899
Views
49
CrossRef citations to date
0
Altmetric
Review

Chitosan-based vaccine adjuvants: incomplete characterization complicates preclinical and clinical evaluation

References

  • Otterlei M, Vårum KM, Ryan L, Espevik T. Characterization of binding and TNF-alpha-inducing ability of chitosans on monocytes: the involvement of CD14. Vaccine 1994;12(9):825-32
  • Anthonsen MW, Vårum KM, Smidsrød O. Solution properties of chitosans: conformation and chain stiffness of chitosans with different degrees of N-acetylation. Carbohydr Polym 1993;22(3):193-201
  • Illum L, Jabbal-Gill I, Hinchcliffe M, et al. Chitosan as a novel nasal delivery system for vaccines. Adv Drug Deliv Rev 2001;51(1-3):81-96
  • Horowitz ST, Roseman S, Blumenthal HJ. The preparation of glucosamine oligosaccharides. I. Separation. J Am Chem Soc 1957;79(18):5046-9
  • Lieder R, Gaware VS, Thormodsson F, et al. Endotoxins affect bioactivity of chitosan derivatives in cultures of bone marrow-derived human mesenchymal stem cells. Acta Biomater 2013;9(1):4771-8
  • Zhang H, Du Y, Yu X, et al. Preparation of chitooligosaccharides from chitosan by a complex enzyme. Carbohydr Res 1999;320(3-4):257-60
  • Arabski M, Lisowska H, Lankoff A, et al. The properties of chitosan complexes with smooth and rough forms of lipopolysaccharides on CHO-K1 cells. Carbohydr Polym 2013;97(2):284-92
  • Knaul JZ, Kasaai MR, Bui VT, Creber KAM. Characterization of deacetylated chitosan and chitosan molecular weight review. Can J Chem 1998;76(11):1699-706
  • Maghami GG, Roberts GAF. Evaluation of the viscometric constants for chitosan. Makromol Chem 1988;189(1):195-200
  • Chitosan vaccine. Available from: www.ncbi.nlm.nih.gov/pubmed/?term=chitosan+vaccine
  • Slütter B, Jiskoot W. Dual role of CpG as immune modulator and physical crosslinker in ovalbumin loaded N-trimethyl chitosan (TMC) nanoparticles for nasal vaccination. J Control Release 2010;148(1):117-21
  • Bal SM, Slütter B, van Riet E, et al. Efficient induction of immune responses through intradermal vaccination with N-trimethyl chitosan containing antigen formulations. J Control Release 2010;142(3):374-83
  • Ghendon Y, Markushin S, Krivtsov G, Akopova I. Chitosan as an adjuvant for parenterally administered inactivated influenza vaccines. Arch Virol 2008;153(5):831-7
  • Ghendon Y, Markushin S, Vasiliev Y, et al. Evaluation of properties of chitosan as an adjuvant for inactivated influenza vaccines administered parenterally. J Med Virol 2009;81(3):494-506
  • Wang X, Zhang W, Liu F, et al. Intranasal immunization with live attenuated influenza vaccine plus chitosan as an adjuvant protects mice against homologous and heterologous virus challenge. Arch Virol 2012;157(8):1451-61
  • Sigma-Alorich. Available from: www.sigmaaldrich.com/catalog/search?interface=All&term=chitosan
  • Li D, Chen JL, Zhang H, et al. Improvement of the immunity of pig to Hog cholera vaccine by recombinant plasmid with porcine interleukin-6 gene and CpG motifs. Vaccine 2011;29(22):3888-94
  • Zhang H, Cheng C, Zheng M, et al. Enhancement of immunity to an Escherichia coli vaccine in mice orally inoculated with a fusion gene encoding porcine interleukin 4 and 6. Vaccine 2007;25(41):7094-101
  • Roy K, Mao HQ, Huang SK, Leong KW. Oral gene delivery with chitosan-DNA nanoparticles generates immunologic protection in a murine model of peanut allergy. Nat Med 1999;5(4):387-91
  • Lemke CD, Graham JB, Geary SM, et al. Chitosan is a surprising negative modulator of cytotoxic CD8+ T cell responses elicited by adenovirus cancer vaccines. Mol Pharm 2011;8(5):1652-61
  • Jean M, Smaoui F, Lavertu M, et al. Chitosan-plasmid nanoparticle formulations for IM and SC delivery of recombinant FGF-2 and PDGF-BB or generation of antibodies. Gene Ther 2009;16(9):1097-110
