Original Research Nano-adjuvanted polio vaccine: Preparation and characterization of chitosan and trimethylchitosan (TMC) nanoparticles loaded with inactivated polio virus and coated with sodium alginate

Document Type : Research Paper

Authors

1 Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

2 Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands

3 Institute for Translational Vaccinology, Bilthoven, Netherlands

Abstract

Objective(s): 
It is proposed that particulate antigens could better interact with the antigen presenting cells (APCs). A fast, simple and scalable process for preparation of polymeric nanoparticles (NPs) is coating of charged antigenic particles, like viruses, with oppositely charged polymers. A second coating with a charged polymer could increase the stability and modify the immunomodulatory potentials of NPs. 
Materials and Methods: 
Negatively charged inactivated polio virus (IPV) was coated with cationic polymers, chitosan (CHT) and trimethylchitosan (TMC) by a simple incubation method. CHT: IPV and TMC: IPV NPs were coated by anionic polymer, sodium alginate (ALG). Physical characteristics and stability of NPs were studied. Cytocompatibility of NPs was checked with MTT assay. DC maturation study was used for evaluation of the NPs potential in interaction with DCs. 
Results: 
Among the various polymer to antigen ratios tested, the least size and PDI and the highest ZP was seen in TMC: IPV (2:1), CHT: IPV (2:1), ALG: TMC: IPV (2:2:1) and ALG: CHT: IPV (4:2:1). The physical stability of TMC: IPV and CHT: IPV was preserved until 15 days. After an early de-association of some part of coated alginate, ALG: CHT: IPV and ALG: TMC: IPC NPs were stable until the end of study (25th day). No one of the NPs formulations had a negative effect on cell viability. Compared with plain IPV, nanoparticulate IPV formulations failed to increase the expression of CD40 and CD86 markers of DCs. 
Conclusion:  
NPs prepared with simple and scalable method, had reasonable physical characteristics, stability and cytocompatibility and could be tested in vivo for their immunoadjuvant potential.

