1,295
Views
157
CrossRef citations to date
0
Altmetric
Review

Trends in vaccine adjuvants

&
Pages 539-550 | Published online: 09 Jan 2014

References

  • Ramon G. Diphtheria toxin and anatoxin. Ann. Inst. Pasteur.38, 1 (1924).
  • Caccamo N, Guggino G, Joosten SA et al. Multifunctional CD4+ T cells correlate with active Mycobacterium tuberculosis infection. Eur. J. Immunol.40(8), 2211–2220 (2010).
  • Piedra PA, Jewell AM, Cron SG, Atmar RL, Glezen WP. Correlates of immunity to respiratory syncytial virus (RSV) associated-hospitalization: establishment of minimum protective threshold levels of serum neutralizing antibodies. Vaccine21(24), 3479–3482 (2003).
  • Mallery DL, McEwan WA, Bidgood SR, Towers GJ, Johnson CM, James LC. Antibodies mediate intracellular immunity through tripartite motif-containing 21 (TRIM21). Proc. Natl Acad. Sci. USA107(46), 19985–19990 (2010).
  • Kennedy R, Celis E. Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol. Rev.222, 129–144 (2008).
  • Rinaudo CD, Telford JL, Rappuoli R, Seib KL. Vaccinology in the genome era. J. Clin. Invest.119(9), 2515–2525 (2009).
  • Pizza M, Scarlato V, Masignani V et al. Identification of vaccine candidates against serogroup B meningococcus by whole-genome sequencing. Science287(5459), 1816–1820 (2000).
  • Moriel DG, Bertoldi I, Spagnuolo A et al. Identification of protective and broadly conserved vaccine antigens from the genome of extraintestinal pathogenic Escherichia coli. Proc. Natl Acad. Sci. USA1107(20), 9072–9077 (2010).
  • Maione D, Margarit I, Rinaudo CD et al. Identification of a universal Group B Streptococcus vaccine by multiple genome screen. Science309(5731), 148–150 (2005).
  • Nakagawa M, Gupta SK, Coleman HN, Sellers MA, Banken JA, Greenfield WW. A favorable clinical trend is associated with CD8 T-cell immune responses to the human papillomavirus type 16 e6 antigens in women being studied for abnormal pap smear results. J. Low. Genit. Tract Dis.14(2), 124–129 (2010).
  • Le Moignic P. Les vaccins en emulsion dans les corps gras ou “lipo-vaccins”. Comptes C. R. Seances Soc. Biol. Fil.79, 201–203 (1916).
  • Glenny AT, Pope C, Waddington H, Wallace U. The antigenic value of toxoid precipitated by potassium alum. J. Pathol. Bacteriol.29, 28–45 (1926).
  • Steinman RM. Dendritic cells in vivo: a key target for a new vaccine science. Immunity29(3), 319–324 (2008).
  • Kalinski P, Hilkens CM, Wieringa EA, Kapsenberg ML. T-cell priming by type-1 and type-2 polarized dendritic cells: the concept of a third signal. Immunol. Today20, 561–567 (1999).
  • Bluestone JA, Mackay CR, O’Shea JJ. Stockinger B. The functional plasticity of T cell subsets. Nat. Rev. Immunol.9, 811–816 (2009).
  • Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol.3, 133–146 (2003).
  • Kaufmann SH, McMichael AJ. Annulling a dangerous liaison: vaccination strategies against AIDS and tuberculosis. Nat. Med.11, S33–S44 (2005).
  • Stevenson MM, Riley EM. Innate immunity to malaria. Nat. Rev. Immunol.4, 169–180 (2004).
  • Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat. Rev. Drug. Discov.5, 471–484 (2006).
  • Happel KI, Dubin PJ, Zheng M et al. Divergent roles of IL-23 and IL-12 in host defense against Klebsiella pneumoniae. J. Exp. Med.202, 761–769 (2005).
  • Dong C. TH17 cells in development: an updated view of their molecular identity and genetic programming. Nat. Rev. Immunol.8, 337–348 (2008).
