A guide to vaccinology: from basic principles to new developments

Andrew J Pollard, Else M Bijker, Andrew J Pollard, Else M Bijker

Abstract

Immunization is a cornerstone of public health policy and is demonstrably highly cost-effective when used to protect child health. Although it could be argued that immunology has not thus far contributed much to vaccine development, in that most of the vaccines we use today were developed and tested empirically, it is clear that there are major challenges ahead to develop new vaccines for difficult-to-target pathogens, for which we urgently need a better understanding of protective immunity. Moreover, recognition of the huge potential and challenges for vaccines to control disease outbreaks and protect the older population, together with the availability of an array of new technologies, make it the perfect time for immunologists to be involved in designing the next generation of powerful immunogens. This Review provides an introductory overview of vaccines, immunization and related issues and thereby aims to inform a broad scientific audience about the underlying immunological concepts.

Conflict of interest statement

A.J.P. is Chair of the UK Department of Health and Social Care’s (DHSC) Joint Committee on Vaccination and Immunisation (JCVI), a member of the World Health Organization (WHO) Strategic Advisory Group of Experts on Immunization (SAGE) and a National Institute for Health Research (NIHR) Senior Investigator. The views expressed in this article do not necessarily represent the views of the DHSC, JCVI, NIHR or WHO. E.M.B. declares no competing interests. Oxford University has entered into a partnership with AstraZeneca for the development of a viral vectored coronavirus vaccine.

Figures

Fig. 1. The impact of vaccination on…
Fig. 1. The impact of vaccination on selected diseases in the UK.
The introduction of vaccination against infectious diseases such as diphtheria (part a), capsular group C meningococcus (part b), polio (part c), Haemophilus influenzae type B (part d), measles (part e) and pertussis (part f) led to a marked decrease in their incidence. Of note, the increase in reports of H. influenzae type B in 2001 led to a catch-up vaccination campaign, after which the incidence reduced. For pertussis, a decline in vaccine coverage led to an increase in cases in the late 1970s and 1980s, but disease incidence reduced again after vaccine coverage increased. Adapted with permission from the Green Book, information for public health professionals on immunisation, Public Health England, contains public sector information licensed under the Open Government Licence v3.0.
Fig. 2. Different types of vaccine.
Fig. 2. Different types of vaccine.
Schematic representation of different types of vaccine against pathogens; the text indicates against which pathogens certain vaccines are licensed and when each type of vaccine was first introduced. BCG, Mycobacterium bovis bacillus Calmette–Guérin.
Fig. 3. The generation of an immune…
Fig. 3. The generation of an immune response to a vaccine.
The immune response following immunization with a conventional protein antigen. The vaccine is injected into muscle and the protein antigen is taken up by dendritic cells, which are activated through pattern recognition receptors (PRRs) by danger signals in the adjuvant, and then trafficked to the draining lymph node. Here, the presentation of peptides of the vaccine protein antigen by MHC molecules on the dendritic cell activates T cells through their T cell receptor (TCR). In combination with signalling (by soluble antigen) through the B cell receptor (BCR), the T cells drive B cell development in the lymph node. Here, the T cell-dependent B cell development results in maturation of the antibody response to increase antibody affinity and induce different antibody isotypes. The production of short-lived plasma cells, which actively secrete antibodies specific for the vaccine protein, produces a rapid rise in serum antibody levels over the next 2 weeks. Memory B cells are also produced, which mediate immune memory. Long-lived plasma cells that can continue to produce antibodies for decades travel to reside in bone marrow niches. CD8+ memory T cells can proliferate rapidly when they encounter a pathogen, and CD8+ effector T cells are important for the elimination of infected cells.
Fig. 4. Immune memory is an important…
Fig. 4. Immune memory is an important feature of vaccine-induced protection.
Antibody levels in the circulation wane after primary vaccination, often to a level below that required for protection. Whether immune memory can protect against a future pathogen encounter depends on the incubation time of the infection, the quality of the memory response and the level of antibodies induced by memory B cells. a | The memory response may be sufficient to protect against disease if there is a long incubation period between pathogen exposure and the onset of symptoms to allow for the 3–4 days required for memory B cells to generate antibody titres above the protective threshold. b | The memory response may not be sufficient to protect against disease if the pathogen has a short incubation period and there is rapid onset of symptoms before antibody levels have reached the protective threshold. c | In some cases, antibody levels after primary vaccination remain above the protective threshold and can provide lifelong immunity.