  • Nguyen S, Winnik FM, Buschmann MD. Improved reproducibility in the determination of the molecular weight of chitosan by analytical size exclusion chromatography. Carbohydr Polym 2009;75(3):528-33
  • Gordon S, Saupe A, McBurney W, et al. Comparison of chitosan nanoparticles and chitosan hydrogels for vaccine delivery. J Pharm Pharmacol 2008;60(12):1591-600
  • Scherließ R, Buske S, Young K, et al. In vivo evaluation of chitosan as an adjuvant in subcutaneous vaccine formulations. Vaccine 2013;31(42):4812-19
  • Xie Y, Zhou NJ, Gong YF, et al. Th immune response induced by H pylori vaccine with chitosan as adjuvant and its relation to immune protection. World J Gastroenterol 2007;13(10):1547-53
  • Ghendon Y, Markushin S, Akopova I, et al. Chitosan as an adjuvant for poliovaccine. J Med Virol 2011;83(5):847-52
  • Jabbal-Gill I, Fisher AN, Rappuoli R, et al. Stimulation of mucosal and systemic antibody responses against Bordetella pertussis filamentous haemagglutinin and recombinant pertussis toxin after nasal administration with chitosan in mice. Vaccine 1998;16(20):2039-46
  • Hashem FM, Fahmy SA, El-Sayed AM, Al-Sawahli MM. Development and evaluation of chitosan microspheres for tetanus, diphtheria and divalent vaccines: a comparative study of subcutaneous and intranasal administration in mice. Pharm Dev Technol 2013;18(5):1175-85
  • Coeshott CM, Smithson SL, Verderber E, et al. Pluronic F127-based systemic vaccine delivery systems. Vaccine 2004;22(19):2396-405
  • Buffa V, Klein K, Fischetti L, Shattock RJ. Evaluation of TLR agonists as potential mucosal adjuvants for HIV gp140 and tetanus toxoid in mice. PLoS One 2012;7(12):e50529
  • de Geus ED, van Haarlem DA, Poetri ON, et al. A lack of antibody formation against inactivated influenza virus after aerosol vaccination in presence or absence of adjuvantia. Vet Immunol Immunopathol 2011;143(1-2):143-7
  • Degen WGJ, van Zuilekom HI, Scholtes NC, et al. Potentiation of humoral immune responses to vaccine antigens by recombinant chicken IL-18 (rChIL-18). Vaccine 2005;23(33):4212-18
  • Khatri K, Goyal AK, Gupta PN, et al. Plasmid DNA loaded chitosan nanoparticles for nasal mucosal immunization against hepatitis B. Int J Pharm 2008;354(1-2):235-41
  • Read RC, Naylor SC, Potter CW, et al. Effective nasal influenza vaccine delivery using chitosan. Vaccine 2005;23(35):4367-74
  • Mills KH, Cosgrove C, McNeela EA, et al. Protective levels of diphtheria-neutralizing antibody induced in healthy volunteers by unilateral priming-boosting intranasal immunization associated with restricted ipsilateral mucosal secretory immunoglobulin A. Infect Immun 2003;71(2):726-32
  • McNeela EA, Jabbal-Gill I, Illum L, et al. Intranasal immunization with genetically detoxified diphtheria toxin induces T cell responses in humans: enhancement of Th2 responses and toxin-neutralizing antibodies by formulation with chitosan. Vaccine 2004;22(8):909-14
  • El-Kamary SS, Pasetti MF, Mendelman PM, et al. Adjuvanted intranasal Norwalk virus-like particle vaccine elicits antibodies and antibody-secreting cells that express homing receptors for mucosal and peripheral lymphoid tissues. J Infect Dis 2010;202(11):1649-58. Erratum in: J Infect Dis 2011;203(7):1036
  • Atmar RL, Bernstein DI, Harro CD, et al. Norovirus vaccine against experimental human Norwalk Virus illness. N Engl J Med 2011;365(23):2178-87
  • Huo Z, Sinha R, McNeela EA, et al. Induction of protective serum meningococcal bactericidal and diphtheria-neutralizing antibodies and mucosal immunoglobulin A in volunteers by nasal insufflations of the Neisseria meningitidis serogroup C polysaccharide-CRM197 conjugate vaccine mixed with chitosan. Infect Immun 2005;73(12):8256-65
