Generic placeholder image

Current Pharmaceutical Biotechnology

Editor-in-Chief

ISSN (Print): 1389-2010
ISSN (Online): 1873-4316

Review Article

A Glance on Nanovaccine: A Potential Approach for Disease Prevention

Author(s): Akash Garg, Rutvi Agrawal*, Himansu Chopra, Talever Singh, Ramkumar Chaudhary and Abhishek Tankara

Volume 25, Issue 11, 2024

Published on: 11 October, 2023

Page: [1406 - 1418] Pages: 13

DOI: 10.2174/0113892010254221231006100659

Price: $65

conference banner
Abstract

There are several vaccines available for preventing various bacterial and viral infections, but still, there are many challenges that require the development of noninvasive, more efficient, and active vaccines. The advancement in biotechnological tools has provided safer antigens, such as nucleic acids, proteins etc., but due to their lower immunogenic property, adjuvants of stronger immune response are required. Nanovaccines are effective vaccines when compared with conventional vaccines as they can induce both Humoral and cell-mediated immune responses and also provide longer immunogenic memory. The nanocarriers used in vaccines act as adjuvant. They provide site-specific delivery of antigens and can be used in conjugation with immunostimulatory molecules for enhancing adjuvant therapy. The nanovaccines avoid degrading cell pathways and provide effective absorption into blood vessels. The higher potential of nanovaccines to treat various diseases, such as acquired immuno deficiency syndrome, cancer, tuberculosis, malaria and many others, along with their immunological mechanisms and different types, have been discussed in the review.

Keywords: Vaccine, nanovaccine, immune response, infectious diseases, humoral, cell-mediated.

