Clin Exp Vaccine Res.  2012 Jul;1(1):50-63. 10.7774/cevr.2012.1.1.50.

Mucosal vaccine adjuvants update

Affiliations
  • 1Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. jhrhee@chonnam.ac.kr
  • 2Department of Microbiology and Research Institute of Vibrio Infections, Chonnam National University Medical School, Gwangju, Korea.
  • 3Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea.
  • 4Fraunhofer Korea Center for Biopharmaceutical Research, Hwasun, Korea.

Abstract

Mucosal vaccination, capable of inducing protective immune responses both in the mucosal and systemic immune compartments, has many advantages and is regarded as a blue ocean in the vaccine industry. Mucosal vaccines can offer lower costs, better accessability, needle-free delivery, and higher capacity of mass immunizations during pandemics. However, only very limited number of mucosal vaccines was approved for human use in the market yet. Generally, induction of immune responses following mucosal immunization requires the co-administration of appropriate adjuvants that can initiate and support the effective collaboration between innate and adaptive immunity. Classically, adjuvant researches were rather empirical than keenly scientific. However, during last several years, fundamental scientific achievements in innate immunity have been translated into the development of new mucosal adjuvants. This review focuses on recent developments in the concepts of adjuvants and innate immunity, mucosal immunity with special interest of vaccine development, and basic and applied researches in mucosal adjuvant.

Keyword

Mucosal; Vaccine; Adjuvant; Innate immunity

MeSH Terms

Achievement
Adaptive Immunity
Cooperative Behavior
Humans
Immunity, Innate
Immunity, Mucosal
Immunization
Mass Vaccination
Pandemics
Vaccination
Vaccines
Vaccines

Figure

  • Fig. 1 The target site of vaccine adjuvants. Most of the recently developed specific adjuvants, such as pattern recognition receptor (PRR) ligands act on signal 0 (antigen recognition and antigen-presenting cells [APCs] activation), and indirectly on signal 2 (co-stimulation). In addition, PRR ligands can act on signal 1 (efficient presentation of the co-administered antigen). Modified from Guy [12].

  • Fig. 2 Pattern recognition receptor (PRR) and signaling (A) Toll-like receptors (TLRs). TLR receptors recognize different microbial associated molecular patterns: the heterodimer of TLR4 and MD-2 recognizes lipopolysaccharide (LPS); TLR2 recognizes triacyl and diacyl portions of lipoproteins together with TLR1 or TLR6, respectively; TLR5 recognizes flagellin; TLR3 recognizes double-stranded RNA; TLR7 recognizes single-stranded RNA and TLR9 recognizes bacterial and viral DNA, the so-called CpG DNA. The signaling pathways of TLRs are mediated by selective usage of adaptor molecules, MyD88, Toll-receptor-associated activator of interferon (TRIF), TIR-associated protein (TIRAP) and Toll-receptor-associated molecule (TRAM). (B) C-type lectins (CLRs), retinoic acid-inducible gene-like receptors (RLRs) and nucleotide binding domain (NOD)-like receptors (NLRs). CLRs recognize carbohydrates on microorganisms via the carbohydrate-binding domain. Dectin-1 is well studied. RLRs are composed of two N-terminal caspase-recruitment domains (CARDs), a central DEAD box helicase/ATPase domain, and a C-terminal regulatory domain (RD). They are localized in the cytoplasm and recognize the genomic RNA of dsRNA viruses, and dsRNA generated as the replication intermediate of ssRNA viruses. RLRs interact with IPS1 via their CARD domains, resulting in type 1 interferon production through IkB kinase, inducible (IKKi)/TANK-binding kinase 1 (TBK1). NLRs are composed of a central NOD and C-terminal leucine-rich repeats (LRRs). NODs activate caspase-1, resulting in processing of pro-interleukin-1β (IL-1β) to mature IL-1β. ASC, apoptosis-associated speck-like protein; IFN, interferon; IPS1, IFN-β promoter stimulator 1; IRF, interferon regulatory factor; MDA-5, melanoma-differentiation-associated gene 5; NF-kB, nuclear factor-kB; NLPR3, NLR family, pyrin domain-containing 3; RIG-1, retinoic acid-inducible gene I. Modified from Akira [15].