Fig. 5. Herd immunity is an important…
Fig. 5. Herd immunity is an important feature of vaccine-induced protection.
The concept of herd immunity for a highly contagious disease such as measles. Susceptible individuals include those who have not yet been immunized (for example, being too young), those who cannot be immunized (for example, as a result of immunodeficiency), those for whom the vaccine did not induce immunity, those for whom initial vaccine-induced immunity has waned and those who refused immunization.
Fig. 6. Immune responses to polysaccharide and…
Fig. 6. Immune responses to polysaccharide and protein–polysaccharide conjugate vaccines.
a | Polysaccharide vaccines induce antibody-producing plasma cells by cross-linking the B cell receptor (BCR). However, affinity maturation of the antibody response and the induction of memory B cells do not occur. b | Protein–polysaccharide conjugate vaccines can engage T cells that recognize the carrier protein, as well as B cells that recognize the polysaccharide. T cells provide help to B cells, leading to affinity maturation and the production of both plasma cells and memory B cells. TCR, T cell receptor. Adapted from ref., Springer Nature Limited.

References

    1. World Health Organization. Global vaccine action plan 2011–2020. WHO (2013).
    1. World Health Organization. Child mortality and causes of death. WHO (2020).
    1. Hatherill M, White RG, Hawn TR. Clinical development of new TB vaccines: recent advances and next steps. Front. Microbiol. 2019;10:3154. doi: 10.3389/fmicb.2019.03154.
    1. Bekker LG, et al. The complex challenges of HIV vaccine development require renewed and expanded global commitment. Lancet. 2020;395:384–388. doi: 10.1016/S0140-6736(19)32682-0.
    1. Matz KM, Marzi A, Feldmann H. Ebola vaccine trials: progress in vaccine safety and immunogenicity. Expert Rev. Vaccines. 2019;18:1229–1242. doi: 10.1080/14760584.2019.1698952.
    1. Ahmed SF, Quadeer AA, McKay MR. Preliminary identification of potential vaccine targets for the COVID-19 coronavirus (SARS-CoV-2) based on SARS-CoV immunological studies. Viruses. 2020;12:254. doi: 10.3390/v12030254.
    1. Pawelec G. Age and immunity: what is “immunosenescence”? Exp. Gerontol. 2018;105:4–9. doi: 10.1016/j.exger.2017.10.024.
    1. Larson HJ. The state of vaccine confidence. Lancet. 2018;392:2244–2246. doi: 10.1016/S0140-6736(18)32608-4.
    1. Robbins JB, et al. Prevention of invasive bacterial diseases by immunization with polysaccharide–protein conjugates. Curr. Top. Microbiol. Immunol. 1989;146:169–180.
    1. Plotkin SA. Updates on immunologic correlates of vaccine-induced protection. Vaccine. 2020;38:2250–2257. doi: 10.1016/j.vaccine.2019.10.046.
    1. Rubin LG, et al. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin. Infect. Dis. 2014;58:e44–e100. doi: 10.1093/cid/cit684.
    1. Milligan R, Paul M, Richardson M, Neuberger A. Vaccines for preventing typhoid fever. Cochrane Database Syst. Rev. 2018;5:CD001261.
    1. WHO. Measles vaccines: WHO position paper — April 2017. Wkly. Epidemiol. Rec. 2017;92:205–227.
    1. Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-first century society. Nat. Rev. Immunol. 2011;11:865–872. doi: 10.1038/nri3085.
    1. Marrack P, McKee AS, Munks MW. Towards an understanding of the adjuvant action of aluminium. Nat. Rev. Immunol. 2009;9:287–293. doi: 10.1038/nri2510.
    1. Wilkins AL, et al. AS03- and MF59-adjuvanted influenza vaccines in children. Front. Immunol. 2017;8:1760. doi: 10.3389/fimmu.2017.01760.
    1. Kaslow DC, Biernaux S. RTS,S: toward a first landmark on the Malaria Vaccine Technology Roadmap. Vaccine. 2015;33:7425–7432. doi: 10.1016/j.vaccine.2015.09.061.