Graphical Abstract
[1]
Sekhon, B.; Sekhon, B.; Saluja, V. Nanovaccines-An overview. Inter. J. Pharma. Frontier Res., 2011, 1, 101-109.
[2]
Griffin, J. A strategic approach to vaccine development: Animal models, monitoring vaccine efficacy, formulation and delivery. Adv. Drug Deliv. Rev., 2002, 54(6), 851-861.
[http://dx.doi.org/10.1016/S0169-409X(02)00072-8] [PMID: 12363434]
[3]
Riedel, S. Edward Jenner and the history of smallpox and vaccination. Proc. Bayl. Univ. Med. Cent., 2005, 18(1), 21-25.
[http://dx.doi.org/10.1080/08998280.2005.11928028] [PMID: 16200144]
[4]
Kaufmann, S.H.E.; Juliana McElrath, M.; Lewis, D.J.M.; Del Giudice, G. Challenges and responses in human vaccine development. Curr. Opin. Immunol., 2014, 28, 18-26.
[http://dx.doi.org/10.1016/j.coi.2014.01.009] [PMID: 24561742]
[5]
Barouch, D.H. Challenges in the development of an HIV-1 vaccine. Nature, 2008, 455(7213), 613-619.
[http://dx.doi.org/10.1038/nature07352] [PMID: 18833271]
[6]
Feinberg, M.B.; Moore, J.P. AIDS vaccine models: Challenging challenge viruses. Nat. Med., 2002, 8(3), 207-210.
[http://dx.doi.org/10.1038/nm0302-207] [PMID: 11875482]
[7]
Nabel, G.J. Challenges and opportunities for development of an AIDS vaccine. Nature, 2001, 410(6831), 1002-1007.
[http://dx.doi.org/10.1038/35073500] [PMID: 11309631]
[8]
Brennan, M.J. The tuberculosis vaccine challenge. Tuberculosis, 2005, 85(1-2), 7-12.
[http://dx.doi.org/10.1016/j.tube.2004.09.001] [PMID: 15687021]
[9]
Raviglione, M.; Sulis, G. Tuberculosis 2015: Burden, challenges and strategy for control and elimination. Infect. Dis. Rep., 2016, 8(2), 6570.
[http://dx.doi.org/10.4081/idr.2016.6570] [PMID: 27403269]
[10]
Siegrist, C.A. The challenges of vaccine responses in early life: Selected examples. J. Comp. Pathol., 2007, 137(1), S4-S9.
[http://dx.doi.org/10.1016/j.jcpa.2007.04.004] [PMID: 17559867]
[11]
Wilson-Welder, J.H.; Torres, M.P.; Kipper, M.J.; Mallapragada, S.K.; Wannemuehler, M.J.; Narasimhan, B. Vaccine adjuvants: Current challenges and future approaches. J. Pharm. Sci., 2009, 98(4), 1278-1316.
[http://dx.doi.org/10.1002/jps.21523] [PMID: 18704954]
[12]
Gheibi Hayat, S.M.; Darroudi, M. Nanovaccine: A novel approach in immunization. J. Cell. Physiol., 2019, 234(8), 12530-12536.
[http://dx.doi.org/10.1002/jcp.28120] [PMID: 30633361]
[13]
Singh, A.; Misra, R.; Mohanty, C.; Sahoo, S.K. Applications of nanotechnology in vaccine delivery. Int. J. Green Nanotechnol. Biomed., 2010, 2(1), B25-B45.
[14]
Cordeiro, A.S.; Alonso, M.J.; de la Fuente, M. Nanoengineering of vaccines using natural polysaccharides. Biotechnol. Adv., 2015, 33(6), 1279-1293.
[http://dx.doi.org/10.1016/j.biotechadv.2015.05.010] [PMID: 26049133]
[15]
Bielinska, A.U.; Chepurnov, A.A.; Landers, J.J.; Janczak, K.W.; Chepurnova, T.S.; Luker, G.D.; Baker, J.R. Jr A novel, killed-virus nasal vaccinia virus vaccine. Clin. Vaccine Immunol., 2008, 15(2), 348-358.
[http://dx.doi.org/10.1128/CVI.00440-07] [PMID: 18057181]
[16]
Skwarczynski, M.; Toth, I. Peptide-based subunit nanovaccines. Curr. Drug Deliv., 2011, 8(3), 282-289.
[http://dx.doi.org/10.2174/156720111795256192] [PMID: 21291373]
[17]
Akagi, T.; Akashi, M. Development of polymeric nanoparticles-based vaccine. Jpn. J. Clin. Med., 2006, 64(2), 279-285.
[18]
Danhier, F.; Feron, O.; Préat, V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J. Control. Release, 2010, 148(2), 135-146.
[http://dx.doi.org/10.1016/j.jconrel.2010.08.027] [PMID: 20797419]
[19]
Jain, K.K. Nanoparticles as targeting ligands. Trends Biotechnol., 2006, 24(4), 143-145.
[http://dx.doi.org/10.1016/j.tibtech.2006.02.004] [PMID: 16488033]
[20]
Singh, R.; Lillard, J.W. Jr Nanoparticle-based targeted drug delivery. Exp. Mol. Pathol., 2009, 86(3), 215-223.
[http://dx.doi.org/10.1016/j.yexmp.2008.12.004] [PMID: 19186176]
[21]
Wang, M.; Thanou, M. Targeting nanoparticles to cancer. Pharmacol. Res., 2010, 62(2), 90-99.
[http://dx.doi.org/10.1016/j.phrs.2010.03.005] [PMID: 20380880]
[22]
Mahmoudi, M.; Sant, S.; Wang, B.; Laurent, S.; Sen, T. Superparamagnetic iron oxide nanoparticles (SPIONs): Development, surface modification and applications in chemotherapy. Adv. Drug Deliv. Rev., 2011, 63(1-2), 24-46.
[http://dx.doi.org/10.1016/j.addr.2010.05.006] [PMID: 20685224]
[23]
Mou, X.; Ali, Z.; Li, S.; He, N. Applications of magnetic nanoparticles in targeted drug delivery system. J. Nanosci. Nanotechnol., 2015, 15(1), 54-62.
[http://dx.doi.org/10.1166/jnn.2015.9585] [PMID: 26328305]
[24]
Mohammed, M.R.; Sher Bahadar, K.; Aslam, J.; Mohd, F.; Abdullah, M.A. Iron Oxide Nanoparticles. In: Nanomaterials; IntechOpen: Rijeka, 2011.
[25]
Bao, G.; Mitragotri, S.; Tong, S. Multifunctional nanoparticles for drug delivery and molecular imaging. Annu. Rev. Biomed. Eng., 2013, 15(1), 253-282.
[http://dx.doi.org/10.1146/annurev-bioeng-071812-152409] [PMID: 23642243]