Cited by  4 articles

Enhancement of antigen-specific humoral immune responses and protein solubility through conjugation of bacterial flagellin, Vibrio vulnificus FlaB, to the N-terminus of porcine epidemic diarrhea virus surface protein antigen S0
Seo-ho Oh, Young-Saeng Kim Cho, Ho-Bin Lee, Sang-Mok Lee, Whee-Soo Kim, Liang Hong, Chong-Su Cho, Yun-Jaie Choi, Sang-Kee Kang
J Vet Sci. 2019;20(6):.    doi: 10.4142/jvs.2019.20.e70.

Intranasal immunization with a flagellin-adjuvanted peptide anticancer vaccine prevents tumor development by enhancing specific cytotoxic T lymphocyte response in a mouse model
Chung Truong Nguyen, Seol Hee Hong, Thuan Trong Ung, Vivek Verma, Soo Young Kim, Joon Haeng Rhee, Shee Eun Lee
Clin Exp Vaccine Res. 2013;2(2):128-134.    doi: 10.7774/cevr.2013.2.2.128.

Towards Vaccine 3.0: new era opened in vaccine research and industry
Joon Haeng Rhee
Clin Exp Vaccine Res. 2014;3(1):1-4.    doi: 10.7774/cevr.2014.3.1.1.

Tetanus toxin fragment C fused to flagellin makes a potent mucosal vaccine
Shee Eun Lee, Chung Truong Nguyen, Soo Young Kim, Thinh Nguyen Thi, Joon Haeng Rhee
Clin Exp Vaccine Res. 2015;4(1):59-67.    doi: 10.7774/cevr.2015.4.1.59.


Reference

1. Chen W, Patel GB, Yan H, Zhang J. Recent advances in the development of novel mucosal adjuvants and antigen delivery systems. Hum Vaccin. 2010. 6:706–714.
Article
2. Lamm ME. Interaction of antigens and antibodies at mucosal surfaces. Annu Rev Microbiol. 1997. 51:311–340.
Article
3. Neutra MR, Kozlowski PA. Mucosal vaccines: the promise and the challenge. Nat Rev Immunol. 2006. 6:148–158.
Article
4. Holmgren J, Czerkinsky C, Eriksson K, Mharandi A. Mucosal immunisation and adjuvants: a brief overview of recent advances and challenges. Vaccine. 2003. 21:Suppl 2. S89–S95.
Article
5. Murphy TV, Gargiullo PM, Massoudi MS, et al. Intussusception among infants given an oral rotavirus vaccine. N Engl J Med. 2001. 344:564–572.
Article
6. Mutsch M, Zhou W, Rhodes P, et al. Use of the inactivated intranasal influenza vaccine and the risk of Bell's palsy in Switzerland. N Engl J Med. 2004. 350:896–903.
Article
7. Fujkuyama Y, Tokuhara D, Kataoka K, et al. Novel vaccine development strategies for inducing mucosal immunity. Expert Rev Vaccines. 2012. 11:367–379.
Article
8. Ramon G. Sur l'augmentation anormale de l'antitoxine chez les chevaux producteurs de serum antidiphtherique. Bull Soc Cent Med Vet. 1925. 101:227–234.
9. Glenny AT. Insoluble precipitates in diphtheria and tetanus immunization. Br Med J. 1930. 2:244–245.
Article
10. Marciani DJ. Vaccine adjuvants: role and mechanisms of action in vaccine immunogenicity. Drug Discov Today. 2003. 8:934–943.
Article
11. Schijns VE. Induction and direction of immune responses by vaccine adjuvants. Crit Rev Immunol. 2001. 21:75–85.
Article
12. Guy B. The perfect mix: recent progress in adjuvant research. Nat Rev Microbiol. 2007. 5:505–517.
Article
13. McKee AS, Munks MW, Marrack P. How do adjuvants work? Important considerations for new generation adjuvants. Immunity. 2007. 27:687–690.
Article
14. Creagh EM, O'Neill LA. TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol. 2006. 27:352–357.
Article
15. Akira S. Innate immunity and adjuvants. Philos Trans R Soc Lond B Biol Sci. 2011. 366:2748–2755.
Article
16. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996. 86:973–983.
Article
17. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006. 124:783–801.
Article
18. Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007. 449:819–826.
Article
19. Shi Z, Cai Z, Sanchez A, et al. A novel Toll-like receptor that recognizes vesicular stomatitis virus. J Biol Chem. 2011. 286:4517–4524.
Article
20. Flacher V, Bouschbacher M, Verronèse E, et al. Human Langerhans cells express a specific TLR profile and differentially respond to viruses and Gram-positive bacteria. J Immunol. 2006. 177:7959–7967.
Article
21. Jarrossay D, Napolitani G, Colonna M, Sallusto F, Lanzavecchia A. Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells. Eur J Immunol. 2001. 31:3388–3393.
Article
22. Schwarz TF. Clinical update of the AS04-adjuvanted human papillomavirus-16/18 cervical cancer vaccine, Cervarix. Adv Ther. 2009. 26:983–998.
Article
23. 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. 2009. 183:6186–6197.
Article
24. Vollmer J, Krieg AM. Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev. 2009. 61:195–204.
Article
25. Duthie MS, Windish HP, Fox CB, Reed SG. Use of defined TLR ligands as adjuvants within human vaccines. Immunol Rev. 2011. 239:178–196.
Article
26. Fritz JH, Ferrero RL, Philpott DJ, Girardin SE. Nod-like proteins in immunity, inflammation and disease. Nat Immunol. 2006. 7:1250–1257.
Article
27. Inohara , Chamaillard , McDonald C, Nuñez G. NOD-LRR proteins: role in host-microbial interactions and inflammatory disease. Annu Rev Biochem. 2005. 74:355–383.
Article
28. Traub S, von Aulock S, Hartung T, Hermann C. MDP and other muropeptides: direct and synergistic effects on the immune system. J Endotoxin Res. 2006. 12:69–85.
