Clin Exp Vaccine Res.  2015 Jan;4(1):23-45. 10.7774/cevr.2015.4.1.23.

Polyionic vaccine adjuvants: another look at aluminum salts and polyelectrolytes

Affiliations
  • 1PharmAthene, Inc., Annapolis, MD, USA. bradford.powell@pharmathene.com
  • 2Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA.

Abstract

Adjuvants improve the adaptive immune response to a vaccine antigen by modulating innate immunity or facilitating transport and presentation. The selection of an appropriate adjuvant has become vital as new vaccines trend toward narrower composition, expanded application, and improved safety. Functionally, adjuvants act directly or indirectly on antigen presenting cells (APCs) including dendritic cells (DCs) and are perceived as having molecular patterns associated either with pathogen invasion or endogenous cell damage (known as pathogen associated molecular patterns [PAMPs] and damage associated molecular patterns [DAMPs]), thereby initiating sensing and response pathways. PAMP-type adjuvants are ligands for toll-like receptors (TLRs) and can directly affect DCs to alter the strength, potency, speed, duration, bias, breadth, and scope of adaptive immunity. DAMP-type adjuvants signal via proinflammatory pathways and promote immune cell infiltration, antigen presentation, and effector cell maturation. This class of adjuvants includes mineral salts, oil emulsions, nanoparticles, and polyelectrolytes and comprises colloids and molecular assemblies exhibiting complex, heterogeneous structures. Today innovation in adjuvant technology is driven by rapidly expanding knowledge in immunology, cross-fertilization from other areas including systems biology and materials sciences, and regulatory requirements for quality, safety, efficacy and understanding as part of the vaccine product. Standardizations will aid efforts to better define and compare the structure, function and safety of adjuvants. This article briefly surveys the genesis of adjuvant technology and then re-examines polyionic macromolecules and polyelectrolyte materials, adjuvants currently not known to employ TLR. Specific updates are provided for aluminum-based formulations and polyelectrolytes as examples of improvements to the oldest and emerging classes of vaccine adjuvants in use.

Keyword

Immunologic adjuvant; Innate immunity; Toll like receptors; Pattern recognition receptors; Nalp3 protein; Alum; Aluminum hydroxide; Polymers; Chitosan; Polyphosphazene; Polyoxidonium

MeSH Terms

Adaptive Immunity
Adjuvants, Immunologic
Allergy and Immunology
Aluminum Hydroxide
Aluminum*
Antigen Presentation
Antigen-Presenting Cells
Bias (Epidemiology)
Chitosan
Colloids
Dendritic Cells
Emulsions
Immunity, Innate
Ligands
Nanoparticles
Polymers
Receptors, Pattern Recognition
Salts*
Systems Biology
Toll-Like Receptors
Vaccines
Adjuvants, Immunologic
Aluminum
Aluminum Hydroxide
Chitosan
Colloids
Emulsions
Ligands
Polymers
Receptors, Pattern Recognition
Salts
Toll-Like Receptors
Vaccines

Figure

  • Fig. 1 Proportion of human vaccines containing adjuvant through stages of history. Circles depict periods in vaccine development with fractional amount containing adjuvant shaded in blue, and diameter proportional to the log of number of different vaccines. A, up to 1899; B, 1900 to 1949; C, 1950 to 2012; D, 2014 U.S. licensed as listed by Food and Development Administration (FDA); E, 2014 in clinical testing as listed by HuVax (http://www.violinet.org). Note that the licensed vaccines group D contains many more multiple products for similar or overlapping indications, most of which are non-adjuvanted, while the experimental group E includes all existing and new candidate adjuvants reported in clinical testing.

  • Fig. 2 Timeline of vaccines and adjuvants development. Shown is an historical view of the origin and development for some early and currently licensed human vaccines, the time needed to translate a scientific discovery (shown in red) into a safe and scalable vaccine technology (orange bars), the evolution of adjuvants, and of some supporting areas of science and technology. The blue bars represent the time needed to develop and implement one or more vaccines derived from the same technology. IPV, inactivated polio virus; OPV, oral polio virus; Hib, Haemophilus influenzae type b; HBV, hepatitis B virus; HPV, human papillomavirus. Reproduced from Rappuoli et al. (2014), with permission of the author and publisher, Proceedings of the National Academy of Sciences [2].