Keywords


1. Amorij JP, Hinrichs W, Frijlink HW, Wilschut JC, Huckriede A. Needle-free influenza vaccination. Lancet Infect Dis. 2010; 10: 699-711.
2. Saluja V, Amorij JP, van Roosmalen ML, et al. Intranasal delivery of influenza subunit vaccine formulated with GEM particles as an adjuvant. AAPS J. 2010; 12: 109-16.
3. Tafaghodi M, Rastegar S. Preparation and in vivo study of dry powder microspheres for nasal immunization. J Drug Target. 2010; 18: 235-42.
4. Amin M, Jaafari MR, Tafaghodi M. Impact of chitosan coating of anionic liposomes on clearance rate, mucosal and systemic immune responses following nasal administration in rabbits. Colloids Surf B Biointerfaces. 2009; 74: 225-9.
5. Slutter B, Hagenaars N, Jiskoot W. Rational design of nasal vaccines. J Drug Target. 2008; 16: 1-17.
6. Hirschberg HJHB, de Wijdeven GGPv, Kraan H, Amorij J-P, Kersten GFA. Bioneedles as alternative delivery system for hepatitis B vaccine. J Control Release. 2010; 147: 211-7.
7. Hagenaars N, Mastrobattista E, Verheul RJ, et al. Physicochemical and immunological characterization of N,N,N-trimethyl chitosan-coated whole inactivated influenza virus vaccine for intranasal administration. Pharm Res. 2009; 26: 1353-64.
8. Tafaghodi M, Saluja V, Kersten GF, et al. Hepatitis B surface antigen nanoparticles coated with chitosan and trimethyl chitosan: Impact of formulation on physicochemical and immunological characteristics. Vaccine. 2012; 30: 5341-8.
9. Koping-Hoggard M, Sanchez A, Alonso MJ. Nanoparticles as carriers for nasal vaccine delivery. Expert Rev Vaccines. 2005; 4: 185-96.
10. Almeida AJ, Alpar HO. Mucosal immunization with antigen-containing microparticles. In: Gander B, Merkle HP, (eds) Antigen delivery systems. New York: Harwood academic publishers, 1997. 207-26.
11. Verheul RJ, Amidi M, van der Wal S, van Riet E, Jiskoot W, Hennink WE. Synthesis, characterization and in vitro biological properties of O-methyl free N,N,N-trimethylated chitosan. Biomaterials. 2008; 29: 3642-9.
12. Mangal S, Pawar D, Garg NK, et al. Pharmaceutical and immunological evaluation of mucoadhesive nanoparticles based delivery system(s) administered intranasally. Vaccine. 2011; 29: 4953-62.
13. Bal SM, Ding Z, van Riet E, Jiskoot W, Bouwstra JA. Advances in transcutaneous vaccine delivery: do all ways lead to Rome? J Control Release. 2010; 148: 266-82.
14. Illum L, Jabbal-Gill I, Hinchcliffe M, Fisher AN, Davis SS. Chitosan as a novel nasal delivery system for vaccines. Adv Drug Deliv Rev. 2001; 51: 81-96.
15. van der Lubben IM, Kersten G, Fretz MM, Beuvery C, Coos Verhoef J, Junginger HE. Chitosan microparticles for mucosal vaccination against diphtheria: oral and nasal efficacy studies in mice. Vaccine. 2003; 21: 1400-8.
16. 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.
17. Boonyo W, Junginger HE, Waranuch N, Polnok A, Pitaksuteepong T. Chitosan and trimethyl chitosan chloride (TMC) as adjuvants for inducing immune responses to ovalbumin in mice following nasal administration. J Control Release. 2007; 121: 168-75.
18. Sadeghi AM, Dorkoosh FA, Avadi MR, Saadat P, Rafiee-Tehrani M, Junginger HE. Preparation, characterization and antibacterial activities of chitosan, N-trimethyl chitosan (TMC) and N-diethylmethyl chitosan (DEMC) nanoparticles loaded with insulin using both the ionotropic gelation and polyelectrolyte complexation methods. Int J Pharm. 2008; 355: 299-306.
19. Amidi M, Romeijn SG, Borchard G, Junginger HE, Hennink WE, Jiskoot W. Preparation and characterization of protein-loaded N-trimethyl chitosan nanoparticles as nasal delivery system. J Control Release. 2006; 111: 107-16.
20. Amidi M, Romeijn SG, Verhoef JC, et al. N-Trimethyl chitosan (TMC) nanoparticles loaded with influenza subunit antigen for intranasal vaccination: Biological properties and immunogenicity in a mouse model. Vaccine. 2007; 25: 144-53.
21. Hagenaars N, Verheul RJ, Mooren I, et al. Relationship between structure and adjuvanticity of N,N,N-trimethyl chitosan (TMC) structural variants in a nasal influenza vaccine. J Control Release. 2009; 140: 126-33.
22. Slutter B, Bal S, Keijzer C, et al. Nasal vaccination with N-trimethyl chitosan and PLGA based nanoparticles: nanoparticle characteristics determine quality and strength of the antibody response in mice against the encapsulated antigen. Vaccine. 2010; 28: 6282-91.
23. Slutter B, Plapied L, Fievez V, et al. Mechanistic study of the adjuvant effect of biodegradable nanoparticles in mucosal vaccination. J Control Release. 2009; 138: 113-21.
24. Demoulins 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.
25. Oliveira CR, Rezende CM, Silva MR, Pego AP, Borges O, Goes AM. A new strategy based on SmRho protein loaded chitosan nanoparticles as a candidate oral vaccine against schistosomiasis. PLoS Negl Trop Dis. 2012; 6: e1894.
26. Bal SM, Slutter B, van Riet E, et al. Efficient induction of immune responses through intradermal vaccination with N-trimethyl chitosan containing antigen formulations. J Control Release. 2009; 142: 374-83.
27. Hagenaars N, Mania M, de Jong P, et al. Role of trimethylated chitosan (TMC) in nasal residence time, local distribution and toxicity of an intranasal influenza vaccine. J Control Release. 2010; 144: 17-24.
28. (28) Muzzarelli RA. Colorimetric determination of chitosan. Anal Biochem. 1998; 260: 255-7.
29. (29) Wischke C, Borchert HH. Increased sensitivity of chitosan determination by a dye binding method. Carbohydr Res. 2006; 341: 2978-9.
30. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983; 65: 55-63.
31. Jafari T, Simchi A, Khakpash N. Synthesis and cytotoxicity assessment of superparamagnetic iron-gold core-shell nanoparticles coated with polyglycerol. J Colloid Interface Sci. 2010; 345: 64-71.
32. De Jong EC, Vieira PL, Kalinski P, et al. Microbial compounds selectively induce Th1 cell-promoting or Th2 cell-promoting dendritic cells in vitro with diverse th cell-polarizing signals. J Immunol. 2002; 168: 1704-9.
33. Bal SM, Slutter 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: 374-83.
34. Borges O, Borchard G, Verhoef JC, de Sousa A, Junginger HE. Preparation of coated nanoparticles for a new mucosal vaccine delivery system. Int J Pharm. 2005; 299: 155-66.
35. Borges O, Cordeiro-da-Silva A, Tavares J, et al. Immune response by nasal delivery of hepatitis B surface antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles. Eur J Pharm Biopharm. 2008; 69: 405-16.
36. Borges O, Silva M, de Sousa A, Borchard G, Junginger HE, Cordeiro-da-Silva A. Alginate coated chitosan nanoparticles are an effective subcutaneous adjuvant for hepatitis B surface antigen. Int Immunopharmacol. 2008; 8: 1773-80.
37. Chen F, Zhang ZR, Huang Y. Evaluation and modification of N-trimethyl chitosan chloride nanoparticles as protein carriers. Int J Pharm. 2007; 336: 166-73.
38. Zhou J, Romero G, Rojas E, Ma L, Moya S, Gao C. Layer by layer chitosan/alginate coatings on poly(lactide-co-glycolide) nanoparticles for antifouling protection and Folic acid binding to achieve selective cell targeting. J Colloid Interface Sci. 2010; 345: 241-7.