[26]
Shahbazi, M-A.; Santos, H.A. Revolutionary impact of nanovaccines on immunotherapy. New Horiz. Transl. Med., 2015, 2(2), 44-50.
[27]
Stammers, T.S.; Erden, Y.J.; Hunt, G. The promise and challenge of nanovaccines and the question of global equity; J Nanotechnology Perceptions, 2013, pp. 16-27.
[http://dx.doi.org/10.4024/N02ST13A.ntp.09.01]
[28]
Smith, D.M.; Simon, J.K.; Baker, J.R., Jr Applications of nanotechnology for immunology. Nat. Rev. Immunol., 2013, 13(8), 592-605.
[http://dx.doi.org/10.1038/nri3488] [PMID: 23883969]
[29]
Zaman, M.; Good, M.F.; Toth, I. Nanovaccines and their mode of action. Methods, 2013, 60(3), 226-231.
[http://dx.doi.org/10.1016/j.ymeth.2013.04.014] [PMID: 23623821]
[30]
Luo, M.; Samandi, L.Z.; Wang, Z.; Chen, Z.J.; Gao, J. Synthetic nanovaccines for immunotherapy. J. Control. Release, 2017, 263, 200-210.
[http://dx.doi.org/10.1016/j.jconrel.2017.03.033] [PMID: 28336379]
[31]
Paulis, L.E.; Mandal, S.; Kreutz, M.; Figdor, C.G. Dendritic cell-based nanovaccines for cancer immunotherapy. Curr. Opin. Immunol., 2013, 25(3), 389-395.
[http://dx.doi.org/10.1016/j.coi.2013.03.001] [PMID: 23571027]
[32]
Köping-Höggård, M.; Sánchez, A.; Alonso, M.J. Nanoparticles as carriers for nasal vaccine delivery. Expert Rev. Vaccines, 2005, 4(2), 185-196.
[http://dx.doi.org/10.1586/14760584.4.2.185] [PMID: 15889992]
[33]
Zaheer, T.; Pal, K.; Zaheer, I. Topical review on nano-vaccinology: Biochemical promises and key challenges. Process Biochem., 2021, 100, 237-244.
[http://dx.doi.org/10.1016/j.procbio.2020.09.028] [PMID: 33013180]
[34]
Bhardwaj, P.; Bhatia, E.; Sharma, S.; Ahamad, N.; Banerjee, R. Advancements in prophylactic and therapeutic nanovaccines. Acta Biomater., 2020, 108, 1-21.
[http://dx.doi.org/10.1016/j.actbio.2020.03.020] [PMID: 32268235]
[35]
Sulczewski, F.B.; Liszbinski, R.B.; Romão, P.R.T.; Rodrigues Junior, L.C. Nanoparticle vaccines against viral infections. Arch. Virol., 2018, 163(9), 2313-2325.
[http://dx.doi.org/10.1007/s00705-018-3856-0] [PMID: 29728911]
[36]
Thomas, C.; Rawat, A.; Hope-Weeks, L.; Ahsan, F. Aerosolized PLA and PLGA nanoparticles enhance humoral, mucosal and cytokine responses to hepatitis B vaccine. Mol. Pharm., 2011, 8(2), 405-415.
[http://dx.doi.org/10.1021/mp100255c] [PMID: 21189035]
[37]
Diwan, M.; Tafaghodi, M.; Samuel, J. Enhancement of immune responses by co-delivery of a CpG oligodeoxynucleotide and tetanus toxoid in biodegradable nanospheres. J. Control. Release, 2002, 85(1-3), 247-262.
[http://dx.doi.org/10.1016/S0168-3659(02)00275-4] [PMID: 12480329]
[38]
Borges, O.; Cordeiro-da-Silva, A.; Tavares, J.; Santarém, N.; de Sousa, A.; Borchard, G.; Junginger, H.E. 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(2), 405-416.
[http://dx.doi.org/10.1016/j.ejpb.2008.01.019] [PMID: 18364251]
[39]
Li, P.; Luo, Z.; Liu, P.; Gao, N.; Zhang, Y.; Pan, H.; Liu, L.; Wang, C.; Cai, L.; Ma, Y. Bioreducible alginate-poly(ethylenimine) nanogels as an antigen-delivery system robustly enhance vaccine-elicited humoral and cellular immune responses. J. Control. Release, 2013, 168(3), 271-279.
[http://dx.doi.org/10.1016/j.jconrel.2013.03.025] [PMID: 23562637]
[40]
Pippa, N.; Gazouli, M.; Pispas, S. Recent advances and future perspectives in polymer-based nanovaccines. Vaccines, 2021, 9(6), 558.
[http://dx.doi.org/10.3390/vaccines9060558] [PMID: 34073648]
[41]
López-Sagaseta, J.; Malito, E.; Rappuoli, R.; Bottomley, M.J. Self-assembling protein nanoparticles in the design of vaccines. Comput. Struct. Biotechnol. J., 2016, 14, 58-68.
[http://dx.doi.org/10.1016/j.csbj.2015.11.001] [PMID: 26862374]
[42]
Qi, M.; Zhang, X.E.; Sun, X.; Zhang, X.; Yao, Y.; Liu, S.; Chen, Z.; Li, W.; Zhang, Z.; Chen, J.; Cui, Z. Intranasal nanovaccine confers homo- and hetero-subtypic influenza protection. Small, 2018, 14(13), e1703207.
[43]
Asadi, K.; Gholami, A. Virosome-based nanovaccines; a promising bioinspiration and biomimetic approach for preventing viral diseases: A review. Int. J. Biol. Macromol., 2021, 182, 648-658.
[http://dx.doi.org/10.1016/j.ijbiomac.2021.04.005] [PMID: 33862071]
[44]
Chasteen, N.D.; Harrison, P.M. Mineralization in ferritin: An efficient means of iron storage. J. Struct. Biol., 1999, 126(3), 182-194.
[http://dx.doi.org/10.1006/jsbi.1999.4118] [PMID: 10441528]
[45]
Kanekiyo, M.; Wei, C.J.; Yassine, H.M.; McTamney, P.M.; Boyington, J.C.; Whittle, J.R.R.; Rao, S.S.; Kong, W.P.; Wang, L.; Nabel, G.J. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature, 2013, 499(7456), 102-106.
[http://dx.doi.org/10.1038/nature12202] [PMID: 23698367]
[46]
Wahome, N.; Pfeiffer, T.; Ambiel, I.; Yang, Y.; Keppler, O.T.; Bosch, V.; Burkhard, P. Conformation-specific display of 4E10 and 2F5 epitopes on self-assembling protein nanoparticles as a potential HIV vaccine. Chem. Biol. Drug Des., 2012, 80(3), 349-357.