  • Fig. 3 A model of multiple mechanisms of adjuvanticity that converge toward dendritic cell (DC) activation. Adjuvants activate DCs either through direct interactions or through cellular intermediaries. toll-like receptor (TLR)-dependent adjuvants (in black) act directly on DCs; however, they can also activate other cells expressing the responding TLR. TLR-independent adjuvants (in red) can act directly either on DCs or on accessory cells. Aluminum salts, PLG, MSU, and Quil-A activate the NLRP3 inflammasome. Aluminum salts and MF59 act on monocytes, macrophages, or granulocytes to induce cytokines that generate a local immunostimulatory environment eventually leading to DC activation. In addition they also promote monocyte differentiation into DCs. MF59 can also activate muscle cells at injection site. It has been suggested that aluminum salts causes local necrosis of stromal cells leading to the release of uric acid, an endogenous danger signal that activates the inflammasome. Mast cell activators such as c48/80 can also act as adjuvants in a DC-dependent mechanism. iNKT activation by a-GalCer presented on CD1d leads to DC activation. Beta glucans activate DC through dectin-1 and ISCOMATRIX directly promotes cross-presentation in DCs [68]. So far, a role by TRLs has not been shown for the adjuvant action of polyelectrolytes. Reproduced from De Greggorio et al. (2009) with slight update and with permission of the author and publisher, Elsevier, Current Opinion in Immunology [68].

  • Fig. 4 Schematic chemical structure of chitosan.

  • Fig. 5 Schematic chemical structure of alginic acid.

  • Fig. 6 Schematic chemical structure of polyoxidonium.

  • Fig. 7 Schematic chemical structure of polyphosphazenes: PCPP (A), PCPP copolymer with oxyethylene groups (B), and PCEP (C). PCPP, poly[di(carboxylatophenoxy) phosphazene]; PCEP, poly[di(carboxylatoethylphenoxy)phosphazene].


