DOI QR코드

DOI QR Code

Oxidized Carbon Nanosphere-Based Subunit Vaccine Delivery System Elicited Robust Th1 and Cytotoxic T Cell Responses

  • Sawutdeechaikul, Pritsana (Graduate Program in Microbiology and Microbial Technology, Department of Microbiology, Faculty of Science, Chulalongkorn University) ;
  • Cia, Felipe (Department of Immunology and Infection, London School of Hygiene and Tropical Medicine) ;
  • Bancroft, Gregory J. (Department of Immunology and Infection, London School of Hygiene and Tropical Medicine) ;
  • Wanichwecharungruang, Supason (Center of Excellence in Materials and Bio-Interfaces, Chulalongkorn University and Department of Chemistry, Faculty of Science, Chulalongkorn University) ;
  • Sittplangkoo, Chutamath (Center of Excellence in Immune-mediated Diseases, Chulalongkorn University) ;
  • Palaga, Tanapat (Graduate Program in Microbiology and Microbial Technology, Department of Microbiology, Faculty of Science, Chulalongkorn University)
  • Received : 2018.09.25
  • Accepted : 2019.01.17
  • Published : 2019.03.28

Abstract

Subunit vaccines are safer and more stable than live vaccines although they have the disadvantage of eliciting poor immune response. To develop a subunit vaccine, an effective delivery system targeting the key elements of the protective immune response is a prerequisite. In this study, oxidized carbon nanospheres (OCNs) were used as a subunit vaccine delivery system and tuberculosis (TB) was chosen as a model disease. TB is among the deadliest infectious diseases worldwide and an effective vaccine is urgently needed. The ability of OCNs to deliver recombinant Mycobacterium tuberculosis (Mtb) proteins, Ag85B and HspX, into bone marrow derived macrophages (BMDMs) and dendritic cells (BMDCs) was investigated. For immunization, OCNs were mixed with the two TB antigens as well as the adjuvant monophosphoryl lipid A (MPL). The protective efficacy was analyzed in vaccinated mice by aerosol Mtb challenge with a virulent strain of Mtb and the bacterial burdens were measured. The results showed that OCNs are highly effective in delivering Mtb proteins into the cytosol of BMDMs and BMDCs. Upon immunization, this vaccine formula induced robust Th1 immune response characterized by cytokine profiles from restimulated splenocytes and specific antibody titer. More importantly, enhanced cytotoxic $CD8^+$ T cell activation was observed. However, it did not reduce the bacteria burden in the lung and spleen from the aerosol Mtb challenge. Taken together, OCNs are highly effective in delivering subunit protein vaccine and induce robust Th1 and $CD8^+$ T cell response. This vaccine delivery system is suitable for application in settings where cell-mediated immune response is needed.

Keywords

References

  1. Moreno-Mendieta SA, Rocha-Zavaleta L, Rodriguez-Sanoja R. 2010. Adjuvants in tuberculosis vaccine development. FEMS Immmunol. Med. Microbiol. 58: 75-84. https://doi.org/10.1111/j.1574-695X.2009.00629.x
  2. Andersen P, Doherty M. 2005. The success and failure of BCG-implications for a novel tuberculosis vaccine. Nat. Rev. Microbiol. 3: 656-662. https://doi.org/10.1038/nrmicro1211
  3. Nunes-Alves C, Booty MG, Carpenter SM, Jayaraman P, Rothchild AC, Behar SM. 2014. In search of a new paradigm for protective immunity to TB. Nat. Rev. Microbiol. 12: 289-299. https://doi.org/10.1038/nrmicro3230
  4. Ottenhoff TH, Kaufmann SH. 2012. Vaccines against tuberculosis: where are we and where do we need to go? PLoS Path. 8: e1002607. https://doi.org/10.1371/journal.ppat.1002607
  5. Fehres CM, Unger WWJ, Garcia-Vallejo JJ, van Kooyk Y. 2014. Understanding the biology of antigen cross-presentation for the design of vaccines against cancer. Front. Immunol. 5: 149. https://doi.org/10.3389/fimmu.2014.00149
  6. Kasturi SP, Pulendran B. 2008. Cross-presentation: avoiding trafficking chaos? Nat. Immunol. 9: 461-463. https://doi.org/10.1038/ni0508-461
  7. Joshi VB, Geary SM, Salem AK. 2013. Biodegradable particles as vaccine delivery systems: size matters. AAPS J. 15: 85-94. https://doi.org/10.1208/s12248-012-9418-6
  8. Smith DM, Simon JK, Baker JR, Jr. 2013. Applications of nanotechnology for immunology. Nat. Rev. Immunol. 13: 592-605. https://doi.org/10.1038/nri3488
  9. Gregory AE, Titball R, Williamson D. 2013. Vaccine delivery using nanoparticles. Front. Cell. Infect. Microbiol. 3: 13.
  10. Couvreur PV, C. 2006. Nanotechnology: Intelligent design to treat complex disease. Pharm. Res. 23: 1417-1450. https://doi.org/10.1007/s11095-006-0284-8
  11. Demento SL, Cui W, Criscione JM, Stern E, Tulipan J, Kaech SM, et al. 2012. Role of sustained antigen release from nanoparticle vaccines in shaping the T cell memory phenotype. Biomaterials 33: 4957-4964. https://doi.org/10.1016/j.biomaterials.2012.03.041
  12. van Dissel JT, Arend SM, Prins C, Bang P, Tingskov PN, Lingnau K, et al. 2010. Ag85B-ESAT-6 adjuvanted with IC31 promotes strong and long-lived Mycobacterium tuberculosis specific T cell responses in naive human volunteers. Vaccine 28: 3571-3581. https://doi.org/10.1016/j.vaccine.2010.02.094
  13. Leleux J, Roy K. 2013. Micro and nanoparticle-based delivery systems for vaccine immunotherapy: an immunological and materials perspective. Adv. Healthc Mater. 2: 72-94. https://doi.org/10.1002/adhm.201200268
  14. Shen H, Ackerman AL, Cody V, Giodini A, Hinson ER, Cresswell P, et al. 2006. Enhanced and prolonged crosspresentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles. Immunology 117: 78-88. https://doi.org/10.1111/j.1365-2567.2005.02268.x
  15. Hirosue S, Kourtis IC, van der Vlies AJ, Hubbell JA, Swartz MA. 2010. Antigen delivery to dendritic cells by poly(propylene sulfide) nanoparticles with disulfide conjugated peptides: Cross-presentation and T cell activation. Vaccine 28: 7897-7906. https://doi.org/10.1016/j.vaccine.2010.09.077
  16. Khademi F, Derakhshan M, Yousefi-Avarvand A, Tafaghodi M. 2018. Potential of polymeric particles as future vaccine delivery systems/adjuvants for parenteral and non-parenteral immunization against tuberculosis: A systematic review. Iran J. Basic Med. Sci. 21: 116-123.
  17. Arayachukeat S, Palaga T, Wanichwecharungruang SP. 2012. Clusters of carbon nanospheres derived from graphene oxide. ACS Appl. Mat. Inter. 4: 6808-6815. https://doi.org/10.1021/am3019959
  18. Arayachukiat S, Seemork J, Pan-In P, Amornwachirabodee K, Sangphech N, Sansureerungsikul T, et al. 2015. Bringing macromolecules into cells and evading endosomes by oxidized carbon nanoparticles. Nano Lett. 15: 3370-3376. https://doi.org/10.1021/acs.nanolett.5b00696
  19. Mata-Haro V, Cekic C, Martin M, Chilton PM, Casella CR, Mitchell TC. 2007. The vaccine adjuvant monophosphoryl lipid a as a TRIF-biased agonist of TLR4. Science 316: 1628-1632. https://doi.org/10.1126/science.1138963
  20. Yuan Y, Crane DD, Simpson RM, Zhu Y, Hickey MJ, Sherman DR, et al. 1998. The 16-kDa ${\alpha}$-crystallin (Acr) protein of Mycobacterium tuberculosis is required for growth in macrophages. Proc. Natl. Acad. Sci. USA 95: 9578-9583. https://doi.org/10.1073/pnas.95.16.9578
  21. Belisle JT, Vissa VD, Sievert T, Takayama K, Brennan PJ, Besra GS. 1997. Role of the major antigen of Mycobacterium tuberculosis in cell wall biogenesis. Science 276: 1420-1422. https://doi.org/10.1126/science.276.5317.1420
  22. Boonyatecha N, Sangphech N, Wongchana W, Kueanjinda P, Palaga T. 2012. Involvement of notch signaling pathway in regulating IL-12 expression via c-Rel in activated macrophages. Mol. Immunol. 51: 255-262. https://doi.org/10.1016/j.molimm.2012.03.017
  23. Nguyen TNY, Padungros P, Wongsrisupphakul P, Sa-Ard-Iam N, Mahanonda R, Matangkasombut O, et al. 2018. Cell wall mannan of Candida krusei mediates dendritic cell apoptosis and orchestrates Th17 polarization via TLR-2/MyD88-dependent pathway. Sci. Rep. 8: 17123. https://doi.org/10.1038/s41598-018-35101-3
  24. Fletcher HA, Schrager L. 2016. TB vaccine development and the End TB Strategy: importance and current status. Trans. R. Soc. Trop. Med. Hyg. 110: 212-218. https://doi.org/10.1093/trstmh/trw016
  25. Grode L, Seiler P, Baumann S, Hess J, Brinkmann V, Eddine AN, et al. 2005. Increased vaccine efficacy against tuberculosis of recombinant Mycobacterium bovis bacille Calmette-Guerin mutants that secrete listeriolysin. J. Clin. Invest. 115: 2472-2479. https://doi.org/10.1172/JCI24617
  26. Hoft DF, Blazevic A, Abate G, Hanekom WA, Kaplan G, Soler JH, et al. 2008. A new recombinant BCG vaccine safely induces significantly enhanced TB-specific immunity in human volunteers. J. Infect. Dis. 198: 1491-1501. https://doi.org/10.1086/592450
  27. Nieuwenhuizen NE, Kaufmann SHE. 2018. Next-generation vaccines based on Bacille Calmette-Guerin. Front Immunol. 9: 121. https://doi.org/10.3389/fimmu.2018.00121
  28. Winau F, Weber S, Sad S, de Diego J, Hoops SL, Breiden B, et al. 2006. Apoptotic vesicles crossprime CD8 T cells and protect against tuberculosis. Immunity 24: 105-117. https://doi.org/10.1016/j.immuni.2005.12.001
  29. Desel C, Dorhoi A, Bandermann S, Grode L, Eisele B, Kaufmann SH. 2011. Recombinant BCG Delta ureC hly+ induces superior protection over parental BCG by stimulating a balanced combination of type 1 and type 17 cytokine responses. J. Infect. Dis. 204: 1573-1584. https://doi.org/10.1093/infdis/jir592
  30. Casella CR, Mitchell TC. 2008. Putting endotoxin to work for us: monophosphoryl lipid A as a safe and effective vaccine adjuvant. Cell Mol. Life Sci. 65: 3231-3240. https://doi.org/10.1007/s00018-008-8228-6
  31. Awasthi S. 2014. Toll-Like receptor-4 modulation for cancer immunotherapy. Front Immunol. 5: 328. https://doi.org/10.3389/fimmu.2014.00328
  32. Neeland MR, Shi W, Collignon C, Taubenheim N, Meeusen ENT, Didierlaurent AM, et al. 2016. The Lymphatic immune response induced by the adjuvant AS01: a comparison of intramuscular and subcutaneous immunization routes. J. Immunol. 197: 2704. https://doi.org/10.4049/jimmunol.1600817
  33. Leleux J, Roy K. 2013. Micro and nanoparticle-based delivery systems for vaccine immunotherapy: an immunological and materials perspective. Adv. Healthc. Mater. 2: 72-94. https://doi.org/10.1002/adhm.201200268
  34. Carletti D, Morais da Fonseca D, Gembre AF, Masson AP, Weijenborg Campos L, Leite LCC, et al. 2013. A single dose of a DNA vaccine encoding apa coencapsulated with 6,6'-trehalose dimycolate in microspheres confers long-term protection against tuberculosis in Mycobacterium bovis BCG-primed mice. Clin. Vaccine immunol. 20: 1162-1169. https://doi.org/10.1128/CVI.00148-13
  35. Ha S-J, Park S-H, Kim H-J, Kim S-C, Kang H-J, Lee E-G, et al. 2006. Enhanced immunogenicity and protective efficacy with the use of interleukin-12-encapsulated microspheres plus AS01B in tuberculosis subunit vaccination. Infect. Immun. 74: 4954-4959. https://doi.org/10.1128/IAI.01781-05
  36. Crotty S. 2014. T follicular helper cell differentiation, function, and roles in disease. Immunity 41: 529-542. https://doi.org/10.1016/j.immuni.2014.10.004
  37. Olsen AW, Williams A, Okkels LM, Hatch G, Andersen P. 2004. Protective effect of a tuberculosis subunit vaccine based on a fusion of Antigen 85B and ESAT-6 in the aerosol guinea pig model. Infect. Immun. 72: 6148-6150. https://doi.org/10.1128/IAI.72.10.6148-6150.2004
  38. Doherty TM, Olsen AW, Weischenfeldt J, Huygen K, D'Souza S, Kondratieva TK, et al. 2004. Comparative analysis of different vaccine constructs expressing defined antigens from Mycobacterium tuberculosis. J. Infect. Dis. 190: 2146-2153. https://doi.org/10.1086/425931
  39. Niu H, Peng J, Bai C, Liu X, Hu L, Luo Y, et al. 2015. Multistage tuberculosis subunit vaccine candidate LT69 provides high protection against Mycobacterium tuberculosis infection in mice. PLoS One 10: e0130641. https://doi.org/10.1371/journal.pone.0130641
  40. Aagaard C, Hoang T, Dietrich J, Cardona P-J, Izzo A, Dolganov G, et al. 2011. A multistage tuberculosis vaccine that confers efficient protection before and after exposure. Nat. Med. 17: 189-194. https://doi.org/10.1038/nm.2285