    1. Pedersen C, et al. Immunization of early adolescent females with human papillomavirus type 16 and 18 L1 virus-like particle vaccine containing AS04 adjuvant. J. Adolesc. Health. 2007;40:564–571. doi: 10.1016/j.jadohealth.2007.02.015.
    1. Mitkus RJ, Hess MA, Schwartz SL. Pharmacokinetic modeling as an approach to assessing the safety of residual formaldehyde in infant vaccines. Vaccine. 2013;31:2738–2743. doi: 10.1016/j.vaccine.2013.03.071.
    1. Eldred BE, Dean AJ, McGuire TM, Nash AL. Vaccine components and constituents: responding to consumer concerns. Med. J. Aust. 2006;184:170–175. doi: 10.5694/j.1326-5377.2006.tb00178.x.
    1. Fijen CA, Kuijper EJ, te Bulte MT, Daha MR, Dankert J. Assessment of complement deficiency in patients with meningococcal disease in The Netherlands. Clin. Infect. Dis. 1999;28:98–105. doi: 10.1086/515075.
    1. Wara DW. Host defense against Streptococcus pneumoniae: the role of the spleen. Rev. Infect. Dis. 1981;3:299–309. doi: 10.1093/clinids/3.2.299.
    1. Gershon AA, et al. Varicella zoster virus infection. Nat. Rev. Dis. Prim. 2015;1:15016. doi: 10.1038/nrdp.2015.16.
    1. Sandmann F, et al. Infant hospitalisations and fatalities averted by the maternal pertussis vaccination programme in England, 2012–2017: post-implementation economic evaluation. Clin. Infect. Dis. 2020;71:1984–1987. doi: 10.1093/cid/ciaa165.
    1. Demicheli V, Barale A, Rivetti A. Vaccines for women for preventing neonatal tetanus. Cochrane Database Syst. Rev. 2015;7:CD002959.
    1. Madhi SA, et al. Influenza vaccination of pregnant women and protection of their infants. N. Engl. J. Med. 2014;371:918–931. doi: 10.1056/NEJMoa1401480.
    1. Madhi SA, Dangor Z. Prospects for preventing infant invasive GBS disease through maternal vaccination. Vaccine. 2017;35:4457–4460. doi: 10.1016/j.vaccine.2017.02.025.
    1. Madhi SA, et al. Respiratory syncytial virus vaccination during pregnancy and effects in infants. N. Engl. J. Med. 2020;383:426–439. doi: 10.1056/NEJMoa1908380.
    1. Young MK, Cripps AW. Passive immunization for the public health control of communicable diseases: current status in four high-income countries and where to next. Hum. Vaccin. Immunother. 2013;9:1885–1893. doi: 10.4161/hv.25311.
    1. Patel M, Lee CK. Polysaccharide vaccines for preventing serogroup A meningococcal meningitis. Cochrane Database Syst. Rev. 2005;3:CD001093.
    1. Moberley S, Holden J, Tatham DP, Andrews RM. Vaccines for preventing pneumococcal infection in adults. Cochrane Database Syst. Rev. 2013;1:CD000422.
    1. Andrews NJ, et al. Serotype-specific effectiveness and correlates of protection for the 13-valent pneumococcal conjugate vaccine: a postlicensure indirect cohort study. Lancet Infect. Dis. 2014;14:839–846. doi: 10.1016/S1473-3099(14)70822-9.
    1. Borrow R, Abad R, Trotter C, van der Klis FR, Vazquez JA. Effectiveness of meningococcal serogroup C vaccine programmes. Vaccine. 2013;31:4477–4486. doi: 10.1016/j.vaccine.2013.07.083.
    1. Ramsay ME, McVernon J, Andrews NJ, Heath PT, Slack MP. Estimating haemophilus influenzae type b vaccine effectiveness in England and Wales by use of the screening method. J. Infect. Dis. 2003;188:481–485. doi: 10.1086/376997.
    1. Pollard AJ, Perrett KP, Beverley PC. Maintaining protection against invasive bacteria with protein-polysaccharide conjugate vaccines. Nat. Rev. Immunol. 2009;9:213–220. doi: 10.1038/nri2494.
    1. Darton TC, et al. Design, recruitment, and microbiological considerations in human challenge studies. Lancet Infect. Dis. 2015;15:840–851. doi: 10.1016/S1473-3099(15)00068-7.