Reference

1. van Panhuis WG, Grefenstette J, Jung SY, et al. Contagious diseases in the United States from 1888 to the present. N Engl J Med. 2013; 369:2152–2158.
Article
2. Rappuoli R, Pizza M, Del Giudice G, De Gregorio E. Vaccines, new opportunities for a new society. Proc Natl Acad Sci U S A. 2014; 111:12288–12293.
Article
3. World Health Organization. Global Vaccine Action Plan 2011-2020. Geneva: WHO;2013.
4. Black RE, Cousens S, Johnson HL, et al. Global, regional, and national causes of child mortality in 2008: a systematic analysis. Lancet. 2010; 375:1969–1987.
Article
5. Rusnak JM, Kortepeter MG, Hawley RJ, Anderson AO, Boudreau E, Eitzen E. Risk of occupationally acquired illnesses from biological threat agents in unvaccinated laboratory workers. Biosecur Bioterror. 2004; 2:281–293.
Article
6. World Health Organization. Global vaccine action plan: monitoring, evaluation and accountability annual report. Geneva: WHO;2014.
7. Mahadevan M, Navarro-Locsin G, Tan HK, et al. A review of the burden of disease due to otitis media in the Asia-Pacific. Int J Pediatr Otorhinolaryngol. 2012; 76:623–635.
Article
8. Memish ZA. Meningococcal disease and travel. Clin Infect Dis. 2002; 34:84–90.
Article
9. Hinman A. Eradication of vaccine-preventable diseases. Annu Rev Public Health. 1999; 20:211–229.
Article
10. Hennessey K, Schluter WW, Wang X, et al. Are we there yet? Assessing achievement of vaccine-preventable disease goals in WHO's Western Pacific Region. Vaccine. 2014; 32:4259–4266.
Article
11. Riedel S. Edward Jenner and the history of smallpox and vaccination. Proc (Bayl Univ Med Cent). 2005; 18:21–25.
Article
12. Henderson DA. The eradication of smallpox: an overview of the past, present, and future. Vaccine. 2011; 29:Suppl 4. D7–D9.
13. Anand A, Pallansch MA, Estivariz CF, Gary H, Wassilak SG. Estimating the likely coverage of inactivated poliovirus vaccine in routine immunization: evidence from demographic and health surveys. J Infect Dis. 2014; 210:Suppl 1. S465–S474.
Article
14. Klepac P, Metcalf CJ, McLean AR, Hampson K. Towards the endgame and beyond: complexities and challenges for the elimination of infectious diseases. Philos Trans R Soc Lond B Biol Sci. 2013; 368:20120137.
Article
15. Lievano F, Galea SA, Thornton M, et al. Measles, mumps, and rubella virus vaccine (M-M-RII): a review of 32 years of clinical and postmarketing experience. Vaccine. 2012; 30:6918–6926.
Article
16. Alicino C, Merlano C, Zappettini S, et al. Routine surveillance of adverse events following immunization as an important tool to monitor vaccine safety: the two-years' experience of the Liguria Region, Italy. Hum Vaccin Immunother. 2015; 11:91–94.
Article
17. Arguedas A, Soley C, Abdelnour A, et al. Assessment of the safety, tolerability and kinetics of the immune response to A/H1N1v vaccine formulations with and without adjuvant in healthy pediatric subjects from 3 through 17 years of age. Hum Vaccin. 2011; 7:58–66.
Article
18. Chang S, O'Connor PM, Slade BA, Woo EJ. U.S. Postlicensure safety surveillance for adolescent and adult tetanus, diphtheria and acellular pertussis vaccines: 2005-2007. Vaccine. 2013; 31:1447–1452.
Article
19. Cottin P, Niedrig M, Domingo C. Safety profile of the yellow fever vaccine Stamaril(R): a 17-year review. Expert Rev Vaccines. 2013; 12:1351–1368.
20. DeStefano F. Vaccine Safety Datalink Research Group. The Vaccine Safety Datalink project. Pharmacoepidemiol Drug Saf. 2001; 10:403–406.
Article
21. Lee CJ, Lee LH, Lu CH, Huang YJ, Chu ML. Safety monitoring in vaccine development and immunization. Acta Paediatr Taiwan. 2006; 47:7–13.
22. Macartney KK, Chiu C, Georgousakis M, Brotherton JM. Safety of human papillomavirus vaccines: a review. Drug Saf. 2013; 36:393–412.
Article
23. Tseng HF, Liu A, Sy L, et al. Safety of zoster vaccine in adults from a large managed-care cohort: a Vaccine Safety Datalink study. J Intern Med. 2012; 271:510–520.
Article
24. Nguyen TH, Vu MH, Nguyen VC, et al. A reduction in chronic hepatitis B virus infection prevalence among children in Vietnam demonstrates the importance of vaccination. Vaccine. 2014; 32:217–222.
Article
25. Teleb N, Lebo E, Ahmed H, et al. Progress toward measles elimination: Eastern Mediterranean Region, 2008-2012. MMWR Morb Mortal Wkly Rep. 2014; 63:511–515.
26. Kapoor R, Kottilil S. Strategies to eliminate HBV infection. Future Virol. 2014; 9:565–585.
Article
27. Andre FE, Booy R, Bock HL, et al. Vaccination greatly reduces disease, disability, death and inequity worldwide. Bull World Health Organ. 2008; 86:140–146.
