277
Views
3
CrossRef citations to date
0
Altmetric
Review

Understanding host immune responses to pneumococcal proteins in the upper respiratory tract to develop serotype-independent pneumococcal vaccines

, , ORCID Icon &
Pages 959-972 | Received 02 Jun 2020, Accepted 26 Oct 2020, Published online: 08 Nov 2020

References

  • Ciapponi A, Elorriaga N, Rojas JI, et al. Epidemiology of pediatric pneumococcal meningitis and bacteremia in Latin America and the caribbean: A systematic review and meta-analysis. Pediatr Infect Dis J. 2014;33(9):971–978.
  • O’Brien KL, Wolfson LJ, Watt JP, et al. Burden of disease caused by streptococcus pneumoniae in children younger than 5 years: global estimates. Lancet. 2009;374:893–902.
  • Mook-Kanamori BB, Geldhoff M, van der Poll T, et al. Pathogenesis and pathophysiology of pneumococcal meningitis. Clin Microbiol Rev. 2011;24(3):557–591.
  • Aligholi M, Emaneini M, Jabalameli F, et al. Antibiotic susceptibility pattern of gram-positive cocci cultured from patients in three university hospitals in Tehran, Iran during 2001–2005. Acta Med. Iran. 2009;47:329–334.
  • Bentley SD, Aanensen DM, Mavroidi A, et al. Genetic analysis of the capsular biosynthetic locus from all 90 pneumococcal serotypes. PLoS Genet. 2006;2(3):262–269.
  • Hausdorff WP, Bryant J, Paradiso PR, et al. Which pneumococcal serogroups cause the most invasive disease: implications for conjugate vaccine formulation and use, part I. Clin Infect Dis. 2000;30(1):100–121.
  • Lijek RS, Weiser JN. Co-infection subverts mucosal immunity in the upper respiratory tract. Curr Opin Immunol. 2012;24(4):417–423.
  • Simell B, Auranen K, Käyhty H, et al. The fundamental link between pneumococcal carriage and disease. Expert Rev Vaccines. 2012;11(7):841–855.
  • Poolman JT, Peeters CC, Van Den Dobbelsteen GP. The history of pneumococcal conjugate vaccine development: dose selection. Expert Rev Vaccines. 2013;12(12):1379–1394.
  • Mitsi E, Roche AM, Reiné J, et al. Agglutination by anti-capsular polysaccharide antibody is associated with protection against experimental human pneumococcal carriage. Mucosal Immunol. 2017;10(2):385–394.
  • McFetridge R, Meulen AS, Folkerth SD, et al. Safety, tolerability, and immunogenicity of 15-valent pneumococcal conjugate vaccine in healthy adults. Vaccine. 2015;33(24):2793–2799.
  • Sobanjo-ter Meulen A, Vesikari T, Malacaman EA, et al. Safety, tolerability and immunogenicity of 15-valent pneumococcal conjugate vaccine in toddlers previously vaccinated with 7-valent pneumococcal conjugate vaccine. J. Pediatr. Infect. Dis. 2015;34(2):186–194.
  • Tai SS. Streptococcus pneumoniae protein vaccine candidates: properties, activities and animal studies. Crit. Rev. Microbiol. 2006;32(3):139–153.
  • Lagousi T, Basdeki P, Routsias J, et al. Novel protein-based pneumococcal vaccines: assessing the use of distinct protein fragments instead of full-length proteins as vaccine antigens. Vaccines (Basel). 2019;7(1):9.
  • Bruyn GAW, Zegers BJM, van Furth R. Mechanisms of host defense against infection with Streptococcus pneumoniae. Clin Infect Dis. 1992;14(1):251–262.
  • Zhang Q, Bernatoniene J, Bagrade L, et al. Regulation of production of mucosal antibody to pneumococcal protein antigens by T-cell-derived gamma interferon and interleukin-10 in children. Infect Immun. 2006;74(8):4735–4743.
  • Mureithi MW, Finn A, Ota MO, et al. T cell memory response to pneumococcal protein antigens in an area of high pneumococcal carriage and disease. J Infect Dis. 2009;200(5):783–793.
  • McAleer JP, Kolls JK. Directing traffic: IL-17 and IL-22 coordinate pulmonary immune defense. Immunol Rev. 2014;260(1):129–144.
  • Darrieux M, Moreno AT, Ferreira DM, et al. Recognition of pneumococcal isolates by antisera raised against PspA fragments from different clades. J Med Microbiol. 2008;57:273–278.
  • Mccool TL, Cate TR, Moy G, et al. The immune response to pneumococcal proteins during experimental human carriage. J Exp Med. 2002;195(3):359–365.
  • Ferreira DM, Neill DR, Bangert M, et al. Controlled human infection and rechallenge with Streptococcus pneumoniae reveals the protective efficacy of carriage in healthy adults. Am J Respir Crit Care Med. 2013;187(8):855–864.
  • Wright AKA, Bangert M, Gritzfeld JF, et al. Experimental human pneumococcal carriage augments IL-17A-dependent T-cell defence of the lung. PLoS Pathog. 2013;9:3.
  • Gilks CF, Ojoo SA, Ojoo JC, et al. Invasive pneumococcal disease in a cohort of predominantly HIV-1 infected female sex-workers in Nairobi, Kenya. Lancet. 1996;347(9003):718–723.
  • Weiser JN, Ferreira DM, Paton JC. Streptococcus pneumoniae: transmission, colonization and invasion. Nat Rev Microbiol. 2018;16(6):355–367.
  • Syrjänen RK, Kilpi TM, Kaijalainen TH, Herva EE, Takala AK. Nasopharyngeal Carriage of Streptococcus pneumoniae in Finnish Children Younger Than 2 Years Old. J Infect Dis. 2001;184:451-9.
  • Dunkelberger JR, Song W-C. Complement and its role in innate and adaptive immune responses. Cell Res. 2010;20:34–50.
  • Roche AM, Richard AL, Rahkola JT, et al. Antibody blocks acquisition of bacterial colonization through agglutination. Mucosal Immunol. 2015;8(1):176–185.
  • Hall-Stoodley L, Nistico L, Sambanthamoorthy K, et al. Characterization of biofilm matrix, degradation by DNase treatment and evidence of capsule downregulation in streptococcus pneumoniae clinical isolates. BMC Microbiol. 2008;8:173.
  • Kim JO, Weiser JN. Association of intrastrain phase variation in quantity of capsular polysaccharide and teichoic acid with the virulence of streptococcus pneumoniae. J Infect Dis. 1998;177(2):368–377.
  • Habets MN, Van Selm S, Van Der Gaast De Jongh CE, et al. A novel flow cytometry-based assay for the quantification of antibody-dependent pneumococcal agglutination. PLoS One. 2017;12:3.
  • Khan MN, Sharma SK, Filkins LM, et al. PcpA of streptococcus pneumoniae mediates adherence to nasopharyngeal and lung epithelial cells and elicits functional antibodies in humans. Microbes Infect. 2012;14(12):1102–1110.
  • Khan MN, Pichichero ME. Vaccine candidates PhtD and PhtE of Streptococcus pneumoniae are adhesins that elicit functional antibodies in humans. Vaccine. 2012;30(18):2900–2907.
  • Papastamatiou T, Routsias JG, Koutsoni O, et al. Evaluation of protective efficacy of selected immunodominant B-cell epitopes within virulent surface proteins of streptococcus pneumoniae. Infect Immun. 2018;86:3.
  • Weiser JN, Austrian R, Sreenivasan PK, et al. Phase variation in pneumococcal opacity: relationship between colonial morphology and nasopharyngeal colonization. Infect Immun. 1994;62(6):2582–2589.
  • Alper CA, Johnson AM, Birtch AG, et al. Human C’3: evidence for the liver as the primary site of synthesis. Science. 1969;163(3864):286–288.
  • Lubbers R, van Essen MF, van Kooten C, et al. Production of complement components by cells of the immune system. Clin Exp Immunol. 2017;188(2):183–194.
  • Glennie S, Gritzfeld JF, Pennington SH, et al. Modulation of nasopharyngeal innate defenses by viral coinfection predisposes individuals to experimental pneumococcal carriage. Mucosal Immunol. 2016;9:56–67.
  • Hammerschmidt S, Agarwal V, Kunert A, et al. The host immune regulator factor H interacts via two contact sites with the PspC protein of streptococcus pneumoniae and mediates adhesion to host epithelial cells. J Immunol. 2007;178(9):5848–5858.
  • Bogaert D, De Groot R, Hermans PWM. Streptococcus pneumoniae colonisation: the key to pneumococcal disease. Lancet Infect Dis. 2004;4(3):144–154.
  • Rajam G, Anderton JM, Carlone GM, et al. Pneumococcal surface adhesin A (PsaA): A review. Crit. Rev. Microbiol. 2008;34(3–4):131–142.
  • Rosenow C, Ryan P, Weiser JN, et al. Contribution of novel choline-binding proteins to adherence, colonization and immunogenicity of streptococcus pneumoniae. Mol Microbiol. 1997;25(5):819–829.
  • Pracht D, Elm C, Gerber J, et al. PavA of Streptococcus pneumoniae modulates adherence, invasion, and meningeal inflammation. Infect Immun. 2005;73(5):2680–2689.
  • Rose L, Shivshankar P, Hinojosa E, et al. Antibodies against PsrP, a novel streptococcus pneumoniae adhesin, block adhesion and protect mice against pneumococcal challenge. J Infect Dis. 2008;198(3):375–383.
  • Rosenow C, Ryan P, Weiser JN, et al. Contribution of novel choline-binding proteins to adherence, colonization and immunogenicity of streptococcus pneumoniae. Mol Microbiol. 1997;25(5):819–829.
  • Romero-Steiner S, Pilishvili T, Sampson JS, et al. Inhibition of pneumococcal adherence to human nasopharyngeal epithelial cells by anti-PsaA antibodies. Clin Diagn Lab Immunol. 2003;10(2):246–251.
  • Romero-Steiner S, Caba J, Rajam G, et al. Adherence of recombinant pneumococcal surface adhesin A (rPsaA)-coated particles to human nasopharyngeal epithelial cells for the evaluation of anti-PsaA functional antibodies. Vaccine. 2006;24:3224–3231.
  • Rajam G, Phillips DJ, White E, et al. A functional epitope of the pneumococcal surface adhesin A activates nasopharyngeal cells and increases bacterial internalization. Microb Pathog. 2008;44(3):186–196.
  • Pichichero ME, Kaur R, Casey JR, et al. Antibody response to streptococcus pneumoniae proteins PhtD, LytB, PcpA, PhtE and Ply after nasopharyngeal colonization and acute otitis media in children. Hum Vaccin Immunother. 2012;8(6):799–805.
  • Kaur R, Surendran N, Ochs M, et al. Human antibodies to PhtD, PcpA, and ply reduce adherence to human lung epithelial cells and murine nasopharyngeal colonization by Streptococcus pneumoniae. Infect Immun. 2014;82(12):5069–5075.
  • Johnston JW, Briles DE, Myers LE, et al. Mn2+-dependent regulation of multiple genes in streptococcus pneumoniae through PsaR and the resultant impact on virulence. Infect Immun. 2006;74(2):1171–1180.
  • Kaur R, Casey JR, Pichichero ME. Serum antibody response to five streptococcus pneumoniae proteins during acute otitis media in otitis-prone and non-otitis-prone children. Pediatr Infect. Dis J. 2011;30(8):645–650.
  • Shak JR, Ludewick HP, Howery KE, et al. Novel role for the streptococcus pneumoniae toxin pneumolysin in the assembly of biofilms. MBio. 2013;4:5.
  • Rubins JB, Paddock AH, Charboneau D, et al. Pneumolysin in pneumococcal adherence and colonization. Microb Pathog. 1998;25(6):337–342.
  • Neill DR, Coward WR, Gritzfeld JF, et al. Density and duration of pneumococcal carriage is maintained by transforming growth factor β1 and T regulatory cells. Am J Respir Crit Care Med. 2014;189(10):1250–1259.
  • Honsa ES, Johnson MDL, Rosch JW. The roles of transition metals in the physiology and pathogenesis of streptococcus pneumoniae. Front Cell Infect Microbiol. 2013;3:92.
  • Ogunniyi AD, Grabowicz M, Mahdi LK, et al. Pneumococcal histidine triad proteins are regulated by the Zn 2+ ‐dependent repressor AdcR and inhibit complement deposition through the recruitment of complement factor H. Faseb J. 2009;23:731–738.
  • Shafeeq S, Kuipers OP, Kloosterman TG. The role of zinc in the interplay between pathogenic streptococci and their hosts. Mol Microbiol. 2013;88(6):1047–1057.
  • Riboldi-Tunnicliffe A, Isaacs NW, Mitchell TJ. 1.2 Å crystal structure of the S. pneumoniae PhtA histidine triad domain a novel zinc binding fold. FEBS Lett. 2005;579(24):5353–5360.
  • Lagousi T, Routsias J, Piperi C, et al. Discovery of immunodominant B cell epitopes within surface pneumococcal virulence proteins in pediatric patients with invasive pneumococcal disease. J Biol Chem. 2015;290(46):27500–27510.
  • Godfroid F, Hermand P, Verlant V, et al. Preclinical evaluation of the Pht proteins as potential cross-protective pneumococcal vaccine antigens. Infect Immun. 2011;79:238–245.
  • Rioux S, Neyt C, Di Paolo E, et al. Transcriptional regulation, occurrence and putative role of the Pht family of streptococcus pneumoniae. Microbiology. 2011;157:335–348.
  • Kallio A, Sepponen K, Hermand P, et al. Role of Pht proteins in attachment of streptococcus pneumoniae to respiratory epithelial cells. Infect Immun. 2014;82:1683–1691.
  • Routsias JG, Kosmopoulou A, Makri A, et al. Zinc ion dependent B-cell epitope, associated with primary sjogren’s syndrome, resides within the putative zinc finger domain of Ro60kD autoantigen: physical and immunologic properties. J Med Chem. 2004;47(17):4327–4334.
  • Briles DE, Ades E, Paton JC, et al. Intranasal immunization of mice with a mixture of the pneumococcal proteins PsaA and PspA is highly protective against nasopharyngeal carriage of streptococcus pneumoniae. Infect Immun. 2000;68:796–800.
  • Voß F, Kohler TP, Meyer T, et al. Intranasal vaccination with lipoproteins confers protection against pneumococcal colonisation. Front Immunol. 2018;9:2405.
  • McCool TL, Weiser JN. Limited role of antibody in clearance of streptococcus pneumoniae in a murine model of colonization. Infect Immun. 2004;72(10):5807–5813.
  • Trzciński K, Thompson C, Malley R, et al. Antibodies to conserved pneumococcal antigens correlate with, but are not required for, protection against pneumococcal colonization induced by prior exposure in a mouse model. Infect Immun. 2005;73(10):7043–7046.
  • Converso TR, Goulart C, Darrieux M, et al. A protein chimera including PspA in fusion with PotD is protective against invasive pneumococcal infection and reduces nasopharyngeal colonization in mice. Vaccine. 2017;35(38):5140–5147.
  • Whaley MJ, Sampson JS, Johnson SE, et al. Concomitant administration of recombinant PsaA and PCV7 reduces streptococcus pneumoniae serotype 19A colonization in a murine model. Vaccine. 2010;28(18):3071–3075.
  • Dhoubhadel BG, Yasunami M, Nguyen HAT, et al. Bacterial load of pneumococcal serotypes correlates with their prevalence and multiple serotypes is associated with acute respiratory infections among children less than 5 years of age. PLoS One. 2014;9:10.
  • Brotons P, Bassat Q, Lanaspa M, et al. Nasopharyngeal bacterial load as a marker for rapid and easy diagnosis of invasive pneumococcal disease in children from Mozambique. PLoS One. 2017;12:9.
  • Pichichero ME. Ten-year study of the stringently defined otitis-prone child in Rochester, NY. Pediatr Infect Dis J. 2016;35(9):1033–1039.
  • Azarian T, Grant LR, Georgieva M, et al. Association of pneumococcal protein antigen serology with age and antigenic profile of colonizing isolates. J Infect Dis. 2017;215(5):713–722.
  • Mendy AL, Agbla SC, Odutola AA, et al. Kinetics of antibodies against pneumococcal proteins and their relationship to nasopharyngeal carriage in the first two months of life. PLoS One. 2017;12:10.
  • Prevaes SMPJ, van Wamel WJB, de Vogel CP, et al. Nasopharyngeal colonization elicits antibody responses to staphylococcal and pneumococcal proteins that are not associated with a reduced risk of subsequent carriage. Infect Immun. 2012;80::2186–2193.
  • Pennington SH, Pojar S, Mitsi E, et al. Polysaccharide-specific memory B cells predict protection against experimental human pneumococcal carriage. Am J Respir Crit Care Med. 2016;194:1523–1531.
  • Odutola A, Ota MOC, Antonio M, et al. Efficacy of a novel, protein-based pneumococcal vaccine against nasopharyngeal carriage of streptococcus pneumoniae in infants: A phase 2, randomized, controlled, observer-blind study. Vaccine. 2017;35:2531–2542.
  • Xu Q, Casey JR, Almudevar A, et al. Correlation of higher antibody levels to pneumococcal proteins with protection from pneumococcal acute otitis media but not protection from nasopharyngeal colonization in young children. Clin Microbiol Infect. 2017;23:7.
  • Andrews NJ, Waight PA, Burbidge P, 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.
  • Lu YJ, Gross J, Bogaert D, et al. Interleukin-17A mediates acquired immunity to pneumococcal colonization. PLoS Pathog. 2008;4:9.
  • Zhang Z, Clarke TB, Weiser JN. Cellular effectors mediating Th17-dependent clearance of pneumococcal colonization in mice. J Clin Invest. 2009;119:1899.
  • Davis KM, Nakamura S, Weiser JN. Nod2 sensing of lysozyme-digested peptidoglycan promotes macrophage recruitment and clearance of S. pneumoniae colonization in mice. J Clin Invest. 2011;121:3666–3676.
  • Ronchetti R, Villa MP, Martella S, et al. Nasal cellularity in 183 unselected schoolchildren aged 9 to 11 years. Pediatrics. 2002;110:1137-42.
  • Jochems SP, Piddock K, Rylance J, et al. Novel analysis of immune cells from nasal microbiopsy demonstrates reliable, reproducible data for immune populations, and superior cytokine detection compared to nasal wash. PLoS One. 2017;12:e0169805.
  • Van Rossum AMC, Lysenko ES, Weiser JN. Host and bacterial factors contributing to the clearance of colonization by Streptococcus pneumoniae in a murine model. Infect Immun. 2005;73:7718–7726.
  • Gwaltney JM, Sande MA, Austrian R, et al. Spread of streptococcus pneumoniae in families. II. relation of transfer of S. pneumoniae to incidence of colds and serum antibody. J Infect Dis. 1975;132:1.
  • Zhang Q, Arnaoutakis K, Murdoch C, et al. Mucosal immune responses to capsular pneumococcal polysaccharides in immunized preschool children and controls with similar nasal pneumococcal colonization rates. Pediatr Infect Dis J. 2004;23(4):307–313.
  • Simell B, Kilpi TM, Käyhty H. Pneumococcal carriage and otitis media induce salivary antibodies to pneumococcal capsular polysaccharides in children. J Infect Dis. 2002;186(8):1106–1114.
  • Malley R, Srivastava A, Lipsitch M, et al. Antibody-independent, interleukin-17A-mediated, cross-serotype immunity to pneumococci in mice immunized intranasally with the cell wall polysaccharide. Infect Immun. 2006;74:2187–2195.
  • Basset A, Thompson CM, Hollingshead SK, et al. Antibody-independent, CD4+ T-cell-dependent protection against pneumococcal colonization elicited by intranasal immunization with purified pneumococcal proteins. Infect Immun. 2007;75:5460–5464.
  • Malley R, Trzcinski K, Srivastava A, et al. CD4+ T cells mediate antibody-independent acquired immunity to pneumococcal colonization. Proc Natl Acad Sci. 2005;102(13):4848–4853.
  • Moens L, Hermand P, Wellens T, et al. Identification of SP1683 as a pneumococcal protein that is protective against nasopharyngeal colonization. Hum.Vaccin Immunother. 2018;14::1234–1242.
  • Zhang Q, Leong SC, McNamara PS, et al. Characterisation of regulatory t cells in nasal associated lymphoid tissue in children: relationships with pneumococcal colonization. PLoS Pathog. 2011;7:8.
  • Pido-Lopez J, Kwok WW, Mitchell TJ, et al. Acquisition of pneumococci specific effector and regulatory Cd4 + T cells localising within human upper respiratory-tract mucosal lymphoid tissue. PLoS Pathog. 2011;7:12.
  • Mubarak A, Ahmed MS, Upile N, et al. A dynamic relationship between mucosal T helper type 17 and regulatory T-cell populations in nasopharynx evolves with age and associates with the clearance of pneumococcal carriage in humans. Clin Microbiol Infect. 2016;22:8.
  • Jiang XL, Zhang GL, Yang T, et al. Association of pneumococcal carriage and expression of Foxp3+ regulatory T cells and Th17 cells in the adenoids of children. Respiration. 2015;90:25–32.
  • Gray C, Ahmed MS, Mubarak A, et al. Activation of memory Th17 cells by domain 4 pneumolysin in human nasopharynx-associated lymphoid tissue and its association with pneumococcal carriage. Mucosal Immunol. 2014;7:705–717.
  • Vuononvirta J, Peltola V, Ilonen J, et al. The gene polymorphism of IL-17 G-152A is associated with increased colonization of streptococcus pneumoniae in young finnish children. Pediatr Infect Dis J. 2015;34:928–932.
  • Gross JE, Lu YJ, Forte S, et al. IL-22 predicts resistance to bacterial colonization in adults with chronic obstructive pulmonary disease. ISPPD7. Tel Aviv, Israel (2010)
  • Lundgren A, Bhuiyan TR, Novak D, et al. Characterization of Th17 responses to streptococcus pneumoniae in humans: comparisons between adults and children in a developed and a developing country. Vaccine. 2012;30:3897–3907.
  • Zhang Q, Bagrade L, Bernatoniene J, et al. Low CD4 T cell immunity to pneumolysin is associated with nasopharyngeal carriage of pneumococci in children. J Infect Dis. 2007;195:1194–1202.
  • Pichichero ME, Kaur R, Casey JR, et al. Antibody response to streptococcus pneumoniae proteins PhtD, LytB, PcpA, PhtE and Ply after nasopharyngeal colonization and acute otitis media in children. Hum Vaccines Immunother. 2012;8:799–805.
  • Goulart C, Rodriguez D, Kanno AI, et al. A combination of recombinant mycobacterium bovis BCG strains expressing pneumococcal proteins induces cellular and humoral immune responses and protects against pneumococcal colonization and sepsis. Clin Vaccine Immunol. 2017;24:e00133-17.
  • Skoberne M, Ferreira DM, Hetherington S, et al. Pneumococcal protein vaccine GEN-004 reduces experimental human pneumococcal carriage in healthy adults. Int Symp Pneumococci Pneumococcal Dis. Glasgow, UK (2016)
  • Mozdzanowska K, Zharikova D, Cudic M, et al. Roles of adjuvant and route of vaccination in antibody response and protection engendered by a synthetic matrix protein 2-based influenza A virus vaccine in the mouse. Virol J. 2007;4:118.
  • Debache K, Guionaud C, Alaeddine F, et al. Intraperitoneal and intranasal vaccination of mice with three distinct recombinant neospora caninum antigens results in differential effects with regard to protection against experimental challenge with neospora caninum tachyzoites. Parasitology. 2010;137:229–240.
  • Svanborg-Edén C, Svennerholm AM. Secretory immunoglobulin A and G antibodies prevent adhesion of escherichia coli to human urinary tract epithelial cells. Infect Immun. 1978;22:790–797.
  • Johnson S, Sypura WD, Gerding DN, et al. Selective neutralization of a bacterial enterotoxin by serum immunoglobulin A in response to mucosal disease. Infect Immun. 1995;63:3166–3173.
  • Mazanec MB, Kaetzel CS, Lamm ME, et al. Intracellular neutralization of virus by immunoglobulin A antibodies. Proc Natl Acad Sci. 1992;89:6901–6905.
  • Underdown BJ, Schiff JM. Immunoglobulin A: strategic defense initiative at the mucosal surface. Annu Rev Immunol. 1986;4:389–417.
  • Brewer JM, Conacher M, Hunter CA, et al. Aluminium hydroxide adjuvant initiates strong antigen-specific Th2 responses in the absence of IL-4- or IL-13-mediated signaling. J Immunol. 1999;163:6448–6454.
  • Marqués JM, Rial A, Muñoz N, et al. Protection against streptococcus pneumoniae serotype 1 acute infection shows a signature of Th17- and IFN-γ-mediated immunity. Immunobiology. 2012;217:420–429.
  • Voß F, van Beek LF, Schwudke D, et al. Lipidation of pneumococcal antigens leads to improved immunogenicity and protection. Vaccines. 2020;8:310.
  • McDaniel LS, Swiatlo E. Should pneumococcal vaccines eliminate nasopharyngeal colonization? MBio. 2016;7::16–17.
  • Castle SC. Clinical relevance of age-related immune dysfunction. Clin Infect Dis. 2000;31:578–585.
  • Haynes L, Eaton SM. The effect of age on the cognate function of CD4+ T cells. Immunol Rev. 2005;205:220–228.
  • Linton P-J, Li SP, Zhang Y, et al. Intrinsic versus environmental influences on T-cell responses in aging. Immunol Rev. 2005;205:207–219.
  • Jiang J, Bennett AJ, Fisher E, et al. Limited expansion of virus-specific CD8 T cells in the aged environment. Mech Ageing Dev. 2009;130:713–721.
  • Goronzy JJ, Hu B, Kim C, et al. Epigenetics of T cell aging. J Leukocyte Biol. 2018;104:691–699.
  • Marwaha AK, Leung NJ, McMurchy AN, et al. TH17 cells in autoimmunity and immunodeficiency: protective or pathogenic? Front Immunol. 2012;3:129.
  • Pichichero ME. Challenges in vaccination of neonates, infants and young children. Vaccine. 2014;32(31):3886–3894.
  • Grijalva CG, Griffin MR, Edwards KM, et al. The role of influenza and parainfluenza infections in nasopharyngeal pneumococcal acquisition among young children. Clin Infect Dis. 2014;58(10):1369–1376.
  • Morpeth SC, Munywoki P, Hammitt LL, et al. Impact of viral upper respiratory tract infection on the concentration of nasopharyngeal pneumococcal carriage among Kenyan children. Sci Rep. 2018;8:11030.
  • Lu YJ, Oliver E, Zhang F, et al. Screening for Th17-dependent pneumococcal vaccine antigens: comparison of murine and human cellular immune responses. Infect Immun. 2018;86:11.
  • Kuipers K, van Selm S, van Opzeeland F, et al. Genetic background impacts vaccine-induced reduction of pneumococcal colonization. Vaccine. 2017;35:5235–5241.
  • Pulendran B, Li S, Nakaya HI. Systems vaccinology. Immunity. 2010;33:516–529.
  • Hagan T, Nakaya HI, Subramaniam S, et al. Systems vaccinology: enabling rational vaccine design with systems biological approaches. Vaccine. 2015;33:5294–5301.
  • Khan MN, Pichichero ME. CD4+ T-cell memory and antibody responses directed against the pneumococcal histidine triad proteins PhtD and PhtE following nasopharyngeal colonization and immunization and their role in protection against pneumococcal colonization in mice. Infect Immun. 2013;81(10):3781–3792.
  • Balachandran P, Brooks-Walter A, Virolainen-Julkunen A, et al. Role of pneumococcal surface protein C in nasopharyngeal carriage and pneumonia and its ability to elicit protection against carriage of streptococcus pneumoniae. Infect Immun. 2002;70(5):2526–2534.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.