    1. Suscovich TJ, et al. Mapping functional humoral correlates of protection against malaria challenge following RTS, S/AS01 vaccination. Sci. Transl Med. 2020;12:eabb4757. doi: 10.1126/scitranslmed.abb4757.
    1. Jin C, et al. Efficacy and immunogenicity of a Vi–tetanus toxoid conjugate vaccine in the prevention of typhoid fever using a controlled human infection model of Salmonella Typhi: a randomised controlled, phase 2b trial. Lancet. 2017;390:2472–2480. doi: 10.1016/S0140-6736(17)32149-9.
    1. Kourtis AP, Read JS, Jamieson DJ. Pregnancy and infection. N. Engl. J. Med. 2014;370:2211–2218. doi: 10.1056/NEJMra1213566.
    1. Malley R, et al. CD4+ T cells mediate antibody-independent acquired immunity to pneumococcal colonization. Proc. Natl Acad. Sci. USA. 2005;102:4848–4853. doi: 10.1073/pnas.0501254102.
    1. Henry B, Baclic O, & National Advisory Committee on Immunization (NACI). Summary of the NACI update on the recommended use of hepatitis B vaccine. Can. Commun. Dis. Rep. 2017;43:104–106. doi: 10.14745/ccdr.v43i05a04.
    1. Kelly DF, Pollard AJ, Moxon ER. Immunological memory: the role of B cells in long-term protection against invasive bacterial pathogens. JAMA. 2005;294:3019–3023. doi: 10.1001/jama.294.23.3019.
    1. McVernon J, Johnson PD, Pollard AJ, Slack MP, Moxon ER. Immunologic memory in Haemophilus influenzae type b conjugate vaccine failure. Arch. Dis. Child. 2003;88:379–383. doi: 10.1136/adc.88.5.379.
    1. McVernon J, et al. Immunologic memory with no detectable bactericidal antibody response to a first dose of meningococcal serogroup C conjugate vaccine at four years. Pediatr. Infect. Dis. J. 2003;22:659–661.
    1. World Health Organization Tetanus vaccines: WHO position paper, February 2017 — recommendations. Vaccine. 2018;36:3573–3575. doi: 10.1016/j.vaccine.2017.02.034.
    1. World Health Organization. Diphtheria vaccine: WHO position paper, August 2017 — recommendations. Vaccine. 2018;36:199–201. doi: 10.1016/j.vaccine.2017.08.024.
    1. Chen Z, He Q. Immune persistence after pertussis vaccination. Hum. Vaccin. Immunother. 2017;13:744–756. doi: 10.1080/21645515.2016.1259780.
    1. Burdin N, Handy LK, Plotkin SA. What is wrong with pertussis vaccine immunity? The problem of waning effectiveness of pertussis vaccines. Cold Spring Harb. Perspect. Biol. 2017;9:a029454. doi: 10.1101/cshperspect.a029454.
    1. WHO. Vaccines and vaccination against yellow fever: WHO Position Paper, June 2013 — recommendations. Vaccine. 2015;33:76–77. doi: 10.1016/j.vaccine.2014.05.040.
    1. Paunio M, et al. Twice vaccinated recipients are better protected against epidemic measles than are single dose recipients of measles containing vaccine. J. Epidemiol. Community Health. 1999;53:173–178. doi: 10.1136/jech.53.3.173.
    1. Zhu S, Zeng F, Xia L, He H, Zhang J. Incidence rate of breakthrough varicella observed in healthy children after 1 or 2 doses of varicella vaccine: results from a meta-analysis. Am. J. Infect. Control. 2018;46:e1–e7. doi: 10.1016/j.ajic.2017.07.029.
    1. Halstead SB, Rojanasuphot S, Sangkawibha N. Original antigenic sin in dengue. Am. J. Trop. Med. Hyg. 1983;32:154–156. doi: 10.4269/ajtmh.1983.32.154.
    1. Kim JH, Skountzou I, Compans R, Jacob J. Original antigenic sin responses to influenza viruses. J. Immunol. 2009;183:3294–3301. doi: 10.4049/jimmunol.0900398.
    1. Vatti A, et al. Original antigenic sin: a comprehensive review. J. Autoimmun. 2017;83:12–21. doi: 10.1016/j.jaut.2017.04.008.