Article
28. Garcon N, Hem S, Friede M. Evolution of adjuvants across the centuries. In : Plotkin S, Ornstein W, Offit P, editors. Vaccines. 6th ed. Philadelphia: Saunders Elsevier;2013. p. 58–70.
29. Glenny AT, Sudmersen HJ. Notes on the production of immunity to diphtheria toxin. J Hyg (Lond). 1921; 20:176–220.
Article
30. Emil von Behring: the founder of serum therapy [Internet]. Nobel Foundation;2001. cited 2014 Nov 18. Available from: http://www.nobelprize.org/nobel_prizes/medicine/laureates/1901/behring-article.html.
31. Kantha SS. The legacy of von Behring and Kitasato. Immunol Today. 1992; 13:374.
Article
32. Plotkin SL, Plotkin SA. A short history of vaccination. In : Plotkin S, Ornstein W, Offit P, editors. Vaccines. 6th ed. Philadelphia: Saunders Elsevier;2013. p. 1–13.
33. The tetanus cases in Camden, N.J. JAMA. 1901; 37:1539–1540.
34. U.S. Food and Drug Administration. Biologics centennial: 100 years of biologics regulation [Internet]. Silver Spring: FDA;2002. cited 2014 Nov 18. Available from: http://www.fda.gov/AboutFDA/WhatWeDo/History/ProductRegulation/SelectionsFromFDLIUpdateSeriesonFDAHistory/ucm091754.htm.
35. Dudzinski DM. Reflections on historical, scientific, and legal issues relevant to designing approval pathways for generic versions of recombinant protein-based therapeutics and monoclonal antibodies. Food Drug Law J. 2005; 60:143–260.
36. Ramon G. Regarding the flocculant ability and immunizing properties of a diphtheria toxin rendered atoxic. C R Hebd Seances Acad Sci. 1923; 177:1338–1340.
37. Glenny AT, Pope CG, Waddington H, Wallace U. Immunological notes: XVII-XXIV. J Pathol Bacteriol. 1926; 29:31–40.
Article
38. Park WH, Schroder MC. Diphtheria toxin-antitoxin and toxoid: a comparison. Am J Public Health Nations Health. 1932; 22:7–16.
39. Baker JN, Gill DG. Precipitated toxoid as an immunizing agent against diphtheria. Am J Public Health Nations Health. 1934; 24:22–24.
Article
40. Ericsson H. Purification and adsorption of diphtheria toxoid. Nature. 1946; 158:350.
Article
41. Holt LB. Purified precipitated diphtheria toxoid of constant composition. Lancet. 1947; 1:282–285.
42. Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-first century society. Nat Rev Immunol. 2011; 11:865–872.
Article
43. Freund J. The effect of paraffin oil and mycobacteria on antibody formation and sensitization: a review. Am J Clin Pathol. 1951; 21:645–656.
Article
44. Petrovsky N, Aguilar JC. Vaccine adjuvants: current state and future trends. Immunol Cell Biol. 2004; 82:488–496.
Article
45. Stuart-Harris CH. Adjuvant influenza vaccines. Bull World Health Organ. 1969; 41:617–621.
46. O'Hagan DT, Ott GS, Nest GV, Rappuoli R, Giudice GD. The history of MF59((R)) adjuvant: a phoenix that arose from the ashes. Expert Rev Vaccines. 2013; 12:13–30.
47. Morel S, Didierlaurent A, Bourguignon P, et al. Adjuvant System AS03 containing alpha-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011; 29:2461–2473.
Article
48. Caillet C, Piras F, Bernard MC, et al. AF03-adjuvanted and non-adjuvanted pandemic influenza A (H1N1) 2009 vaccines induce strong antibody responses in seasonal influenza vaccine-primed and unprimed mice. Vaccine. 2010; 28:3076–3079.
Article
49. Fox CB, Baldwin SL, Duthie MS, Reed SG, Vedvick TS. Immunomodulatory and physical effects of oil composition in vaccine adjuvant emulsions. Vaccine. 2011; 29:9563–9572.
Article
50. Rietschel ET, Cavaillon JM. Richard Pfeiffer and Alexandre Besredka: creators of the concept of endotoxin and anti-endotoxin. Microbes Infect. 2003; 5:1407–1414.
Article
51. Nauts HC, Swift WE, Coley BL. The treatment of malignant tumors by bacterial toxins as developed by the late William B. Coley, M.D., reviewed in the light of modern research. Cancer Res. 1946; 6:205–216.
52. Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Immunol. 2001; 1:135–145.
Article
53. Medzhitov R, Janeway CA Jr. Innate immunity: impact on the adaptive immune response. Curr Opin Immunol. 1997; 9:4–9.
Article
54. Reed SG, Orr MT, Fox CB. Key roles of adjuvants in modern vaccines. Nat Med. 2013; 19:1597–1608.
Article
55. De Gregorio E, Rappuoli R. From empiricism to rational design: a personal perspective of the evolution of vaccine development. Nat Rev Immunol. 2014; 14:505–514.
Article
56. Petrovsky N. Vaccine adjuvants: in search of new paradigms. Interview by Jenaid Rees. Expert Rev Vaccines. 2013; 12:723–726.
57. Koff WC, Burton DR, Johnson PR, et al. Accelerating next-generation vaccine development for global disease prevention. Science. 2013; 340:1232910.
Article
58. Brito LA, Malyala P, O'Hagan DT. Vaccine adjuvant formulations: a pharmaceutical perspective. Semin Immunol. 2013; 25:130–145.
Article
59. Li L, Saade F, Petrovsky N. The future of human DNA vaccines. J Biotechnol. 2012; 162:171–182.
Article
60. Williams A, Flavell RA, Eisenbarth SC. The role of NOD-like receptors in shaping adaptive immunity. Curr Opin Immunol. 2010; 22:34–40.
Article
61. Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010; 33:492–503.
Article
62. Carter D, Reed SG. Role of adjuvants in modeling the immune response. Curr Opin HIV AIDS. 2010; 5:409–413.
Article
63. Rappuoli R. Reverse vaccinology. Curr Opin Microbiol. 2000; 3:445–450.
Article
64. Vos Q, Lees A, Wu ZQ, Snapper CM, Mond JJ. B-cell activation by T-cell-independent type 2 antigens as an integral part of the humoral immune response to pathogenic microorganisms. Immunol Rev. 2000; 176:154–170.
Article
65. Wetzler LM. Innate immune function of the neisserial porins and the relationship to vaccine adjuvant activity. Future Microbiol. 2010; 5:749–758.
Article
66. Massari P, Gunawardana J, Liu X, Wetzler LM. Meningococcal porin PorB prevents cellular apoptosis in a toll-like receptor 2- and NF-kappaB-independent manner. Infect Immun. 2010; 78:994–1003.
Article
67. Taylor DN, Treanor JJ, Sheldon EA, et al. Development of VAX128, a recombinant hemagglutinin (HA) influenza-flagellin fusion vaccine with improved safety and immune response. Vaccine. 2012; 30:5761–5769.
Article
68. De Gregorio E, D'Oro U, Wack A. Immunology of TLR-independent vaccine adjuvants. Curr Opin Immunol. 2009; 21:339–345.
Article
69. Kay E, Scotland RS, Whiteford JR. Toll-like receptors: Role in inflammation and therapeutic potential. Biofactors. 2014; 40:284–294.
Article
70. Cervantes JL, Weinerman B, Basole C, Salazar JC. TLR8: the forgotten relative revindicated. Cell Mol Immunol. 2012; 9:434–438.
Article
71. Carta S, Tassi S, Pettinati I, Delfino L, Dinarello CA, Rubartelli A. The rate of interleukin-1beta secretion in different myeloid cells varies with the extent of redox response to Toll-like receptor triggering. J Biol Chem. 2011; 286:27069–27080.
Article
72. Isakov E, Weisman-Shomer P, Benhar M. Suppression of the pro-inflammatory NLRP3/interleukin-1beta pathway in macrophages by the thioredoxin reductase inhibitor auranofin. Biochim Biophys Acta. 2014; 1840:3153–3161.
Article
73. Dasu MR, Isseroff RR. Toll-like receptors in wound healing: location, accessibility, and timing. J Invest Dermatol. 2012; 132:1955–1958.
Article
74. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010; 10:826–837.
Article
75. Muralidharan S, Mandrekar P. Cellular stress response and innate immune signaling: integrating pathways in host defense and inflammation. J Leukoc Biol. 2013; 94:1167–1184.
Article
76. Mount A, Koernig S, Silva A, Drane D, Maraskovsky E, Morelli AB. Combination of adjuvants: the future of vaccine design. Expert Rev Vaccines. 2013; 12:733–746.
Article
77. Kuroda E, Coban C, Ishii KJ. Particulate adjuvant and innate immunity: past achievements, present findings, and future prospects. Int Rev Immunol. 2013; 32:209–220.
Article
78. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011; 469:221–225.
Article
79. Flach TL, Ng G, Hari A, et al. Alum interaction with dendritic cell membrane lipids is essential for its adjuvanticity. Nat Med. 2011; 17:479–487.
Article
80. Seubert A, Calabro S, Santini L, et al. Adjuvanticity of the oil-in-water emulsion MF59 is independent of Nlrp3 inflammasome but requires the adaptor protein MyD88. Proc Natl Acad Sci U S A. 2011; 108:11169–11174.
Article
81. Wilson NS, Duewell P, Yang B, et al. Inflammasome-dependent and -independent IL-18 production mediates immunity to the ISCOMATRIX adjuvant. J Immunol. 2014; 192:3259–3268.
Article
82. Baroja-Mazo A, Martin-Sanchez F, Gomez AI, et al. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014; 15:738–748.
Article
83. 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. 2009; 183:4403–4414.
Article
84. Hutchison S, Benson RA, Gibson VB, Pollock AH, Garside P, Brewer JM. Antigen depot is not required for alum adjuvanticity. FASEB J. 2012; 26:1272–1279.
Article
85. Watkinson A, Soliakov A, Ganesan A, et al. Increasing the potency of an alhydrogel-formulated anthrax vaccine by minimizing antigen-adjuvant interactions. Clin Vaccine Immunol. 2013; 20:1659–1668.
Article
86. Iyer V, Hu L, Liyanage MR, et al. Preformulation characterization of an aluminum salt-adjuvanted trivalent recombinant protein-based vaccine candidate against Streptococcus pneumoniae. J Pharm Sci. 2012; 101:3078–3090.
Article
87. Soliakov A, Kelly IF, Lakey JH, Watkinson A. Anthrax sub-unit vaccine: the structural consequences of binding rPA83 to Alhydrogel(R). Eur J Pharm Biopharm. 2012; 80:25–32.
Article
88. Exley C, Siesjo P, Eriksson H. The immunobiology of aluminium adjuvants: how do they really work. Trends Immunol. 2010; 31:103–109.
Article
89. Lu F, Hogenesch H. Kinetics of the inflammatory response following intramuscular injection of aluminum adjuvant. Vaccine. 2013; 31:3979–3986.
Article
90. Rimaniol AC, Gras G, Verdier F, et al. Aluminum hydroxide adjuvant induces macrophage differentiation towards a specialized antigen-presenting cell type. Vaccine. 2004; 22:3127–3135.
Article
91. Mold M, Eriksson H, Siesjo P, Darabi A, Shardlow E, Exley C. Unequivocal identification of intracellular aluminium adjuvant in a monocytic THP-1 cell line. Sci Rep. 2014; 4:6287.
Article
92. He Y. Vaccine adjuvant informatics: from data integration and analysis to rational vaccine adjuvant design. Front Immunol. 2014; 5:32.
Article
93. Sayers S, Ulysse G, Xiang Z, He Y. Vaxjo: a web-based vaccine adjuvant database and its application for analysis of vaccine adjuvants and their uses in vaccine development. J Biomed Biotechnol. 2012; 2012:831486.
Article
94. Hartung DM, Zarin DA, Guise JM, McDonagh M, Paynter R, Helfand M. Reporting discrepancies between the ClinicalTrials.gov results database and peer-reviewed publications. Ann Intern Med. 2014; 160:477–483.
Article
95. Zarin DA, Tse T, Williams RJ, Califf RM, Ide NC. The ClinicalTrials.gov results database--update and key issues. N Engl J Med. 2011; 364:852–860.
Article
96. Levast B, Berri M, Wilson HL, Meurens F, Salmon H. Development of gut immunoglobulin A production in piglet in response to innate and environmental factors. Dev Comp Immunol. 2014; 44:235–244.
Article
97. Gupta RK, Rost BE, Relyveld E, Siber GR. Adjuvant properties of aluminum and calcium compounds. Pharm Biotechnol. 1995; 6:229–248.
Article
98. Hem SL, White JL. Structure and properties of aluminum-containing adjuvants. Pharm Biotechnol. 1995; 6:249–276.
Article
99. Li X, Aldayel AM, Cui Z. Aluminum hydroxide nanoparticles show a stronger vaccine adjuvant activity than traditional aluminum hydroxide microparticles. J Control Release. 2014; 173:148–157.
Article
100. Barbaud A, Deschildre A, Waton J, Raison-Peyron N, Trechot P. Hypersensitivity and vaccines: an update. Eur J Dermatol. 2013; 23:135–141.
Article
101. Pittman PR, Kim-Ahn G, Pifat DY, et al. Anthrax vaccine: immunogenicity and safety of a dose-reduction, route-change comparison study in humans. Vaccine. 2002; 20:1412–1420.
Article
102. Hogenesch H. Mechanism of immunopotentiation and safety of aluminum adjuvants. Front Immunol. 2012; 3:406.
Article
103. Jiang D, Premachandra GS, Johnston C, Hem SL. Structure and adsorption properties of commercial calcium phosphate adjuvant. Vaccine. 2004; 23:693–698.
Article
104. 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
105. 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
106. Schinwald A, Donaldson K. Use of back-scatter electron signals to visualise cell/nanowires interactions in vitro and in vivo: frustrated phagocytosis of long fibres in macrophages and compartmentalisation in mesothelial cells in vivo. Part Fibre Toxicol. 2012; 9:34.
Article
107. Williams GR, Fierens K, Preston SG, et al. Immunity induced by a broad class of inorganic crystalline materials is directly controlled by their chemistry. J Exp Med. 2014; 211:1019–1025.
Article
108. Sun B, Ji Z, Liao YP, et al. Engineering an effective immune adjuvant by designed control of shape and crystallinity of aluminum oxyhydroxide nanoparticles. ACS Nano. 2013; 7:10834–10849.
Article
109. Mori A, Oleszycka E, Sharp FA, et al. The vaccine adjuvant alum inhibits IL-12 by promoting PI3 kinase signaling while chitosan does not inhibit IL-12 and enhances Th1 and Th17 responses. Eur J Immunol. 2012; 42:2709–2719.
Article
110. Neumann S, Burkert K, Kemp R, Rades T, Rod Dunbar P, Hook S. Activation of the NLRP3 inflammasome is not a feature of all particulate vaccine adjuvants. Immunol Cell Biol. 2014; 92:535–542.
Article
111. Lu F, Boutselis I, Borch RF, Hogenesch H. Control of antigen-binding to aluminum adjuvants and the immune response with a novel phosphonate linker. Vaccine. 2013; 31:4362–4367.
Article
112. Lindblad EB. Aluminium compounds for use in vaccines. Immunol Cell Biol. 2004; 82:497–505.
Article
113. Kabanov VA. From synthetic polyelectrolytes to polymer-subunit vaccines. Pure Appl Chem. 2004; 76:1659–1677.
Article
114. Andrianov AK. Polyphosphazene vaccine delivery vehicles: state of development and perspectives. In : Andrianov AK, editor. Polyphosphazenes for biomedical applications. Hoboken: John Wiley & Sons Inc.;2009. p. 47–63.
115. Muzzarelli RA. Chitins and chitosans as immunoadjuvants and non-allergenic drug carriers. Mar Drugs. 2010; 8:292–312.
Article
116. Amidi M, Mastrobattista E, Jiskoot W, Hennink WE. Chitosan-based delivery systems for protein therapeutics and antigens. Adv Drug Deliv Rev. 2010; 62:59–82.
Article
117. Jayakumar R, Prabaharan M, Sudheesh Kumar PT, Nair SV, Tamura H. Biomaterials based on chitin and chitosan in wound dressing applications. Biotechnol Adv. 2011; 29:322–337.
Article
118. You J, Li W, Yu C, et al. Amphiphilically modified chitosan cationic nanoparticles for drug delivery. J Nanopart Res. 2013; 15:2123.
Article
119. Nishimura K, Nishimura S, Nishi N, Saiki I, Tokura S, Azuma I. Immunological activity of chitin and its derivatives. Vaccine. 1984; 2:93–99.
Article
120. Lee CG. Chitin, chitinases and chitinase-like proteins in allergic inflammation and tissue remodeling. Yonsei Med J. 2009; 50:22–30.
Article
121. Lee CG, Da Silva CA, Dela Cruz CS, et al. Role of chitin and chitinase/chitinase-like proteins in inflammation, tissue remodeling, and injury. Annu Rev Physiol. 2011; 73:479–501.
Article
122. Zaharoff DA, Rogers CJ, Hance KW, Schlom J, Greiner JW. Chitosan solution enhances both humoral and cell-mediated immune responses to subcutaneous vaccination. Vaccine. 2007; 25:2085–2094.
Article
123. Zaharoff DA, Rogers CJ, Hance KW, Schlom J, Greiner JW. Chitosan solution enhances the immunoadjuvant properties of GM-CSF. Vaccine. 2007; 25:8673–8686.
Article
124. Scherliess R, Buske S, Young K, Weber B, Rades T, Hook S. In vivo evaluation of chitosan as an adjuvant in subcutaneous vaccine formulations. Vaccine. 2013; 31:4812–4819.
Article
125. Vasiliev YM. Chitosan-based vaccine adjuvants: incomplete characterization complicates preclinical and clinical evaluation. Expert Rev Vaccines. 2015; 14:37–53.
Article
126. Murano E. Use of natural polysaccharides in the microencapsulation techniques. J Appl Ichthyol. 1998; 14:245–249.
Article
127. Dobakhti F, Naghibi T, Taghikhani M, et al. Adjuvanticity effect of sodium alginate on subcutaneously injected BCG in BALB/c mice. Microbes Infect. 2009; 11:296–301.
Article
128. Mata E, Igartua M, Patarroyo ME, Pedraz JL, Hernandez RM. Enhancing immunogenicity to PLGA microparticulate systems by incorporation of alginate and RGD-modified alginate. Eur J Pharm Sci. 2011; 44:32–40.
Article
129. Dyakonova VA, Dambaeva SV, Pinegin BV, Khaitov RM. Study of interaction between the polyoxidonium immunomodulator and the human immune system cells. Int Immunopharmacol. 2004; 4:1615–1623.
Article
130. Dambaeva SV, Mazurov DV, Golubeva NM, D'Yakonova VA, Pinegin BV, Khaitov RM. Effect of polyoxidonium on the phagocytic activity of human peripheral blood leukocytes. Russ J Immunol. 2003; 8:53–60.
131. Denisov AA, Korobovtseva YS, Karpova OM, et al. Immunopotentiation of live brucellosis vaccine by adjuvants. Vaccine. 2010; 28:Suppl 5. F17–F22.
Article
132. Toptygina A, Semikina E, Alioshkin V. Influence of an immunopotentiator polyoxidonium on cytokine profile and antibody production in children vaccinated with Priorix. Arch Physiol Biochem. 2012; 118:197–203.
Article
133. Andrianov AK, Marin A, Roberts BE. Polyphosphazene polyelectrolytes: a link between the formation of noncovalent complexes with antigenic proteins and immunostimulating activity. Biomacromolecules. 2005; 6:1375–1379.
Article
134. Andrianov AK, Chen J, LeGolvan MP. Poly(dichlorophosphazene) as a precursor for biologically active polyphosphazenes: synthesis, characterization, and stabilization. Macromolecules. 2004; 37:414–420.
Article
135. Andrianov AK, LeGolvan MP. Characterization of poly [di(carboxylatophenoxy)-phosphazene] by an aqueous gel permeation chromatography. J Appl Polym Sci. 1996; 60:2289–2295.
Article
136. Andrianov AK, Svirkin YY, LeGolvan MP. Synthesis and biologically relevant properties of polyphosphazene polyacids. Biomacromolecules. 2004; 5:1999–2006.
Article
137. Andrianov AK, DeCollibus DP, Gillis HA, Kha HH, Marin A. Polyphosphazene immunoadjuvants for intradermal vaccine delivery. In : Andrianov AK, editor. Polyphosphazene for biomedical applications. Hoboken: John Wiley & Sons Inc.;2009. p. 101–116.
138. Andrianov AK, Decollibus DP, Marin A, Webb A, Griffin Y, Webby RJ. PCPP-formulated H5N1 influenza vaccine displays improved stability and dose-sparing effect in lethal challenge studies. J Pharm Sci. 2011; 100:1436–1443.
Article
139. Payne LG, Jenkins SA, Woods AL, et al. Poly[di(carboxylatophenoxy)phosphazene] (PCPP) is a potent immunoadjuvant for an influenza vaccine. Vaccine. 1998; 16:92–98.
Article
140. Payne LG, Van Nest G, Barchfeld GL, Siber GR, Gupta RK, Jenkins SA. PCPP as a parenteral adjuvant for diverse antigens. Dev Biol Stand. 1998; 92:79–87.
141. Mutwiri G, Benjamin P, Soita H, et al. Poly[di(sodium carboxylatoethylphenoxy)phosphazene] (PCEP) is a potent enhancer of mixed Th1/Th2 immune responses in mice immunized with influenza virus antigens. Vaccine. 2007; 25:1204–1213.
Article
142. Lu Y, Salvato MS, Pauza CD, et al. Utility of SHIV for testing HIV-1 vaccine candidates in macaques. J Acquir Immune Defic Syndr Hum Retrovirol. 1996; 12:99–106.
Article
143. Wu JY, Wade WF, Taylor RK. Evaluation of cholera vaccines formulated with toxin-coregulated pilin peptide plus polymer adjuvant in mice. Infect Immun. 2001; 69:7695–7702.
Article
144. Le Cam NN, Ronco J, Francon A, Blondeau C, Fanget B. Adjuvants for influenza vaccine. Res Immunol. 1998; 149:19–23.
Article
145. Plotkin SA. Vaccines: past, present and future. Nat Med. 2005; 11:4 Suppl. S5–11.
Article
146. Ison MG, Mills J, Openshaw P, Zambon M, Osterhaus A, Hayden F. Current research on respiratory viral infections: Fourth International Symposium. Antiviral Res. 2002; 55:227–278.
Article
147. Kim JH, Kirsch EA, Gilliam B. A phase I, open label, dose ranging trial of the Pasteur Merieux Connaught (PMC) oligomeric HIV-1 Gp160mn/LAI-2 vaccine in HIV seronegative adults. Abstracts of the 37th Annual Meeting of the Infectious Diseases Society of America. Philadelphia, PA, USA: 1999. 11. 18. 21.
148. Mutwiri G, Benjamin P, Soita H, Babiuk LA. Co-administration of polyphosphazenes with CpG oligodeoxynucleotides strongly enhances immune responses in mice immunized with hepatitis B virus surface antigen. Vaccine. 2008; 26:2680–2688.
Article
149. Istrate C, Hinkula J, Charpilienne A, et al. Parenteral administration of RF 8-2/6/7 rotavirus-like particles in a one-dose regimen induce protective immunity in mice. Vaccine. 2008; 26:4594–4601.
Article
150. Palmer CD, Ninkovic J, Prokopowicz ZM, et al. The effect of stable macromolecular complexes of ionic polyphosphazene on HIV Gag antigen and on activation of human dendritic cells and presentation to T-cells. Biomaterials. 2014; 35:8876–8886.
Article
151. Andrianov AK, Sargent JR, Sule SS, et al. Synthesis, physico-chemical properties and immunoadjuvant activity of water-soluble phosphazene polyacids. J Bioact Compat Polym. 1998; 13:243–256.
Article
152. Andrianov AK, Marin A, Chen J. Synthesis, properties, and biological activity of poly[di(sodium carboxylatoethylphenoxy)phosphazene]. Biomacromolecules. 2006; 7:394–399.
Article
153. Johansson E, Istrate C, Charpilienne A, et al. Amount of maternal rotavirus-specific antibodies influence the outcome of rotavirus vaccination of newborn mice with virus-like particles. Vaccine. 2008; 26:778–785.
Article
154. Awate S, Wilson HL, Lai K, Babiuk LA, Mutwiri G. Activation of adjuvant core response genes by the novel adjuvant PCEP. Mol Immunol. 2012; 51:292–303.
Article
155. Awate S, Wilson HL, Singh B, Babiuk LA, Mutwiri G. The adjuvant PCEP induces recruitment of myeloid and lymphoid cells at the injection site and draining lymph node. Vaccine. 2014; 32:2420–2427.
Article
156. Andrianov AK, DeCollibus DP, Gillis HA, et al. Poly[di (carboxylatophenoxy)phosphazene] is a potent adjuvant for intradermal immunization. Proc Natl Acad Sci U S A. 2009; 106:18936–18941.
Article
157. Andrianov AK, Marin A, DeCollibus DP. Microneedles with intrinsic immunoadjuvant properties: microfabrication, protein stability, and modulated release. Pharm Res. 2011; 28:58–65.
Article
158. Vaccines Europe. The vaccine industry in figures [Internet]. Brussels: EFPIA/Vaccines Europe. 2014. cited 2014 Nov 18. Available from: http://www.vaccineseurope.eu/about-vaccines-europe/vaccines-europe-in-figures/.
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