[http://dx.doi.org/10.1111/j.1747-0285.2012.01423.x] [PMID: 22650354]
[47]
Vijayan, V.; Mohapatra, A.; Uthaman, S.; Park, I.K. Recent advances in nanovaccines using biomimetic immunomodulatory materials. Pharmaceutics, 2019, 11(10), 534.
[http://dx.doi.org/10.3390/pharmaceutics11100534] [PMID: 31615112]
[48]
Gomes, A.; Mohsen, M.; Bachmann, M. Harnessing nanoparticles for immunomodulation and vaccines. Vaccines, 2017, 5(1), 6.
[http://dx.doi.org/10.3390/vaccines5010006] [PMID: 28216554]
[49]
Garg, H.; Mehmetoglu-Gurbuz, T.; Joshi, A. Virus Like Particles (VLP) as multivalent vaccine candidate against Chikungunya, Japanese Encephalitis, Yellow Fever and Zika Virus. Sci. Rep., 2020, 10(1), 4017.
[http://dx.doi.org/10.1038/s41598-020-61103-1] [PMID: 32132648]
[50]
Urakami, A.; Sakurai, A.; Ishikawa, M.; Yap, M.L.; Flores-Garcia, Y.; Haseda, Y.; Aoshi, T.; Zavala, F.P.; Rossmann, M.G.; Kuno, S.; Ueno, R.; Akahata, W. Development of a novel virus-like particle vaccine platform that mimics the immature form of alphavirus. Clin. Vaccine Immunol., 2017, 24(7), e00090-e17.
[http://dx.doi.org/10.1128/CVI.00090-17] [PMID: 28515133]
[51]
Roldão, A.; Mellado, M.C.M.; Castilho, L.R.; Carrondo, M.J.T.; Alves, P.M. Virus-like particles in vaccine development. Expert Rev. Vaccines, 2010, 9(10), 1149-1176.
[http://dx.doi.org/10.1586/erv.10.115] [PMID: 20923267]
[52]
Zhao, L.; Zhu, Z.; Ma, L.; Li, Y. O/W nanoemulsion as an adjuvant for an inactivated H3N2 influenza vaccine: Based on particle properties and mode of carrying. Int. J. Nanomedicine, 2020, 15, 2071-2083.
[http://dx.doi.org/10.2147/IJN.S232677] [PMID: 32273703]
[53]
Duinkerken, S.; Horrevorts, S.K.; Kalay, H.; Ambrosini, M.; Rutte, L.; de Gruijl, T.D.; Garcia-Vallejo, J.J.; van Kooyk, Y. Glyco-Dendrimers as intradermal anti-tumor vaccine targeting multiple skin DC subsets. Theranostics, 2019, 9(20), 5797-5809.
[http://dx.doi.org/10.7150/thno.35059] [PMID: 31534520]
[54]
Olczak, P.; Roden, R.B.S. Progress in L2-Based prophylactic vaccine development for protection against diverse human papillomavirus genotypes and associated diseases. Vaccines, 2020, 8(4), 568.
[http://dx.doi.org/10.3390/vaccines8040568] [PMID: 33019516]
[55]
Tao, W.; Ziemer, K.S.; Gill, H.S. Gold nanoparticle–M2e conjugate coformulated with CpG induces protective immunity against influenza A virus. Nanomedicine, 2014, 9(2), 237-251.
[http://dx.doi.org/10.2217/nnm.13.58] [PMID: 23829488]
[56]
Mousavi, S.M.; Zarei, M.; Hashemi, S.A.; Ramakrishna, S.; Chiang, W.H.; Lai, C.W.; Gholami, A. Gold nanostars-diagnosis, bioimaging and biomedical applications. Drug Metab. Rev., 2020, 52(2), 299-318.
[http://dx.doi.org/10.1080/03602532.2020.1734021] [PMID: 32150480]
[57]
Gholami, A.; Mousavi, S.M.; Hashemi, S.A.; Ghasemi, Y.; Chiang, W.H.; Parvin, N. Current trends in chemical modifications of magnetic nanoparticles for targeted drug delivery in cancer chemotherapy. Drug Metab. Rev., 2020, 52(1), 205-224.
[PMID: 32083952]
[58]
Climent, N.; García, I.; Marradi, M.; Chiodo, F.; Miralles, L.; Maleno, M.J.; Gatell, J.M.; García, F.; Penadés, S.; Plana, M. Loading dendritic cells with gold nanoparticles (GNPs) bearing HIV-peptides and mannosides enhance HIV-specific T cell responses. Nanomedicine, 2018, 14(2), 339-351.
[http://dx.doi.org/10.1016/j.nano.2017.11.009] [PMID: 29157976]
[59]
Lindblad, E.B. Aluminium adjuvants—in retrospect and prospect. Vaccine, 2004, 22(27-28), 3658-3668.
[http://dx.doi.org/10.1016/j.vaccine.2004.03.032] [PMID: 15315845]
[60]
Frey, A.; Mantis, N.; Kozlowski, P.A.; Quayle, A.J.; Bajardi, A.; Perdomo, J.J.; Robey, F.A.; Neutra, M.R. Immunization of mice with peptomers covalently coupled to aluminum oxide nanoparticles. Vaccine, 1999, 17(23-24), 3007-3019.
[http://dx.doi.org/10.1016/S0264-410X(99)00163-2] [PMID: 10462236]
[61]
Frey, A.; Neutra, M.R.; Robey, F.A. Peptomer aluminum oxide nanoparticle conjugates as systemic and mucosal vaccine candidates: Synthesis and characterization of a conjugate derived from the C4 domain of HIV-1MN gp120. Bioconjug. Chem., 1997, 8(3), 424-433.
[http://dx.doi.org/10.1021/bc970036p] [PMID: 9177850]
[62]
Chiu, D.; Zhou, W.; Kitayaporn, S.; Schwartz, D.T.; Murali-Krishna, K.; Kavanagh, T.J.; Baneyx, F. Biomineralization and size control of stable calcium phosphate core-protein shell nanoparticles: potential for vaccine applications. Bioconjug. Chem., 2012, 23(3), 610-617.
[http://dx.doi.org/10.1021/bc200654v] [PMID: 22263898]
[63]
Powell, T.J.; Palath, N.; DeRome, M.E.; Tang, J.; Jacobs, A.; Boyd, J.G. Synthetic nanoparticle vaccines produced by layer-by-layer assembly of artificial biofilms induce potent protective T-cell and antibody responses in vivo. Vaccine, 2011, 29(3), 558-569.
[http://dx.doi.org/10.1016/j.vaccine.2010.10.001] [PMID: 20951665]
[64]