  • King C. New insights into the differentiation and function of T follicular helper cells. Nat. Rev. Immunol.9(11), 757–766 (2009).
  • Jerne NK. The generative grammar of the immune system. EMBO J.4, 847–852 (1985).
  • Schijns VEJC. Induction and direction of immune responses by vaccine adjuvants. Crit. Rev. Immunol.21, 75–85 (2001).
  • Schijns VEJC. Immunological concepts of vaccine adjuvant activity. Curr. Opin. Immunol.12, 456–463 (2000).
  • Dupuis M, McDonald DM, Ott G. Distribution of adjuvant MF59 and antigen gD2 after intramuscular injection in mice. Vaccine18(5–6), 434–439 (1999).
  • Tanaka T, Legat A, Adam E et al. DiC14-amidine cationic liposomes stimulate myeloid dendritic cells through Toll-like receptor 4. Eur. J. Immunol.38(5), 1351–1357 (2008).
  • Zinkernagel RM, Ehl S, Aichele P, Oehen S, Kundig T, Hengartner H. Antigen localisation regulates immune responses in a dose- and time-dependent fashion: a geographical view of immune reactivity. Immunol. Rev.156, 199–209 (1997).
  • Herbert WJ. Antigenicity of soluble protein in the presence of high levels of antibody: a possible mode of action of the antigen adjuvants. Nature210, 747–748 (1966).
  • Herbert WJ. The mode of action of mineral-oil emulsion adjuvants on antibody production in mice. Immunology14, 301–318 (1968).
  • Holm BC, Svelander L, Bucht A, Lorentzen JC. The arthritogenic adjuvant squalene does not accumulate in joints, but gives rise to pathogenic cells in both draining and non-draining lymph nodes. Clin. Exp. Immunol.127, 430–435 (2002).
  • Seubert A, Monaci E, Pizza M, O’Hagan DT, Wack A. The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells. J. Immunol.180, 5402–5412 (2004).
  • Mosca F, Tritto E, Muzzi A et al. molecular and cellular signatures of human adjuvants Proc. Natl Acad. Sci. USA105, 10501–10506 (2008).
  • Phythian-Adams AT, Cook PC, Lundie RJ et al. CD11c depletion severely disrupts Th2 induction and development in vivo. J. Exp. Med.207(10), 2089–2096 (2010).
  • Neill DR, Wong SH, Bellosi A et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature464(7293), 1367–1370 (2010).
  • Kool M, Soullié T, van Nimwegen M et al. Alum adjuvant boosts adaptive immunity by inducing uric acid and activating inflammatory dendritic cells. J. Exp. Med.205(4), 869–882 (2008).
  • Jordan MB, Mills DM, Kappler J, Marrack P, Cambier JC. Promotion of B cell immune responses via an alum-induced myeloid cell population. Science304(5678), 1808–1810 (2004).
  • Eng NF, Garlapati S, Gerdts V, Potter A, Babiuk LA, Mutwiri GK. The potential of polyphosphazenes for delivery of vaccine antigens and immunotherapeutic agents. Curr. Drug Deliv.7(1), 13–20 (2010).
  • Estevan M, Gamazo C, Grilló MJ, Del Barrio GG, Blasco JM, Irache JM. Experiments on a sub-unit vaccine encapsulated in microparticles and its efficacy against Brucella melitensis in mice. Vaccine24(19), 4179–4187 (2006).
  • Lavelle EC, O’Hagan DT. Delivery systems and adjuvants for oral vaccines. Expert Opin. Drug Deliv.3(6), 747–762 (2006).
  • Janeway CA Jr. The immune system evolved to discriminate infectious nonself from noninfectious self. Immunol. Today13, 11–16 (1992).
  • Poltorak A, He X, Smirnova I et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in TLR4 gene. Science282, 2085–2088 (1998).
  • Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu. Rev. Immunol.21, 335–376 (2003).
  • Janeway CA Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb. Symp. Quant. Biol.54, 1–13 (1989).
  • Didierlaurent AM, Morel S, Lockman L et al. AS04, an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity. J. Immunol.183(10), 6186–6197 (2009).
  • Gavin AL, Hoebe K, Duong B et al. Adjuvant-enhanced antibody responses in the absence of Toll-like receptor signaling. Science314, 1936–1938 (2006).
  • McKee AS, Munks MW, MacLeod MK et al. Alum induces innate immune responses through macrophage and mast cell sensors, but these sensors are not required for alum to act as an adjuvant for specific immunity. J. Immunol.183(7), 4403–4414 (2009).
  • Calabro S, Tortoli M, Baudner BC et al. Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine29(9), 1812–1823 (2011).
  • Ting JP, Lovering RC, Alnemri ES et al. The NLR gene family: a standard nomenclature. Immunity28(3), 285–287 (2008).
  • Ishikawa E, Ishikawa T, Morita YS et al. Direct recognition of the mycobacterial glycolipid, trehalose dimycolate, by C-type lectin Mincle. J. Exp. Med.206(13), 2879–2888 (2009).
  • Schoenen H, Bodendorfer B, Hitchens K et al. Cutting edge: Mincle is essential for recognition and adjuvanticity of the mycobacterial cord factor and its synthetic analog trehalose-dibehenate. J. Immunol.184(6), 2756–2760 (2010).
  • Brown GD, Gordon S. Immune recognition. A new receptor for β-glucans. Nature413(6851), 36–37 (2001).
  • Brown GD, Herre J, Williams DL, Willment JA, Marshall AS, Gordon S. Dectin-1 mediates the biological effects of β-glucans. J. Exp. Med.197(9), 1119–1124 (2003).
  • Unterholzner L, Keating SE, Baran M et al. IFI16 is an innate immune sensor for intracellular DNA. Nat. Immunol.11(11), 997–1004 (2010).
  • Goodridge HS, Underhill DM. Fungal Recognition by TLR2 and Dectin-1. Handb. Exp. Pharmacol.183, 87–109 (2008).
  • Ma R, Du JL, Huang J, Chang-you W. Additive effects of CpG ODN and R-848 as adjuvants on augmenting immune responses to HBsAg vaccination. Biochem. Biophys. Res. Commun.361(2), 537–542 (2007).
  • Matzinger P. Tolerance, danger and the extended family. Ann. Rev. Immunol.12, 991–1045 (1994).
  • Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature425(6957), 516–521 (2003).
  • Harris J, Sharp FA, Lavelle EC. The role of inflammasomes in the immunostimulatory effects of particulate vaccine adjuvants. Eur. J. Immunol.40(3), 634–638 (2010).
  • Kool M, Pétrilli V, De Smedt T et al. Cutting edge: alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J. Immunol.181(6), 3755–3759 (2008).
  • Franchi L, Núñez G. The Nlrp3 inflammasome is critical for aluminium hydroxide-mediated IL-1β secretion but dispensable for adjuvant activity. Eur. J. Immunol.38(8), 2085–2089 (2008).
  • Sharp FA, Ruane D, Claass B et al. Uptake of particulate vaccine adjuvants by dendritic cells activates the NALP3 inflammasome. Proc. Natl Acad. Sci. USA106, 80–875 (2009).
  • Gross O, Poeck H, Bscheider M et al. Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature459(7245), 433–436 (2009).
  • Allen IC, Scull MA, Moore CB et al. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity30(4), 556–565 (2009).
  • Willingham SB, Allen IC, Bergstralh DT et al. NLRP3 (NALP3, Cryopyrin) facilitates in vivo caspase-1 activation, necrosis, and HMGB1 release via inflammasome-dependent and -independent pathways. J. Immunol.183(3), 2008–2015 (2009).
  • McNeela EA, Burke Á, Neill DR et al. Pneumolysin activates the NLRP3 inflammasome and promotes proinflammatory cytokines independently of TLR4. PLoS Pathog.6(11), e1001191 (2010).
  • Broz P, Newton K, Lamkanfi M, Mariathasan S, Dixit VM, Monack DM. Redundant roles for inflammasome receptors NLRP3 and NLRC4 in host defense against Salmonella. J. Exp. Med.207(8), 1745–1755 (2010).