Article
29. Meylan E, Tschopp J, Karin M. Intracellular pattern recognition receptors in the host response. Nature. 2006. 442:39–44.
Article
30. Schroder K, Tschopp J. The inflammasomes. Cell. 2010. 140:821–832.
Article
31. Holt LB. Quantitative studies in diphtheria prophylaxis: the second response. Br J Exp Pathol. 1950. 31:233–241.
32. Iyer S, HogenEsch H, Hem SL. Relationship between the degree of antigen adsorption to aluminum hydroxide adjuvant in interstitial fluid and antibody production. Vaccine. 2003. 21:1219–1223.
Article
33. Li H, Nookala S, Re F. Aluminum hydroxide adjuvants activate caspase-1 and induce IL-1beta and IL-18 release. J Immunol. 2007. 178:5271–5276.
Article
34. Eisenbarth SC, Colegio OR, O'Connor W, Sutterwala FS, Flavell RA. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008. 453:1122–1126.
Article
35. 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. 2008. 181:3755–3759.
Article
36. Hornung V, Bauernfeind F, Halle A, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008. 9:847–856.
Article
37. Franchi L, Nunez G. The Nlrp3 inflammasome is critical for aluminium hydroxide-mediated IL-1beta secretion but dispensable for adjuvant activity. Eur J Immunol. 2008. 38:2085–2089.
Article
38. Matzinger P. The danger model: a renewed sense of self. Science. 2002. 296:301–305.
Article
39. Spreafico R, Ricciardi-Castagnoli P, Mortellaro A. The controversial relationship between NLRP3, alum, danger signals and the next-generation adjuvants. Eur J Immunol. 2010. 40:638–642.
Article
40. Sharp FA, Ruane D, Claass B, et al. Uptake of particulate vaccine adjuvants by dendritic cells activates the NALP3 inflammasome. Proc Natl Acad Sci U S A. 2009. 106:870–875.
Article
41. Li H, Willingham SB, Ting JP, Re F. Cutting edge: inflammasome activation by alum and alum's adjuvant effect are mediated by NLRP3. J Immunol. 2008. 181:17–21.
Article
42. Yoneyama M, Fujita T. RNA recognition and signal transduction by RIG-I-like receptors. Immunol Rev. 2009. 227:54–65.
Article
43. Yoneyama M, Kikuchi M, Natsukawa T, et al. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004. 5:730–737.
Article
44. Takeuchi O, Akira S. Innate immunity to virus infection. Immunol Rev. 2009. 227:75–86.
Article
45. Kato H, Takeuchi O, Sato S, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006. 441:101–105.
Article
46. Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009. 461:788–792.
Article
47. Takaoka A, Wang Z, Choi MK, et al. DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature. 2007. 448:501–505.
Article
48. Gonzalez-Navajas JM, Lee J, David M, Raz E. Immunomodulatory functions of type I interferons. Nat Rev Immunol. 2012. 12:125–135.
Article
49. Lavelle EC, Murphy C, O'Neill LA, Creagh EM. The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis. Mucosal Immunol. 2010. 3:17–28.
Article
50. Varol C, Vallon-Eberhard A, Elinav E, et al. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009. 31:502–512.
Article
51. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007. 204:1757–1764.
Article
52. Kiyono H, Fukuyama S. NALT-versus Peyer's-patch-mediated mucosal immunity. Nat Rev Immunol. 2004. 4:699–710.
53. Yuki Y, Kiyono H. New generation of mucosal adjuvants for the induction of protective immunity. Rev Med Virol. 2003. 13:293–310.
Article
54. Kim DY, Sato A, Fukuyama S, et al. The airway antigen sampling system: respiratory M cells as an alternative gateway for inhaled antigens. J Immunol. 2011. 186:4253–4262.
Article
55. Yamamoto M, Pascual DW, Kiyono H. M cell-targeted mucosal vaccine strategies. Curr Top Microbiol Immunol. 2012. 354:39–52.
Article
56. Chen K, Cerutti A. Vaccination strategies to promote mucosal antibody responses. Immunity. 2010. 33:479–491.
Article
57. Kunisawa J, Nochi T, Kiyono H. Immunological commonalities and distinctions between airway and digestive immunity. Trends Immunol. 2008. 29:505–513.
Article
58. Holmgren J, Czerkinsky C. Mucosal immunity and vaccines. Nat Med. 2005. 11:4 Suppl. S45–S53.
Article
59. Lawson LB, Norton EB, Clements JD. Defending the mucosa: adjuvant and carrier formulations for mucosal immunity. Curr Opin Immunol. 2011. 23:414–420.
Article
60. Mishra N, Goyal AK, Tiwari S, et al. Recent advances in mucosal delivery of vaccines: role of mucoadhesive/biodegradable polymeric carriers. Expert Opin Ther Pat. 2010. 20:661–679.
Article
61. Pavot V, Rochereau N, Genin C, Verrier B, Paul S. New insights in mucosal vaccine development. Vaccine. 2012. 30:142–154.
Article
62. Eriksson K, Holmgren J. Recent advances in mucosal vaccines and adjuvants. Curr Opin Immunol. 2002. 14:666–672.
Article
63. Jabbal-Gill I, Watts P, Smith A. Chitosan-based delivery systems for mucosal vaccines. Expert Opin Drug Deliv. 2012. 06. 19. [Epub] http://dx.doi.org/10.1517/17425247.2012.697455.
Article
64. Freytag LC, Clements JD. Mucosal adjuvants. Vaccine. 2005. 23:1804–1813.
Article
65. Bracho G, Lastre M, del Campo J, et al. Proteoliposome derived cochleate as novel adjuvant. Vaccine. 2006. 24:Suppl 2. S2-30–S2-31.
Article
66. Cox E, Verdonck F, Vanrompay D, Goddeeris B. Adjuvants modulating mucosal immune responses or directing systemic responses towards the mucosa. Vet Res. 2006. 37:511–539.
Article
67. Coulter A, Harris R, Davis R, et al. Intranasal vaccination with ISCOMATRIX adjuvanted influenza vaccine. Vaccine. 2003. 21:946–949.
Article
68. Sanders MT, Deliyannis G, Pearse MJ, McNamara MK, Brown LE. Single dose intranasal immunization with ISCOMATRIX vaccines to elicit antibody-mediated clearance of influenza virus requires delivery to the lower respiratory tract. Vaccine. 2009. 27:2475–2482.
Article
69. Vujanic A, Snibson KJ, Wee JL, et al. Long-term antibody and immune memory response induced by pulmonary delivery of the influenza Iscomatrix vaccine. Clin Vaccine Immunol. 2012. 19:79–83.
Article
70. Lee SE, Kim SY, Jeong BC, et al. A bacterial flagellin, Vibrio vulnificus FlaB, has a strong mucosal adjuvant activity to induce protective immunity. Infect Immun. 2006. 74:694–702.
Article
71. Nguyen CT, Kim SY, Kim MS, Lee SE, Rhee JH. Intranasal immunization with recombinant PspA fused with a flagellin enhances cross-protective immunity against Streptococcus pneumoniae infection in mice. Vaccine. 2011. 29:5731–5739.
Article
72. Hong SH, Byun YH, Nguyen CT, et al. Intranasal administration of a flagellin-adjuvanted inactivated influenza vaccine enhances mucosal immune responses to protect mice against lethal infection. Vaccine. 2012. 30:466–474.
Article
73. Chen W, Kuolee R, Yan H. The potential of 3',5'-cyclic diguanylic acid (c-di-GMP) as an effective vaccine adjuvant. Vaccine. 2010. 28:3080–3085.
Article
74. Yan H, KuoLee R, Tram K, et al. 3',5'-Cyclic diguanylic acid elicits mucosal immunity against bacterial infection. Biochem Biophys Res Commun. 2009. 387:581–584.
Article
75. Mowat AM, Smith RE, Donachie AM, Furrie E, Grdic D, Lycke N. Oral vaccination with immune stimulating complexes. Immunol Lett. 1999. 65:133–140.
Article
76. Noda K, Kodama S, Umemoto S, Abe N, Hirano T, Suzuki M. Nasal vaccination with P6 outer membrane protein and alpha-galactosylceramide induces nontypeable Haemophilus influenzae-specific protective immunity associated with NKT cell activation and dendritic cell expansion in nasopharynx. Vaccine. 2010. 28:5068–5074.
Article
77. Lee YS, Lee KA, Lee JY, et al. An alpha-GalCer analogue with branched acyl chain enhances protective immune responses in a nasal influenza vaccine. Vaccine. 2011. 29:417–425.
Article