Skwarczynski, M.; Toth, I. Recent advances in peptide-based subunit nanovaccines. Nanomedicine, 2014, 9(17), 2657-2669.
[http://dx.doi.org/10.2217/nnm.14.187] [PMID: 25529569]
[65]
Wang, N.; Chen, M.; Wang, T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J. Control. Release, 2019, 303, 130-150.
[http://dx.doi.org/10.1016/j.jconrel.2019.04.025] [PMID: 31022431]
[66]
He, H.; Yuan, D.; Wu, Y.; Cao, Y. Pharmacokinetics and pharmacodynamics modeling and simulation systems to support the development and regulation of liposomal drugs. Pharmaceutics, 2019, 11(3), 110.
[http://dx.doi.org/10.3390/pharmaceutics11030110] [PMID: 30866479]
[67]
Sharma, S.; Mukkur, T.K.S.; Benson, H.A.E.; Chen, Y. Pharmaceutical aspects of intranasal delivery of vaccines using particulate systems. J. Pharm. Sci., 2009, 98(3), 812-843.
[http://dx.doi.org/10.1002/jps.21493] [PMID: 18661544]
[68]
Khatri, K.; Goyal, A.K.; Gupta, P.N.; Mishra, N.; Mehta, A.; Vyas, S.P. Surface modified liposomes for nasal delivery of DNA vaccine. Vaccine, 2008, 26(18), 2225-2233.
[http://dx.doi.org/10.1016/j.vaccine.2008.02.058] [PMID: 18396362]
[69]
Li, S.; Rizzo, M.A.; Bhattacharya, S.; Huang, L. Characterization of cationic lipid-protamine–DNA (LPD) complexes for intravenous gene delivery. Gene Ther., 1998, 5(7), 930-937.
[http://dx.doi.org/10.1038/sj.gt.3300683] [PMID: 9813664]
[70]
Li, S.; Huang, L. In vivo gene transfer via intravenous administration of cationic lipid–protamine–DNA (LPD) complexes. Gene Ther., 1997, 4(9), 891-900.
[http://dx.doi.org/10.1038/sj.gt.3300482] [PMID: 9349425]
[71]
Moon, J.J.; Suh, H.; Polhemus, M.E.; Ockenhouse, C.F.; Yadava, A.; Irvine, D.J. Antigen-displaying lipid-enveloped PLGA nanoparticles as delivery agents for a Plasmodium vivax malaria vaccine. PLoS One, 2012, 7(2), e31472.
[http://dx.doi.org/10.1371/journal.pone.0031472] [PMID: 22328935]
[72]
Facciolà, A.; Visalli, G.; Laganà, P.; La Fauci, V.; Squeri, R.; Pellicanò, G.F.; Nunnari, G.; Trovato, M.; Di Pietro, A. The new era of vaccines: The “nanovaccinology”. Eur. Rev. Med. Pharmacol. Sci., 2019, 23(16), 7163-7182.
[PMID: 31486519]
[73]
Pulendran, B.; Ahmed, R. Immunological mechanisms of vaccination. Nat. Immunol., 2011, 12(6), 509-517.
[http://dx.doi.org/10.1038/ni.2039] [PMID: 21739679]
[74]
Blok, B.A.; Arts, R.J.W.; van Crevel, R.; Benn, C.S.; Netea, M.G. Trained innate immunity as underlying mechanism for the long-term, nonspecific effects of vaccines. J. Leukoc. Biol., 2015, 98(3), 347-356.
[http://dx.doi.org/10.1189/jlb.5RI0315-096R] [PMID: 26150551]
[75]
Demento, S.L.; Siefert, A.L.; Bandyopadhyay, A.; Sharp, F.A.; Fahmy, T.M. Pathogen-associated molecular patterns on biomaterials: A paradigm for engineering new vaccines. Trends Biotechnol., 2011, 29(6), 294-306.
[http://dx.doi.org/10.1016/j.tibtech.2011.02.004] [PMID: 21459467]
[76]
Toy, R.; Roy, K. Engineering nanoparticles to overcome barriers to immunotherapy. Bioeng. Transl. Med., 2016, 1(1), 47-62.
[http://dx.doi.org/10.1002/btm2.10005] [PMID: 29313006]
[77]
Kalluru, R.; Fenaroli, F.; Westmoreland, D.; Ulanova, L.; Maleki, A.; Roos, N.; Paulsen Madsen, M.; Koster, G.; Egge-Jacobsen, W.; Wilson, S.; Roberg-Larsen, H.; Khuller, G.K.; Singh, A.; Nyström, B.; Griffiths, G. Poly(lactide-co-glycolide)-rifampicin nanoparticles efficiently clear Mycobacterium bovis BCG infection in macrophages and remain membrane-bound in phago-lysosomes. J. Cell Sci., 2013, 126(Pt 14), 3043-3054.
[PMID: 23687375]
[78]
Vasievich, E.A.; Chen, W.; Huang, L. Enantiospecific adjuvant activity of cationic lipid DOTAP in cancer vaccine. Cancer Immunol. Immunother., 2011, 60(5), 629-638.
[http://dx.doi.org/10.1007/s00262-011-0970-1] [PMID: 21267720]
[79]
Gross, B.P.; Wongrakpanich, A.; Francis, M.B.; Salem, A.K.; Norian, L.A. A therapeutic microparticle-based tumor lysate vaccine reduces spontaneous metastases in murine breast cancer. AAPS J., 2014, 16(6), 1194-1203.
[http://dx.doi.org/10.1208/s12248-014-9662-z] [PMID: 25224145]
[80]
Restifo, N.P.; Dudley, M.E.; Rosenberg, S.A. Adoptive immunotherapy for cancer: Harnessing the T cell response. Nat. Rev. Immunol., 2012, 12(4), 269-281.
[http://dx.doi.org/10.1038/nri3191] [PMID: 22437939]
[81]
Hussein, W.M.; Liu, T.Y.; Jia, Z.; McMillan, N.A.J.; Monteiro, M.J.; Toth, I.; Skwarczynski, M. Multiantigenic peptide–polymer conjugates as therapeutic vaccines against cervical cancer. Bioorg. Med. Chem., 2016, 24(18), 4372-4380.
[http://dx.doi.org/10.1016/j.bmc.2016.07.036] [PMID: 27475535]
[82]
Walsh, K.P.; Mills, K.H.G. Dendritic cells and other innate determinants of T helper cell polarisation. Trends Immunol., 2013, 34(11), 521-530.
[http://dx.doi.org/10.1016/j.it.2013.07.006] [PMID: 23973621]
[83]