  • Morein B, Villacrés-Eriksson M, Lövgren-Bengtsson K. Iscom, a delivery system for parenteral and mucosal vaccination. Dev. Biol. Stand.92, 33–39 (1998).
  • Aichinger MC, Ginzler M, Weghuber J et al. Adjuvating the adjuvant: facilitated delivery of an immunomodulatory oligonucleotide to TLR9 by a cationic antimicrobial peptide in dendritic cells. Vaccine29(3), 426–436 (2010).
  • Lay M, Callejo B, Chang S et al. Cationic lipid/DNA complexes (JVRS-100) combined with influenza vaccine (Fluzone) increases antibody response, cellular immunity and antigenically drifted protection. Vaccine27(29), 3811–3820 (2009).
  • Eckl-Dorna J, Batista FD. BCR-mediated uptake of antigen linked to TLR9 ligand stimulates B-cell proliferation and antigen-specific plasma cell formation. Blood113(17), 3969–3977 (2009).
  • Kamath AT, Valenti MP, Rochat AF et al. Protective anti-mycobacterial T cell responses through exquisite in vivo activation of vaccine-targeted dendritic cells. Eur. J. Immunol.38(5), 1247–1256 (2008).
  • Wack A, Baudner BC, Hilbert AK et al. Combination adjuvants for the induction of potent, long-lasting antibody and T-cell responses to influenza vaccine in mice. Vaccine26(4), 552–561 (2008).
  • Chabalgoity JA, Baz A, Rial A, Grille S. The relevance of cytokines for development of protective immunity and rational design of vaccines. Cytokine Growth Factor Rev.18, 195–207 (2007).
  • Wickelgren I. Mouse studies question importance of Toll-like receptors to vaccines. Science314, 1859–1860 (2006).
  • Reddy ST, Swartz MA, Hubbell JA. Targeting dendritic cells with biomaterials: developing the next generation of vaccines. Trends Immunol.27(12), 573–579 (2006).
  • Bonifaz LC, Bonnyay DP, Charalambous A et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J. Exp. Med.199(6), 815–824 (2004).
  • Wang RF, Wang HY. Enhancement of antitumor immunity by prolonging antigen presentation on dendritic cells. Nat. Biotechnol.20(2), 149–154 (2002).
  • Jansen T, Hofmans MP, Theelen MJ, Schijns VEJC. Structure-activity relations of water-in-oil vaccine formulations and induced antigen-specific antibody responses. Vaccine 11, 23(8), 1053–1060 (2005).
  • Jansen T, Hofmans MP, Theelen MJ, Manders FG, Schijns VEJC. Dose and timing requirements for immunogenicity of viral poultry vaccine antigen: investigations of emulsion-based depot function. Avian Pathol.36(5), 361–365 (2007).
  • O’Hagan DT, Valiante NM. Recent advances in the discovery and delivery of vaccine adjuvants. Nat. Rev. Drug Discov.2(9), 727–735 (2003).
  • Guy B. The prefect mix: recent progress in adjuvant research. Nat. Rev. Micr.5(7), 505–517 (2007).
  • Mizel SB, Bates JT. Flagellin as an adjuvant: cellular mechanisms and potential. J. Immunol.185(10), 5677–5682 (2010).
  • Eckl-Dorna J, Batista FD. BCR-mediated uptake of antigen linked to TLR9 ligand stimulates B-cell proliferation and antigen-specific plasma cell formation. Blood113(17), 3969–3977 (2009).
  • Kapadia D, Fong L. CTLA-4 blockade: autoimmunity as treatment. J. Clin. Oncol.23, 8926–8928 (2005).
  • Saraiva M, O’Garra A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol.10(3), 170–181 (2010).
  • Yang L, Pang Y, Moses HL. TGF-β and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol.31(6), 220–227 (2010).
  • Siegrist CA, Aspinall R. B-cell responses to vaccination at the extremes of age. Nat. Rev. Immunol.9, 185–194 (2009).

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.