78. Courtney AN, Nehete PN, Nehete BP, Thapa P, Zhou D, Sastry KJ. Alpha-galactosylceramide is an effective mucosal adjuvant for repeated intranasal or oral delivery of HIV peptide antigens. Vaccine. 2009. 27:3335–3341.
Article
79. Lindqvist M, Persson J, Thorn K, Harandi AM. The mucosal adjuvant effect of alpha-galactosylceramide for induction of protective immunity to sexually transmitted viral infection. J Immunol. 2009. 182:6435–6443.
Article
80. de Haan L, Hirst TR. Cholera toxin and related enterotoxins: a cell biological and immunological perspective. J Nat Toxins. 2000. 9:281–297.
81. van Ginkel FW, Jackson RJ, Yoshino N, et al. Enterotoxin-based mucosal adjuvants alter antigen trafficking and induce inflammatory responses in the nasal tract. Infect Immun. 2005. 73:6892–6902.
Article
82. Eriksson K, Fredriksson M, Nordström I, Holmgren J. Cholera toxin and its B subunit promote dendritic cell vaccination with different influences on Th1 and Th2 development. Infect Immun. 2003. 71:1740–1747.
Article
83. George-Chandy A, Eriksson K, Lebens M, Nordström I, Schön E, Holmgren J. Cholera toxin B subunit as a carrier molecule promotes antigen presentation and increases CD40 and CD86 expression on antigen-presenting cells. Infect Immun. 2001. 69:5716–5725.
Article
84. Pizza M, Giuliani MM, Fontana MR, et al. Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants. Vaccine. 2001. 19:2534–2541.
Article
85. Mowat AM, Donachie AM, Jagewall S, et al. CTA1-DD-immune stimulating complexes: a novel, rationally designed combined mucosal vaccine adjuvant effective with nanogram doses of antigen. J Immunol. 2001. 167:3398–3405.
Article
86. Sanchez J, Wallerstrom G, Fredriksson M, Angstrom J, Holmgren J. Detoxification of cholera toxin without removal of its immunoadjuvanticity by the addition of (STa-related) peptides to the catalytic subunit. A potential new strategy to generate immunostimulants for vaccination. J Biol Chem. 2002. 277:33369–33377.
87. Norton EB, Lawson LB, Freytag LC, Clements JD. Characterization of a mutant Escherichia coli heat-labile toxin, LT(R192G/L211A), as a safe and effective oral adjuvant. Clin Vaccine Immunol. 2011. 18:546–551.
Article
88. Anosova NG, Chabot S, Shreedhar V, Borawski JA, Dickinson BL, Neutra MR. Cholera toxin, E. coli heat-labile toxin, and non-toxic derivatives induce dendritic cell migration into the follicle-associated epithelium of Peyer's patches. Mucosal Immunol. 2008. 1:59–67.
Article
89. Fahlen-Yrlid L, Gustafsson T, Westlund J, et al. CD11c (high) dendritic cells are essential for activation of CD4+ T cells and generation of specific antibodies following mucosal immunization. J Immunol. 2009. 183:5032–5041.
Article
90. Chang SY, Cha HR, Igarashi O, et al. Cutting edge: Langerin+ dendritic cells in the mesenteric lymph node set the stage for skin and gut immune system cross-talk. J Immunol. 2008. 180:4361–4365.
Article
91. Datta SK, Sabet M, Nguyen KP, et al. Mucosal adjuvant activity of cholera toxin requires Th17 cells and protects against inhalation anthrax. Proc Natl Acad Sci U S A. 2010. 107:10638–10643.
Article
92. Lu YJ, Yadav P, Clements JD, et al. Options for inactivation, adjuvant, and route of topical administration of a killed, unencapsulated pneumococcal whole-cell vaccine. Clin Vaccine Immunol. 2010. 17:1005–1012.
Article
93. Meza-Sanchez D, Perez-Montesinos G, Sanchez-Garcia J, Moreno J, Bonifaz LC. Intradermal immunization in the ear with cholera toxin and its non-toxic beta subunit promotes efficient Th1 and Th17 differentiation dependent on migrating DCs. Eur J Immunol. 2011. 41:2894–2904.
94. Raghavan S, Ostberg AK, Flach CF, et al. Sublingual immunization protects against Helicobacter pylori infection and induces T and B cell responses in the stomach. Infect Immun. 2010. 78:4251–4260.
Article
95. Dubin PJ, Kolls JK. Th17 cytokines and mucosal immunity. Immunol Rev. 2008. 226:160–171.
Article
96. Paavonen J, Naud P, Salmeron J, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009. 374:301–314.
97. Schwarz TF, Spaczynski M, Schneider A, et al. Immunogenicity and tolerability of an HPV-16/18 AS04-adjuvanted prophylactic cervical cancer vaccine in women aged 15-55 years. Vaccine. 2009. 27:581–587.
Article
98. Cranage MP, Fraser CA, Cope A, et al. Antibody responses after intravaginal immunisation with trimeric HIV-1 CN54 clade C gp140 in Carbopol gel are augmented by systemic priming or boosting with an adjuvanted formulation. Vaccine. 2011. 29:1421–1430.
Article
99. Huang CF, Wu TC, Chu YH, Hwang KS, Wang CC, Peng HJ. Effect of neonatal sublingual vaccination with native or denatured ovalbumin and adjuvant CpG or cholera toxin on systemic and mucosal immunity in mice. Scand J Immunol. 2008. 68:502–510.
Article
100. Pesce I, Monaci E, Muzzi A, et al. Intranasal administration of CpG induces a rapid and transient cytokine response followed by dendritic and natural killer cell activation and recruitment in the mouse lung. J Innate Immun. 2010. 2:144–159.
Article
101. Bode C, Zhao G, Steinhagen F, Kinjo T, Klinman DM. CpG DNA as a vaccine adjuvant. Expert Rev Vaccines. 2011. 10:499–511.
Article
102. Samatey FA, Imada K, Nagashima S, et al. Structure of the bacterial flagellar protofilament and implications for a switch for supercoiling. Nature. 2001. 410:331–337.
Article
103. Ramos HC, Rumbo M, Sirard JC. Bacterial flagellins: mediators of pathogenicity and host immune responses in mucosa. Trends Microbiol. 2004. 12:509–517.
Article
104. Staats HF, Bradney CP, Gwinn WM, et al. Cytokine requirements for induction of systemic and mucosal CTL after nasal immunization. J Immunol. 2001. 167:5386–5394.
Article
105. Eo SK, Lee S, Kumaraguru U, Rouse BT. Immunopotentiation of DNA vaccine against herpes simplex virus via co-delivery of plasmid DNA expressing CCR7 ligands. Vaccine. 2001. 19:4685–4693.
Article
106. Lillard JW Jr, Boyaka PN, Taub DD, McGhee JR. RANTES potentiates antigen-specific mucosal immune responses. J Immunol. 2001. 166:162–169.
Article
107. Thompson AL, Johnson BT, Sempowski GD, et al. Maximal adjuvant activity of nasally delivered IL-1alpha requires adjuvant-responsive CD11c(+) cells and does not correlate with adjuvant-induced in vivo cytokine production. J Immunol. 2012. 188:2834–2846.
Article
108. Agrawal S, Gupta S, Agrawal A. Human dendritic cells activated via dectin-1 are efficient at priming Th17, cytotoxic CD8 T and B cell responses. PLoS One. 2010. 5:e13418.
Article
109. McGowen AL, Hale LP, Shelburne CP, Abraham SN, Staats HF. The mast cell activator compound 48/80 is safe and effective when used as an adjuvant for intradermal immunization with Bacillus anthracis protective antigen. Vaccine. 2009. 27:3544–3552.
Article
110. Merluzzi S, Frossi B, Gri G, Parusso S, Tripodo C, Pucillo C. Mast cells enhance proliferation of B lymphocytes and drive their differentiation toward IgA-secreting plasma cells. Blood. 2010. 115:2810–2817.
Article
111. Hu KF, Lovgren-Bengtsson K, Morein B. Immunostimulating complexes (ISCOMs) for nasal vaccination. Adv Drug Deliv Rev. 2001. 51:149–159.
Article
112. Kersten G, Hirschberg H. Antigen delivery systems. Expert Rev Vaccines. 2004. 3:453–462.
Article
113. Romling U, Amikam D. Cyclic di-GMP as a second messenger. Curr Opin Microbiol. 2006. 9:218–228.
Article
114. Pedersen GK, Ebensen T, Gjeraker IH, et al. Evaluation of the sublingual route for administration of influenza H5N1 virosomes in combination with the bacterial second messenger c-di-GMP. PLoS One. 2011. 6:e26973.
Article
Full Text Links
  • CEVR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr