009.07.024.
    1. Taddio A, et al. A randomized trial of the effect of vaccine injection speed on acute pain in infants. Vaccine. 2016;34:4672–4677. doi: 10.1016/j.vaccine.2016.08.023.
    1. Vargas KM, Koil A, Dehority W. Recurrent sterile abscesses after immunization with aluminum-adjuvant based. Vaccin. Clin. Pediatr. (Phila.) 2018;57:733–737. doi: 10.1177/0009922817728702.
    1. Zijlstra E, Jahnke J, Fischer A, Kapitza C, Forst T. Impact of Injection speed, volume, and site on pain sensation. J. Diabetes Sci. Technol. 2018;12:163–168. doi: 10.1177/1932296817735121.
    1. Strugnell R, Zepp F, Cunningham AL, Tantawhichien T. Vaccine antigens. Underst. Mod. Vaccin.: Perspect. Vaccinol. 2011;1:61–88.
    1. Kanesa-thasan N, Shaw A, Stoddard JJ, Vernon TM. Ensuring the optimal safety of licensed vaccines: a perspective of the vaccine research, development, and manufacturing companies. Pediatrics. 2011;127:S16–S22. doi: 10.1542/peds.2010-1722D.
    1. Willame C, et al. Pain caused by measles, mumps, and rubella vaccines: a systematic literature review. Vaccine. 2017;35:5551–5558. doi: 10.1016/j.vaccine.2017.08.068.
    1. Lee, A. W., Saldutti, L. P., Wolfson, L. J., Stek, J. E. & Kuter, B. J. Response to Willame, et al., published in Vaccine 35 (2017) 5551-5558, entitled “Pain caused by measles, mumps, and rubella vaccines: a systematic literature review”. Vaccine 36, 6587–6588 (2018).
    1. Willame C, Lin L, Vetter V, Baril L, Praet N. Reply to Lee et al.‘s letter to the editor pertaining to our publication entitled “Pain caused by measles, mumps, and rubella vaccines: a systematic literature review”. Vaccine. 2018;36:6589–6590. doi: 10.1016/j.vaccine.2018.08.025.
    1. Nony P, et al. Impact of osmolality on burning sensations during and immediately after intramuscular injection of 0.5 ml of vaccine suspensions in healthy adults. Vaccine. 2001;19:3645–3651. doi: 10.1016/S0264-410X(01)00098-6.
    1. Vergara R, et al. Reduced-antigen-content-diphtheria-tetanus-acellular-pertussis and inactivated polio vaccine as a booster for adolescents 10 to 14 years of age. Eur. J. Pediatr. 2005;164:377–382. doi: 10.1007/s00431-005-1650-y.
    1. Garçon N, Vaughn DW, Didierlaurent AM. Development and evaluation of AS03, an Adjuvant System containing α-tocopherol and squalene in an oil-in-water emulsion. Expert Rev. Vaccin. 2012;11:349–366. doi: 10.1586/erv.11.192.
    1. Leroux-Roels G. Unmet needs in modern vaccinology: adjuvants to improve the immune response. Vaccine. 2010;28:C25–C36. doi: 10.1016/j.vaccine.2010.07.021.
    1. Baay M, Bollaerts K, Verstraeten T. A systematic review and meta-analysis on the safety of newly adjuvanted vaccines among older adults. Vaccine. 2018;36:4207–4214. doi: 10.1016/j.vaccine.2018.06.004.
    1. Leroux-Roels G, et al. Impact of adjuvants on CD4(+) T cell and B cell responses to a protein antigen vaccine: Results from a phase II, randomized, multicenter trial. Clin. Immunol. 2016;169:16–27. doi: 10.1016/j.clim.2016.05.007.
    1. Lecrenier N, et al. Development of a recombinant adjuvanted herpes zoster subunit vaccine and its implications for shingles prevention. Expert Rev. Vaccin. 2018;17:619–634. doi: 10.1080/14760584.2018.1495565.
    1. Cunningham AL, et al. Efficacy of the Herpes Zoster Subunit Vaccine in Adults 70 Years of Age or Older. N. Engl. J. Med. 2016;375:1019–1032. doi: 10.1056/NEJMoa1603800.
    1. Jacobsen SJ, et al. Observational safety study of febrile convulsion following first dose MMRV vaccination in a managed care setting. Vaccine. 2009;27:4656–4661. doi: 10.1016/j.vaccine.2009.05.056.
    1. Schink T, Holstiege J, Kowalzik F, Zepp F, Garbe E. Risk of febrile convulsions after MMRV vaccination in comparison to MMR or MMR+V vaccination. Vaccine. 2014;32:645–650. doi: 10.1016/j.vaccine.2013.12.011.
    1. World Health Organization. Reducing pain at the time of vaccination: WHO position paper, September 2015. Week Epidemiol. Rec. 2015;90:505–516.
    1. Taddio A, et al. Reducing pain during vaccine injections: clinical practice guideline. CMAJ. 2015;187:975–982. doi: 10.1503/cmaj.150391.
    1. Arane K, Behboudi A, Goldman RD. Virtual reality for pain and anxiety management in children. Can. Fam. Physician. 2017;63:932–934.
    1. Das RR, Panigrahi I, Naik SS. The effect of prophylactic antipyretic administration on post-vaccination adverse reactions and antibody response in children: a systematic review. PLoS ONE. 2014;9:e106629. doi: 10.1371/journal.pone.0106629.
    1. Saleh E, Moody MA, Walter EB. Effect of antipyretic analgesics on immune responses to vaccination. Hum. Vaccin. Immunother. 2016;12:2391–2402. doi: 10.1080/21645515.2016.1183077.
    1. Jackson ML, Bellamy A, Wolff M, Hill H, Jackson LA. Low-dose aspirin use does not diminish the immune response to monovalent H1N1 influenza vaccine in older adults. Epidemiol. Infect. 2016;144:768–771. doi: 10.1017/S0950268815002058.
    1. Gross PA, et al. Vaccine immune response and side effects with the use of acetaminophen with influenza vaccine. Clin. Diagn. Lab. Immunol. 1994;1:134–138.
    1. Doedee AM, et al. Effects of prophylactic and therapeutic paracetamol treatment during vaccination on hepatitis B antibody levels in adults: two open-label, randomized controlled trials. PLoS ONE. 2014;9:e98175. doi: 10.1371/journal.pone.0098175.
    1. Bexsero® (Meningococcal Group B Vaccine). Prescribing information. (2018).
    1. European Medicines Agency. Infanrix hexa product information. (2018).
    1. Larson HJ, et al. The State of Vaccine Confidence 2016: Global Insights Through a 67-Country Survey. EBioMedicine. 2016;12:295–301. doi: 10.1016/j.ebiom.2016.08.042.
    1. Karafillakis E, Larson HJ, ADVANCE consortium. The benefit of the doubt or doubts over benefits? A systematic literature review of perceived risks of vaccines in European populations. Vaccine. 2017;35:4840–4850. doi: 10.1016/j.vaccine.2017.07.061.
    1. Paterson P, et al. Vaccine hesitancy and healthcare providers. Vaccine. 2016;34:6700–6706. doi: 10.1016/j.vaccine.2016.10.042.
    1. Falup-Pecurariu O, et al. Effects of prophylactic ibuprofen and paracetamol administration on the immunogenicity and reactogenicity of the 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugated vaccine (PHiD-CV) co-administered with DTPa-combined vaccines in children: an open-label, randomized, controlled, non-inferiority trial. Hum. Vaccin. Immunother. 2017;13:649–660. doi: 10.1080/21645515.2016.1223001.
    1. Karambin MM, Heidarzadeh A, Sharghy R, Dalili S, Hashemian H. Effects of administering prophylactic acetaminophen on short-term complications of vaccination in 6-month-old infants. Int. J. Prev. Med. 2015;6:124. doi: 10.4103/2008-7802.172380.

Source: PubMed

3
Tilaa