  • VandeVord PJ, Matthew HW, DeSilva SP, et al. Evaluation of the biocompatibility of a chitosan scaffold in mice. J Biomed Mater Res 2002;59(3):585-90
  • Han HD, Nam DE, Seo DH, et al. Preparation and biodegradation of thermosensitive chitosan hydrogel as a function of pH and temperature. Macromol Res 2004;12(5):507-11
  • Dehghan S, Kheiri MT, Tabatabaiean M, et al. Dry-powder form of chitosan nanospheres containing influenza virus and adjuvants for nasal immunization. Arch Pharm Res 2013;36(8):981-92
  • Wischke C, Borchert HH, Zimmermann J, et al. Stable cationic microparticles for enhanced model antigen delivery to dendritic cells. J Control Release 2006;114(3):359-68
  • Heuking S, Adam-Malpel S, Sublet E, et al. Stimulation of human macrophages (THP-1) using Toll-like receptor-2 (TLR-2) agonist decorated nanocarriers. J Drug Target 2009;17(8):662-70
  • Romøren K, Thu BJ, Evensen Ø. Immersion delivery of plasmid DNA. II. A study of the potentials of a chitosan based delivery system in rainbow trout (Oncorhynchus mykiss) fry. J Control Release 2002;85(1-3):215-25
  • Huang YC, Vieira A, Huang KL, et al. Pulmonary inflammation caused by chitosan microparticles. J Biomed Mater Res A 2005;75(2):283-7
  • Huang YC, Yeh MK, Cheng SN, Chiang CH. The characteristics of betamethasone-loaded chitosan microparticles by spray-drying method. J Microencapsul 2003;20(4):459-72
  • Litamoi JK, Ayelet G, Rweyemamu MM. Evaluation of the xerovac process for the preparation of heat tolerant contagious bovine pleuropneumonia (CBPP) vaccine. Vaccine 2005;23(20):2573-9
  • Kissmann J, Ausar SF, Foubert TR, et al. Physical stabilization of Norwalk virus-like particles. J Pharm Sci 2008;97(10):4208-18
  • Calvo P, Remuñan-López C, Vila-Jato JL, Alonso MJ. Chitosan and chitosan/ethylene oxide-propylene oxide block copolymer nanoparticles as novel carriers for proteins and vaccines. Pharm Res 1997;14(10):1431-6
  • Mi FL, Shyu SS, Chen CT, Schoung JY. Porous chitosan microsphere for controlling the antigen release of Newcastle disease vaccine: preparation of antigen-adsorbed microsphere and in vitro release. Biomaterials 1999;20(17):1603-12
  • Vicente S, Goins BA, Sanchez A, et al. Biodistribution and lymph node retention of polysaccharide-based immunostimulating nanocapsules. Vaccine 2014;32(15):1685-92
  • Kirby DJ, Kaur R, Agger EM, et al. Developing solid particulate vaccine adjuvants: surface bound antigen favours a humoural response, whereas entrapped antigen shows a tendency for cell mediated immunity. Curr Drug Deliv 2013;10(3):268-78
  • Park J, Babensee JE. Differential functional effects of biomaterials on dendritic cell maturation. Acta Biomater 2012;8(10):3606-17
  • Koppolu B, Zaharoff DA. The effect of antigen encapsulation in chitosan particles on uptake, activation and presentation by antigen presenting cells. Biomaterials 2013;34(9):2359-69
  • Verheul RJ, Hagenaars N, van Es T, et al. A step-by-step approach to study the influence of N-acetylation on the adjuvanticity of N,N,N-trimethyl chitosan (TMC) in an intranasal nanoparticulate influenza virus vaccine. Eur J Pharm Sci 2012;45(4):467-74
  • Nishimura K, Nishimura S, Nishi N, et al. Immunological activity of chitin and its derivatives. Vaccine 1984;2(1):93-9
  • Jeong HJ, Koo HN, Oh EY, et al. Nitric oxide production by high molecular weight water-soluble chitosan via nuclear factor-κB activation. Int J Immunopharmacol 2000;22(11):923-33
  • Dou J, Tan C, Du Y, et al. Effects of chitooligosaccharides on rabbit neutrophils in vitro. Carbohydr Polym 2007;69(2):209-13
  • Démoulins T, Bassi I, Thomann-Harwood L, et al. Alginate-coated chitosan nanogel capacity to modulate the effect of TLR ligands on blood dendritic cells. Nanomedicine 2013;9(6):806-17
  • Li H, Willingham SB, Ting JPY, Re F. Cutting edge: inflammasome activation by alum and alum’s adjuvant effect are mediated by NLRP3. J Immunol 2008;181(1):17-21
  • Ji Q, Deng J, Yu X, et al. Modulation of pro-inflammatory mediators in LPS-stimulated human periodontal ligament cells by chitosan and quaternized chitosan. Carbohydr Polym 2013;92(1):824-9
  • Chen XG, Zheng L, Wang Z, et al. Molecular affinity and permeability of different molecular weight chitosan membranes. J Agric Food Chem 2002;50(21):5915-18
  • Naberezhnykh GA, Gorbach VI, Likhatskaya GN, et al. Interaction of N-acylated and N-alkylated chitosans included in liposomes with lipopolysaccharide of gram-negative bacteria. Biochemistry (Mosc) 2013;78(3):301-8
  • Naberezhnykh GA, Gorbach VI, Likhatskaya GN, et al. Interaction of chitosans and their N-acylated derivatives with lipopolysaccharide of gram-negative bacteria. Biochemistry (Mosc) 2008;73(4):432-41
  • Li J, Shao Y, Chen Z, et al. Membrane cartridges for endotoxin removal from interferon preparations. J Chromatogr B Analyt Technol Biomed Life Sci 2003;791(1-2):55-61
  • Qiao Y, Bai XF, Du YG. Chitosan oligosaccharides protect mice from LPS challenge by attenuation of inflammation and oxidative stress. Int Immunopharmacol 2011;11(1):121-7
  • Hagan KA, Reedy CR, Uchimoto ML, et al. An integrated, valveless system for microfluidic purification and reverse transcription-PCR amplification of RNA for detection of infectious agents. Lab Chip 2011;11(5):957-61
  • Qin C, Du Y, Xiao L, et al. Enzymic preparation of water-soluble chitosan and their antitumor activity. Int J Biol Macromol 2002;31(1-3):111-17
  • Mellegård H, Strand SP, Christensen BE, et al. Antibacterial activity of chemically defined chitosans: influence of molecular weight, degree of acetylation and test organism. Int J Food Microbiol 2011;148(1):48-54
  • Helander IM, Nurmiaho-Lassila EL, Ahvenainen R, et al. Chitosan disrupts the barrier properties of the outer membrane of gram-negative bacteria. Int J Food Microbiol 2001;71(2-3):235-44
  • Ishihara C, Yoshimatsu K, Tsuji M, et al. Anti-viral activity of sulfated chitin derivatives against Friend murine leukaemia and herpes simplex type-1 viruses. Vaccine 1993;11(6):670-4
  • Mori Y, Ono T, Miyahira Y, et al. Antiviral activity of silver nanoparticle/chitosan composites against H1N1 influenza A virus. Nanoscale Res Lett 2013;8(1):93
  • Han XY, Ma YF, Lv MY, et al. Chitosan-zinc chelate improves intestinal structure and mucosal function and decreases apoptosis in ileal mucosal epithelial cells in weaned pigs. Br J Nutr 2014;111(8):1405-11
  • Jiang X, van der Horst A, van Steenbergen MJ, et al. Molar-mass characterization of cationic polymers for gene delivery by aqueous size-exclusion chromatography. Pharm Res 2006;23(3):595-603
  • Tan SC, Khor E, Tan TK, Wong SM. The degree of deacetylation of chitosan: advocating the first derivative UV-spectrophotometry method of determination. Talanta 1998;45(4):713-19
  • Sieval AB, Thanou M, Kotzé AF, et al. Preparation and NMR characterization of highly substituted N-trimethyl chitosan chloride. Carbohydr Polym 1998;36(2-3):157-65
  • Dornish M, Kaplan D, Skaugrud O. Standards and guidelines for biopolymers in tissue-engineered medical products: ASTM alginate and chitosan standard guides. American Society for Testing and Materials. Ann N Y Acad Sci 2001;944:388-97
  • Pok S, Myers JD, Madihally SV, Jacot JG. A multilayered scaffold of a chitosan and gelatin hydrogel supported by a PCL core for cardiac tissue engineering. Acta Biomater 2012;9(3):5630-42
  • Suzuki M, Mikami T, Matsumoto T, Suzuki S. Gelation of limulus lysate by synthetic dextran derivatives. Microbiol Immunol 1977;21(8):419-25

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.