    1. Statista Research Department. Herd immunity threshold for selected global diseases as of 2013. Statista (2013).
    1. Plans-Rubio P. The vaccination coverage required to establish herd immunity against influenza viruses. Prev. Med. 2012;55:72–77. doi: 10.1016/j.ypmed.2012.02.015.
    1. Trotter CL, Andrews NJ, Kaczmarski EB, Miller E, Ramsay ME. Effectiveness of meningococcal serogroup C conjugate vaccine 4 years after introduction. Lancet. 2004;364:365–367. doi: 10.1016/S0140-6736(04)16725-1.
    1. Trotter CL, Maiden MC. Meningococcal vaccines and herd immunity: lessons learned from serogroup C conjugate vaccination programs. Expert. Rev. Vaccines. 2009;8:851–861. doi: 10.1586/erv.09.48.
    1. Tabrizi SN, et al. Assessment of herd immunity and cross-protection after a human papillomavirus vaccination programme in Australia: a repeat cross-sectional study. Lancet Infect. Dis. 2014;14:958–966. doi: 10.1016/S1473-3099(14)70841-2.
    1. Brisson M, et al. Population-level impact, herd immunity, and elimination after human papillomavirus vaccination: a systematic review and meta-analysis of predictions from transmission-dynamic models. Lancet Public Health. 2016;1:e8–e17. doi: 10.1016/S2468-2667(16)30001-9.
    1. Trunz BB, Fine P, Dye C. Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet. 2006;367:1173–1180. doi: 10.1016/S0140-6736(06)68507-3.
    1. Barker, L. & Hussey, G. The Immunological Basis for Immunization Series: Module 5: Tuberculosis (World Health Organization, 2011).
    1. Eisenhut M, et al. BCG vaccination reduces risk of infection with Mycobacterium tuberculosis as detected by γ interferon release assay. Vaccine. 2009;27:6116–6120. doi: 10.1016/j.vaccine.2009.08.031.
    1. Verrall AJ, et al. Early clearance of Mycobacterium tuberculosis: the INFECT case contact cohort study in Indonesia. J. Infect. Dis. 2020;221:1351–1360.
    1. Pollard AJ, Finn A, Curtis N. Non-specific effects of vaccines: plausible and potentially important, but implications uncertain. Arch. Dis. Child. 2017;102:1077–1081. doi: 10.1136/archdischild-2015-310282.
    1. Higgins JP, et al. Association of BCG, DTP, and measles containing vaccines with childhood mortality: systematic review. BMJ. 2016;355:i5170. doi: 10.1136/bmj.i5170.
    1. Mina MJ, et al. Measles virus infection diminishes preexisting antibodies that offer protection from other pathogens. Science. 2019;366:599–606. doi: 10.1126/science.aay6485.
    1. Mina MJ, Metcalf CJ, de Swart RL, Osterhaus AD, Grenfell BT. Long-term measles-induced immunomodulation increases overall childhood infectious disease mortality. Science. 2015;348:694–699. doi: 10.1126/science.aaa3662.
    1. Falsey AR, Treanor JJ, Tornieporth N, Capellan J, Gorse GJ. Randomized, double-blind controlled phase 3 trial comparing the immunogenicity of high-dose and standard-dose influenza vaccine in adults 65 years of age and older. J. Infect. Dis. 2009;200:172–180. doi: 10.1086/599790.
    1. DiazGranados CA, et al. Efficacy of high-dose versus standard-dose influenza vaccine in older adults. N. Engl. J. Med. 2014;371:635–645. doi: 10.1056/NEJMoa1315727.
    1. Schnyder JL, et al. Fractional dose of intradermal compared to intramuscular and subcutaneous vaccination—a systematic review and meta-analysis. Travel. Med. Infect. Dis. 2020;37:101868. doi: 10.1016/j.tmaid.2020.101868.
    1. Voysey M, et al. The influence of maternally derived antibody and infant age at vaccination on infant vaccine responses: an individual participant meta-analysis. JAMA Pediatr. 2017;171:637–646. doi: 10.1001/jamapediatrics.2017.0638.
    1. Caceres VM, Strebel PM, Sutter RW. Factors determining prevalence of maternal antibody to measles virus throughout infancy: a review. Clin. Infect. Dis. 2000;31:110–119. doi: 10.1086/313926.
    1. Belnoue E, et al. APRIL is critical for plasmablast survival in the bone marrow and poorly expressed by early-life bone marrow stromal cells. Blood. 2008;111:2755–2764. doi: 10.1182/blood-2007-09-110858.
    1. Pace D, et al. Immunogenicity of reduced dose priming schedules of serogroup C meningococcal conjugate vaccine followed by booster at 12 months in infants: open label randomised controlled trial. BMJ. 2015;350:h1554. doi: 10.1136/bmj.h1554.
    1. Timens W, Boes A, Rozeboom-Uiterwijk T, Poppema S. Immaturity of the human splenic marginal zone in infancy. Possible contribution to the deficient infant immune response. J. Immunol. 1989;143:3200–3206.
    1. Peset Llopis MJ, Harms G, Hardonk MJ, Timens W. Human immune response to pneumococcal polysaccharides: complement-mediated localization preferentially on CD21-positive splenic marginal zone B cells and follicular dendritic cells. J. Allergy Clin. Immunol. 1996;97:1015–1024. doi: 10.1016/S0091-6749(96)80078-9.
    1. Claesson BA, et al. Protective levels of serum antibodies stimulated in infants by two injections of Haemophilus influenzae type b capsular polysaccharide–tetanus toxoid conjugate. J. Pediatr. 1989;114:97–100. doi: 10.1016/S0022-3476(89)80611-0.
    1. Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immun. Ageing. 2019;16:25. doi: 10.1186/s12979-019-0164-9.
    1. Nikolich-Žugich J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat. Rev. Immunol. 2008;8:512–522. doi: 10.1038/nri2318.
    1. Kadambari S, Klenerman P, Pollard AJ. Why the elderly appear to be more severely affected by COVID-19: the potential role of immunosenescence and CMV. Rev. Med. Virol. 2020;30:e2144. doi: 10.1002/rmv.2144.
    1. Domnich A, et al. Effectiveness of MF59-adjuvanted seasonal influenza vaccine in the elderly: a systematic review and meta-analysis. Vaccine. 2017;35:513–520. doi: 10.1016/j.vaccine.2016.12.011.
    1. Lal H, et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N. Engl. J. Med. 2015;372:2087–2096. doi: 10.1056/NEJMoa1501184.
    1. World Health Assembly. The Expanded Programme on Immunization: the 1974 resolution by the World Health Assembly. Assign. Child. 1985;69-72:87–88.
    1. Voysey M, Pollard AJ, Sadarangani M, Fanshawe TR. Prevalence and decay of maternal pneumococcal and meningococcal antibodies: a meta-analysis of type-specific decay rates. Vaccine. 2017;35:5850–5857. doi: 10.1016/j.vaccine.2017.09.002.
    1. Farrington P, et al. A new method for active surveillance of adverse events from diphtheria/tetanus/pertussis and measles/mumps/rubella vaccines. Lancet. 1995;345:567–569. doi: 10.1016/S0140-6736(95)90471-9.
    1. Pinto MV, Bihari S, Snape MD. Immunisation of the immunocompromised child. J. Infect. 2016;72(Suppl):S13–S22. doi: 10.1016/j.jinf.2016.04.017.
    1. Seligman SJ. Risk groups for yellow fever vaccine-associated viscerotropic disease (YEL-AVD) Vaccine. 2014;32:5769–5775. doi: 10.1016/j.vaccine.2014.08.051.
    1. Gellin BG, Maibach EW, Marcuse EK. Do parents understand immunizations? A national telephone survey. Pediatrics. 2000;106:1097–1102. doi: 10.1542/peds.106.5.1097.
    1. Offit PA, et al. Addressing parents’ concerns: do multiple vaccines overwhelm or weaken the infant’s immune system? Pediatrics. 2002;109:124–129. doi: 10.1542/peds.109.1.124.
    1. Centers for Disease Control and Prevention. Multiple vaccinations at once. CDC (2020).
    1. Stowe J, Andrews N, Taylor B, Miller E. No evidence of an increase of bacterial and viral infections following measles, mumps and rubella vaccine. Vaccine. 2009;27:1422–1425. doi: 10.1016/j.vaccine.2008.12.038.
    1. Otto S, et al. General non-specific morbidity is reduced after vaccination within the third month of life — the Greifswald study. J. Infect. 2000;41:172–175. doi: 10.1053/jinf.2000.0718.
    1. Griffin MR, Taylor JA, Daugherty JR, Ray WA. No increased risk for invasive bacterial infection found following diphtheria–tetanus–pertussis immunization. Pediatrics. 1992;89:640–642. doi: 10.1542/peds.89.4.640.
    1. Aaby P, et al. Non-specific beneficial effect of measles immunisation: analysis of mortality studies from developing countries. BMJ. 1995;311:481–485. doi: 10.1136/bmj.311.7003.481.
    1. Glanz JM, et al. Association between estimated cumulative vaccine antigen exposure through the first 23 months of life and non-vaccine-targeted infections from 24 through 47 months of age. JAMA. 2018;319:906–913. doi: 10.1001/jama.2018.0708.
    1. Bohlke K, et al. Risk of anaphylaxis after vaccination of children and adolescents. Pediatrics. 2003;112:815–820. doi: 10.1542/peds.112.4.815.
    1. Nohynek H, et al. AS03 adjuvanted AH1N1 vaccine associated with an abrupt increase in the incidence of childhood narcolepsy in Finland. PLoS ONE. 2012;7:e33536. doi: 10.1371/journal.pone.0033536.
    1. Miller E, et al. Risk of narcolepsy in children and young people receiving AS03 adjuvanted pandemic A/H1N1 2009 influenza vaccine: retrospective analysis. BMJ. 2013;346:f794. doi: 10.1136/bmj.f794.
    1. Hallberg P, et al. Pandemrix-induced narcolepsy is associated with genes related to immunity and neuronal survival. EBioMedicine. 2019;40:595–604. doi: 10.1016/j.ebiom.2019.01.041.
    1. DeStefano F, Shimabukuro TT. The MMR vaccine and autism. Annu. Rev. Virol. 2019;6:585–600. doi: 10.1146/annurev-virology-092818-015515.
    1. DeStefano F, Bodenstab HM, Offit PA. Principal controversies in vaccine safety in the United States. Clin. Infect. Dis. 2019;69:726–731. doi: 10.1093/cid/ciz135.
    1. Moro PL, Haber P, McNeil MM. Challenges in evaluating post-licensure vaccine safety: observations from the Centers for Disease Control and Prevention. Expert Rev. Vaccines. 2019;18:1091–1101. doi: 10.1080/14760584.2019.1676154.
    1. Peck M, et al. Global routine vaccination coverage, 2018. MMWR Morb. Mortal. Wkly. Rep. 2019;68:937–942. doi: 10.15585/mmwr.mm6842a1.
    1. World Health Organization. Immunization coverage. WHO (2020).
    1. World Health Organization. More than 9.4 million children vaccinated against typhoid fever in Sindh. WHO (2019).
    1. World Health Organization. More than 140,000 die from measles as cases surge worldwide. WHO (2019).
    1. World Health Organization. Disease outbreaks. WHO (2020).
    1. Rerks-Ngarm S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 2009;361:2209–2220. doi: 10.1056/NEJMoa0908492.
    1. Fauci AS, Marovich MA, Dieffenbach CW, Hunter E, Buchbinder SP. Immunology. Immune activation with HIV vaccines. Science. 2014;344:49–51. doi: 10.1126/science.1250672.
    1. Agnandji ST, et al. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N. Engl. J. Med. 2012;367:2284–2295. doi: 10.1056/NEJMoa1208394.
    1. Killeen GF, et al. Developing an expanded vector control toolbox for malaria elimination. BMJ Glob. Health. 2017;2:e000211. doi: 10.1136/bmjgh-2016-000211.
    1. Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect. Dis. 2012;12:36–44. doi: 10.1016/S1473-3099(11)70295-X.
    1. Skowronski DM, et al. Low 2012–13 influenza vaccine effectiveness associated with mutation in the egg-adapted H3N2 vaccine strain not antigenic drift in circulating viruses. PLoS ONE. 2014;9:e92153. doi: 10.1371/journal.pone.0092153.
    1. Raymond DD, et al. Conserved epitope on influenza-virus hemagglutinin head defined by a vaccine-induced antibody. Proc. Natl Acad. Sci. USA. 2018;115:168–173. doi: 10.1073/pnas.1715471115.
    1. Tameris MD, et al. Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previously vaccinated with BCG: a randomised, placebo-controlled phase 2b trial. Lancet. 2013;381:1021–1028. doi: 10.1016/S0140-6736(13)60177-4.
    1. Tait DR, et al. Final analysis of a trial of M72/AS01(E) vaccine to prevent tuberculosis. N. Engl. J. Med. 2019;381:2429–2439. doi: 10.1056/NEJMoa1909953.
    1. Kobayashi M, et al. WHO consultation on group B streptococcus vaccine development: report from a meeting held on 27–28 April 2016. Vaccine. 2019;37:7307–7314. doi: 10.1016/j.vaccine.2016.12.029.
    1. Inoue N, Abe M, Kobayashi R, Yamada S. Vaccine development for cytomegalovirus. Adv. Exp. Med. Biol. 2018;1045:271–296. doi: 10.1007/978-981-10-7230-7_13.
    1. Schleiss MR, Permar SR, Plotkin SA. Progress toward development of a vaccine against congenital cytomegalovirus infection. Clin. Vaccine Immunol. 2017;24:e00268–e00317.
    1. World Health Organization. Ageing and health. WHO (2018).
    1. Rauch S, Jasny E, Schmidt KE, Petsch B. New vaccine technologies to combat outbreak situations. Front. Immunol. 2018;9:1963. doi: 10.3389/fimmu.2018.01963.
    1. Jeyanathan M, et al. Immunological considerations for COVID-19 vaccine strategies. Nat. Rev. Immunol. 2020;20:615–632. doi: 10.1038/s41577-020-00434-6.
    1. Koff WC, Schenkelberg T. The future of vaccine development. Vaccine. 2020;38:4485–4486. doi: 10.1016/j.vaccine.2019.07.101.
    1. van Riel D, de Wit E. Next-generation vaccine platforms for COVID-19. Nat. Mater. 2020;19:810–812. doi: 10.1038/s41563-020-0746-0.
    1. Rollier CS, Reyes-Sandoval A, Cottingham MG, Ewer K, Hill AV. Viral vectors as vaccine platforms: deployment in sight. Curr. Opin. Immunol. 2011;23:377–382. doi: 10.1016/j.coi.2011.03.006.
    1. Corbett KS, et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature. 2020;586:567–571. doi: 10.1038/s41586-020-2622-0.
    1. Polack, F. P. et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N. Engl. J. Med.10.1056/NEJMoa2034577 (2020).
    1. Wallis J, Shenton DP, Carlisle RC. Novel approaches for the design, delivery and administration of vaccine technologies. Clin. Exp. Immunol. 2019;196:189–204. doi: 10.1111/cei.13287.
    1. Zhang C, Maruggi G, Shan H, Li J. Advances in mRNA vaccines for infectious diseases. Front. Immunol. 2019;10:594. doi: 10.3389/fimmu.2019.00594.
    1. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines — a new era in vaccinology. Nat. Rev. Drug Discov. 2018;17:261–279. doi: 10.1038/nrd.2017.243.
    1. Crank MC, et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science. 2019;365:505–509. doi: 10.1126/science.aav9033.
    1. Mascola JR, Fauci AS. Novel vaccine technologies for the 21st century. Nat. Rev. Immunol. 2020;20:87–88. doi: 10.1038/s41577-019-0243-3.
    1. Kanekiyo M, Ellis D, King NP. New vaccine design and delivery technologies. J. Infect. Dis. 2019;219:S88–S96. doi: 10.1093/infdis/jiy745.
    1. Peyraud N, et al. Potential use of microarray patches for vaccine delivery in low- and middle-income countries. Vaccine. 2019;37:4427–4434. doi: 10.1016/j.vaccine.2019.03.035.
    1. Rouphael NG, et al. The safety, immunogenicity, and acceptability of inactivated influenza vaccine delivered by microneedle patch (TIV-MNP 2015): a randomised, partly blinded, placebo-controlled, phase 1 trial. Lancet. 2017;390:649–658. doi: 10.1016/S0140-6736(17)30575-5.
    1. Davenport RJ, Satchell M, Shaw-Taylor LMW. The geography of smallpox in England before vaccination: a conundrum resolved. Soc. Sci. Med. 2018;206:75–85. doi: 10.1016/j.socscimed.2018.04.019.

Source: PubMed

3
Se inscrever