Tran, E.; Turcotte, S.; Gros, A.; Robbins, P.F.; Lu, Y.C.; Dudley, M.E.; Wunderlich, J.R.; Somerville, R.P.; Hogan, K.; Hinrichs, C.S.; Parkhurst, M.R.; Yang, J.C.; Rosenberg, S.A. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science, 2014, 344(6184), 641-645.
[http://dx.doi.org/10.1126/science.1251102] [PMID: 24812403]
[84]
Berkowska, M.A.; Driessen, G.J.A.; Bikos, V.; Grosserichter-Wagener, C.; Stamatopoulos, K.; Cerutti, A.; He, B.; Biermann, K.; Lange, J.F.; van der Burg, M.; van Dongen, J.J.M.; van Zelm, M.C. Human memory B cells originate from three distinct germinal center-dependent and -independent maturation pathways. Blood, 2011, 118(8), 2150-2158.
[http://dx.doi.org/10.1182/blood-2011-04-345579] [PMID: 21690558]
[85]
Huang, H.; Benoist, C.; Mathis, D. Rituximab specifically depletes short-lived autoreactive plasma cells in a mouse model of inflammatory arthritis. Proc. Natl. Acad. Sci., 2010, 107(10), 4658-4663.
[http://dx.doi.org/10.1073/pnas.1001074107] [PMID: 20176942]
[86]
Levine, T.P.; Chain, B.M.; Brodsky, F. The cell biology of antigen processing. Crit. Rev. Biochem. Mol. Biol., 1991, 26(5-6), 439-473.
[http://dx.doi.org/10.3109/10409239109086790] [PMID: 1722142]
[87]
Seidman, J.C.; Richard, S.A.; Viboud, C.; Miller, M.A. Quantitative review of antibody response to inactivated seasonal influenza vaccines. Influenza Other Respir. Viruses, 2012, 6(1), 52-62.
[http://dx.doi.org/10.1111/j.1750-2659.2011.00268.x] [PMID: 21668661]
[88]
McHugh, K.J.; Guarecuco, R.; Langer, R.; Jaklenec, A. Single-injection vaccines: Progress, challenges, and opportunities. J. Control. Release, 2015, 219, 596-609.
[http://dx.doi.org/10.1016/j.jconrel.2015.07.029] [PMID: 26254198]
[89]
de Titta, A.; Ballester, M.; Julier, Z.; Nembrini, C.; Jeanbart, L.; van der Vlies, A.J.; Swartz, M.A.; Hubbell, J.A. Nanoparticle conjugation of CpG enhances adjuvancy for cellular immunity and memory recall at low dose. Proc. Natl. Acad. Sci., 2013, 110(49), 19902-19907.
[http://dx.doi.org/10.1073/pnas.1313152110] [PMID: 24248387]
[90]
Demento, S.L.; Cui, W.; Criscione, J.M.; Stern, E.; Tulipan, J.; Kaech, S.M.; Fahmy, T.M. Role of sustained antigen release from nanoparticle vaccines in shaping the T cell memory phenotype. Biomaterials, 2012, 33(19), 4957-4964.
[http://dx.doi.org/10.1016/j.biomaterials.2012.03.041] [PMID: 22484047]
[91]
Rice-Ficht, A.C.; Arenas-Gamboa, A.M.; Kahl-McDonagh, M.M.; Ficht, T.A. Polymeric particles in vaccine delivery. Curr. Opin. Microbiol., 2010, 13(1), 106-112.
[http://dx.doi.org/10.1016/j.mib.2009.12.001] [PMID: 20079678]
[92]
Reddy, S.T.; van der Vlies, A.J.; Simeoni, E.; Angeli, V.; Randolph, G.J.; O’Neil, C.P.; Lee, L.K.; Swartz, M.A.; Hubbell, J.A. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat. Biotechnol., 2007, 25(10), 1159-1164.
[http://dx.doi.org/10.1038/nbt1332] [PMID: 17873867]
[93]
Kanchan, V.; Katare, Y.K.; Panda, A.K. Memory antibody response from antigen loaded polymer particles and the effect of antigen release kinetics. Biomaterials, 2009, 30(27), 4763-4776.
[http://dx.doi.org/10.1016/j.biomaterials.2009.05.075] [PMID: 19540583]
[94]
Fan, Y.; Moon, J. Nanoparticle drug delivery systems designed to improve cancer vaccines and immunotherapy. Vaccines, 2015, 3(3), 662-685.
[http://dx.doi.org/10.3390/vaccines3030662] [PMID: 26350600]
[95]
Park, Y.M.; Lee, S.J.; Kim, Y.S.; Lee, M.H.; Cha, G.S.; Jung, I.D.; Kang, T.H.; Han, H.D. Nanoparticle-based vaccine delivery for cancer immunotherapy. Immune Netw., 2013, 13(5), 177-183.
[http://dx.doi.org/10.4110/in.2013.13.5.177] [PMID: 24198742]
[96]
Yadav, H.K.S.; Dibi, M.; Mohammad, A.; Srouji, A.E. Nanovaccines formulation and applications-a review. J. Drug Deliv. Sci. Technol., 2018, 44, 380-387.
[http://dx.doi.org/10.1016/j.jddst.2018.01.015]
[97]
Shi, G.N.; Zhang, C.N.; Xu, R.; Niu, J.F.; Song, H.J.; Zhang, X.Y.; Wang, W.W.; Wang, Y.M.; Li, C.; Wei, X.Q.; Kong, D.L. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials, 2017, 113, 191-202.
[http://dx.doi.org/10.1016/j.biomaterials.2016.10.047] [PMID: 27816821]
[98]
Zeng, Q.; Li, H.; Jiang, H.; Yu, J.; Wang, Y.; Ke, H.; Gong, T.; Zhang, Z.; Sun, X. Tailoring polymeric hybrid micelles with lymph node targeting ability to improve the potency of cancer vaccines. Biomaterials, 2017, 122, 105-113.
[http://dx.doi.org/10.1016/j.biomaterials.2017.01.010] [PMID: 28110170]
[99]
D’Amico, C.; Fontana, F.; Cheng, R.; Santos, H.A. Development of vaccine formulations: Past, present, and future. Drug Deliv. Transl. Res., 2021, 11(2), 353-372.
[http://dx.doi.org/10.1007/s13346-021-00924-7] [PMID: 33598818]
[100]
Tang, Y.; Fan, W.; Chen, G.; Zhang, M.; Tang, X.; Wang, H.; Zhao, P.; Xu, Q.; Wu, Z.; Lin, X.; Huang, Y. Recombinant cancer nanovaccine for targeting tumor-associated macrophage and remodeling tumor microenvironment. Nano Today, 2021, 40, 101244.
[http://dx.doi.org/10.1016/j.nantod.2021.101244]
[101]
Jiang, M.; Zhao, L.; Cui, X.; Wu, X.; Zhang, Y.; Guan, X.; Ma, J.; Zhang, W. Cooperating minimalist nanovaccine with PD-1 blockade for effective and feasible cancer immunotherapy. J. Adv. Res., 2022, 35, 49-60.
[http://dx.doi.org/10.1016/j.jare.2021.08.011] [PMID: 35003793]
[102]
Wang, D.; Gu, W.; Chen, W.; Zhou, J.; Yu, L.; Kook Kim, B.; Zhang, X.; Seung Kim, J. Advanced nanovaccines based on engineering nanomaterials for accurately enhanced cancer immunotherapy. Coord. Chem. Rev., 2022, 472, 214788.
[http://dx.doi.org/10.1016/j.ccr.2022.214788]
[103]
Xu, F.; Yuan, Y.; Wang, Y.; Yin, Q. Emerging peptide-based nanovaccines: From design synthesis to defense against cancer and infection. Biomed. Pharmacother., 2023, 158, 114117.
[http://dx.doi.org/10.1016/j.biopha.2022.114117] [PMID: 36528914]
[104]
Yi, Y.; Yu, M.; Li, W.; Zhu, D.; Mei, L.; Ou, M. Vaccine-like nanomedicine for cancer immunotherapy. J. Control. Release, 2023, 355, 760-778.
[http://dx.doi.org/10.1016/j.jconrel.2023.02.015] [PMID: 36822241]
[105]
Vasil’ev Iu, M. Avian influenza vaccines. Vopr. Virusol., 2008, 53(6), 4-15.
[PMID: 19172900]
[106]
Neuhaus, V.; Chichester, J.A.; Ebensen, T.; Schwarz, K.; Hartman, C.E.; Shoji, Y.; Guzmán, C.A.; Yusibov, V.; Sewald, K.; Braun, A. A new adjuvanted nanoparticle-based H1N1 influenza vaccine induced antigen-specific local mucosal and systemic immune responses after administration into the lung. Vaccine, 2014, 32(26), 3216-3222.
[http://dx.doi.org/10.1016/j.vaccine.2014.04.011] [PMID: 24731807]
[107]
Dhakal, S.; Goodman, J.; Bondra, K.; Lakshmanappa, Y.S.; Hiremath, J.; Shyu, D.L.; Ouyang, K.; Kang, K.; Krakowka, S.; Wannemuehler, M.J.; Won Lee, C.; Narasimhan, B.; Renukaradhya, G.J. Polyanhydride nanovaccine against swine influenza virus in pigs. Vaccine, 2017, 35(8), 1124-1131.
[http://dx.doi.org/10.1016/j.vaccine.2017.01.019] [PMID: 28117173]
[108]
Ross, K.A.; Loyd, H.; Wu, W.; Huntimer, L.; Ahmed, S.; Sambol, A.; Broderick, S.; Flickinger, Z.; Rajan, K.; Bronich, T.; Mallapragada, S.; Wannemuehler, M.J.; Carpenter, S.; Narasimhan, B. Hemagglutinin-based polyanhydride nanovaccines against H5N1 influenza elicit protective virus neutralizing titers and cell-mediated immunity. Int. J. Nanomedicine, 2014, 10, 229-243.
[PMID: 25565816]
[109]
Kumar, R.; Ray, P.C.; Datta, D.; Bansal, G.P.; Angov, E.; Kumar, N. Nanovaccines for malaria using Plasmodium falciparum antigen Pfs25 attached gold nanoparticles. Vaccine, 2015, 33(39), 5064-5071.
[http://dx.doi.org/10.1016/j.vaccine.2015.08.025] [PMID: 26299750]
[110]
Ansari, M.A.; Zubair, S.; Mahmood, A.; Gupta, P.; Khan, A.A.; Gupta, U.D.; Arora, A.; Owais, M. RD antigen based nanovaccine imparts long term protection by inducing memory response against experimental murine tuberculosis. PLoS One, 2011, 6(8), e22889.
[http://dx.doi.org/10.1371/journal.pone.0022889] [PMID: 21853054]
[111]
Dhanasooraj, D.; Kumar, R.A.; Mundayoor, S. Vaccine delivery system for tuberculosis based on nano-sized hepatitis B virus core protein particles. Int. J. Nanomedicine, 2013, 8, 835-843.
[PMID: 23486691]
[112]
Chesson, C.B.; Huante, M.; Nusbaum, R.J.; Walker, A.G.; Clover, T.M.; Chinnaswamy, J.; Endsley, J.J.; Rudra, J.S. Nanoscale peptide self-assemblies boost BCG-primed cellular immunity against mycobacterium tuberculosis. Sci. Rep., 2018, 8(1), 12519.
[http://dx.doi.org/10.1038/s41598-018-31089-y] [PMID: 30131591]
[113]
Aikins, M.E.; Bazzill, J.; Moon, J.J. Vaccine nanoparticles for protection against HIV infection. Nanomedicine, 2017, 12(6), 673-682.
[http://dx.doi.org/10.2217/nnm-2016-0381] [PMID: 28244816]
[114]
Rostami, H.; Ebtekar, M.; Ardestani, M.S.; Yazdi, M.H.; Mahdavi, M. Co-utilization of a TLR5 agonist and nano-formulation of HIV-1 vaccine candidate leads to increased vaccine immunogenicity and decreased immunogenic dose: A preliminary study. Immunol. Lett., 2017, 187, 19-26.
[http://dx.doi.org/10.1016/j.imlet.2017.05.002] [PMID: 28479111]
[115]
a) Malik, T.; Chauhan, G.; Rath, G.; Kesarkar, R. N.; Chowdhary, A. S.; Goyal, A. K. Efaverinz and nano-gold-loaded mannosylated niosomes: A host cell-targeted topical HIV-1 prophylaxis via thermogel system. Artificial cells, nanomedicine, and biotechnology, 2018, 46(1), 79-90.
[http://dx.doi.org/10.1080/21691401.2017.1414054];
b) Gazzi A, Fusco L, Orecchioni M, Ferrari S, Franzoni G, Yan JS, et al. Graphene, other carbon nanomaterials and the immune system: toward nanoimmunity-by-design. J. Phys. Mater., 2020, 3(3), 034009.
[116]
Al-Hatamleh, M.A.I.; Hatmal, M.M.; Alshaer, W.; Rahman, E.N.S.E.A.; Mohd-Zahid, M.H.; Alhaj-Qasem, D.M.; Yean, C.Y.; Alias, I.Z.; Jaafar, J.; Ferji, K.; Six, J.L. Uskoković V.; Yabu, H.; Mohamud, R. COVID-19 infection and nanomedicine applications for development of vaccines and therapeutics: An overview and future perspectives based on polymersomes. Eur. J. Pharmacol., 2021, 896, 173930.
[http://dx.doi.org/10.1016/j.ejphar.2021.173930] [PMID: 33545157]
[117]
Singh Sekh, B. Nanoprobes and their applications in veterinary medicine and animal health. Res. J. Nanoscience and Nano., 2012, 2(1), 1-16.
[http://dx.doi.org/10.3923/rjnn.2012.1.16] [PMID: 22523944]
[118]
In application of nano-vaccines in veterinary medicine, 2007.
[119]
Brock, K.V. The persistence of bovine viral diarrhea virus. Biologicals. Journal of the International Association of Biological Standardization, 2003, 31(2), 133-135.
[120]
Bruschke, C.; Moormann, R.J.; van Oirschot, J.T.; van Rijn, P.A. A subunit vaccine based on glycoprotein E2 of bovine virus diarrhea virus induces fetal protection in sheep against homologous challenge. Vaccine, 1997, 15(17-18), 1940-1945.
[http://dx.doi.org/10.1016/S0264-410X(97)00125-4] [PMID: 9413105]
[121]
Thomas, C.; Young, N.J.; Heaney, J.; Collins, M.E.; Brownlie, J. Evaluation of efficacy of mammalian and baculovirus expressed E2 subunit vaccine candidates to bovine viral diarrhoea virus. Vaccine, 2009, 27(17), 2387-2393.
[http://dx.doi.org/10.1016/j.vaccine.2009.02.010] [PMID: 19428855]
[122]
Pecora, A.; Aguirreburualde, M.S.P.; Aguirreburualde, A.; Leunda, M.R.; Odeon, A.; Chiavenna, S.; Bochoeyer, D.; Spitteler, M.; Filippi, J.L.; Dus Santos, M.J.; Levy, S.M.; Wigdorovitz, A. Safety and efficacy of an E2 glycoprotein subunit vaccine produced in mammalian cells to prevent experimental infection with bovine viral diarrhoea virus in cattle. Vet. Res. Commun., 2012, 36(3), 157-164.
[http://dx.doi.org/10.1007/s11259-012-9526-x] [PMID: 22639081]
[123]
Snider, M.; Garg, R.; Brownlie, R.; van den Hurk, J.V.; Hurk, S.D.L. The bovine viral diarrhea virus E2 protein formulated with a novel adjuvant induces strong, balanced immune responses and provides protection from viral challenge in cattle. Vaccine, 2014, 32(50), 6758-6764.
[http://dx.doi.org/10.1016/j.vaccine.2014.10.010] [PMID: 25454860]
[124]
Mahony, D.; Cavallaro, A.S.; Mody, K.T.; Xiong, L.; Mahony, T.J.; Qiao, S.Z.; Mitter, N. In vivo delivery of bovine viral diahorrea virus, E2 protein using hollow mesoporous silica nanoparticles. Nanoscale, 2014, 6(12), 6617-6626.
[http://dx.doi.org/10.1039/C4NR01202J] [PMID: 24811899]
[125]
Mody, K.T.; Mahony, D.; Cavallaro, A.S.; Zhang, J.; Zhang, B.; Mahony, T.J.; Yu, C.; Mitter, N. Silica Vesicle Nanovaccine Formulations Stimulate Long-Term Immune responses to the bovine viral diarrhoea virus E2 protein. PLoS One, 2015, 10(12), e0143507.
[http://dx.doi.org/10.1371/journal.pone.0143507] [PMID: 26630001]
[126]
Mahony, D.; Mody, K.T.; Cavallaro, A.S.; Hu, Q.; Mahony, T.J.; Qiao, S.; Mitter, N. Immunisation of sheep with bovine viral diarrhoea virus, e2 protein using a freeze-dried hollow silica mesoporous nanoparticle formulation. PLoS One, 2015, 10(11), e0141870.
[http://dx.doi.org/10.1371/journal.pone.0141870] [PMID: 26535891]
[127]
Maina, T.W.; Grego, E.A.; Boggiatto, P.M.; Sacco, R.E.; Narasimhan, B.; McGill, J.L. Applications of nanovaccines for disease prevention in cattle. Front. Bioeng. Biotechnol., 2020, 8, 608050.
[http://dx.doi.org/10.3389/fbioe.2020.608050] [PMID: 33363134]
[128]
Thukral, A.; Ross, K.; Hansen, C.; Phanse, Y.; Narasimhan, B.; Steinberg, H.; Talaat, A.M. A single dose polyanhydride-based nanovaccine against paratuberculosis infection. NPJ Vaccines, 2020, 5(1), 15.
[http://dx.doi.org/10.1038/s41541-020-0164-y] [PMID: 32128256]
[129]
Bernocchi, B. Porous maltodextrin nanoparticles for the intranasal delivery of vaccines Nanoparticules de maltodextrine pour l’administration intranasale des vaccins; , 2016. Available from: https://theses.hal.science/tel-01480950/document
[130]
Bernocchi, B.; Carpentier, R.; Betbeder, D. Nasal nanovaccines. Int. J. Pharm., 2017, 530(1-2), 128-138.
[http://dx.doi.org/10.1016/j.ijpharm.2017.07.012] [PMID: 28698066]
[131]
Hafner, A. Lovrić J.; Lakoš, G.P.; Pepić I. Nanotherapeutics in the EU: An overview on current state and future directions. Int. J. Nanomedicine, 2014, 9, 1005-1023.
[PMID: 24600222]
[132]
Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal formulations in clinical use: An updated review. Pharmaceutics, 2017, 9(4), 12.
[http://dx.doi.org/10.3390/pharmaceutics9020012] [PMID: 28346375]
[133]
Pereira, V.B.; Zurita-Turk, M.; Saraiva, T.D.L.; De Castro, C.P.; Souza, B.M.; Mancha Agresti, P.; Lima, F.A.; Pfeiffer, V.N.; Azevedo, M.S.P.; Rocha, C.S.; Pontes, D.S.; Azevedo, V.; Miyoshi, A. DNA vaccines approach: From concepts to applications. World J. Vaccines, 2014, 4(2), 50-71.
[http://dx.doi.org/10.4236/wjv.2014.42008]
[134]
Dewangan, H.K.; Raghuvanshi, A.; Shah, K. Emerging trends and future challenges of nanovaccine delivery via nasal route. Curr. Drug Targets, 2022.
[PMID: 36475350]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy