2,159
Views
24
CrossRef citations to date
0
Altmetric
Review

Nanovaccine: an emerging strategy

&
Pages 1273-1290 | Received 02 Jun 2021, Accepted 21 Sep 2021, Published online: 29 Sep 2021

References

  • Zhao L, Seth A, Wibowo N, et al. Nanoparticle vaccines. Vaccine. 2014;32:327–337.
  • HogenEsch H, O’Hagan DT, Fox CB. Optimizing the utilization of aluminum adjuvants in vaccines: you might just get what you want. NPJ Vaccines. 2018;3:51.
  • O’Hagan DT, Friedland LR, Hanon E, et al. Towards an evidence based approach for the development of adjuvanted vaccines. Curr Opin Immunol. 2017;47:93–102.
  • Fries CN, Curvino EJ, Chen JL, et al. Advances in nanomaterial vaccine strategies to address infectious diseases impacting global health. Nat Nanotechnol. 2021;16:1–14.
  • Petkar KC, Patil SM, Chavhan SS, et al. An overview of nanocarrier-based adjuvants for vaccine delivery. Pharmaceutics. 2021;13:455.
  • Pardi N, Hogan MJ, Porter FW, et al. mRNA vaccines — a new era in vaccinology. Nat Rev Drug Discov. 2018;17:261–279.
  • Sharma J, Shepardson K, Johns LL, et al. A self-adjuvanted, modular, antigenic VLP for rapid response to influenza virus variability. ACS Appl Mater Interfaces. 2020;12:18211–18224.
  • Shin MD, Shukla S, Chung YH, et al. COVID-19 vaccine development and a potential nanomaterial path forward. Nat Nanotechnol. 2020;15:646–655.
  • Kaczanowska S, Joseph AM, Davila E. TLR agonists: our best frenemy in cancer immunotherapy. J Leukoc Biol. 2013;93:847–863.
  • Greenwood B. The contribution of vaccination to global health: past, present and future. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130433.
  • Akkaya M, Kwak K, Pierce SK. B cell memory: building two walls of protection against pathogens. Nat Rev Immunol. 2020;20:229–238.
  • Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles to new developments. Nat Rev Immunol. 2021;21:83–100.
  • Pulendran BS, Arunachalam P, O’Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov. 2021;6:1–22.
  • Mandal S, Khandalavala K, Pham R, et al. Cellulose acetate phthalate and antiretroviral nanoparticle fabrications for hiv pre-exposure prophylaxis. Polymers (Basel). 2017;9:423.
  • Jones RB, Mueller S, Kumari S, et al. Antigen recognition-triggered drug delivery mediated by nanocapsule-functionalized cytotoxic T-cells. Biomaterials. 2017;117:44–53.
  • Brouwer PJM, Antanasijevic A, Berndsen Z, et al. Enhancing and shaping the immunogenicity of native-like HIV-1 envelope trimers with a two-component protein nanoparticle. Nat Commun. 2019;10:4272.
  • Pardi N, LaBranche CC, Ferrari G, et al. Characterization of HIV-1 nucleoside-modified mrna vaccines in rabbits and rhesus macaques. Mol Ther Nucleic Acids. 2019;15:36–47.
  • Aroh C, Wang Z, Dobbs N, et al. Innate immune activation by cGMP-AMP nanoparticles leads to potent and long-acting antiretroviral response against HIV-1. J Immunol. 2017;199:3840–3848.
  • Shahbazi R, Sghia-Hughes G, Reid JL, et al. Targeted homology-directed repair in blood stem and progenitor cells with CRISPR nanoformulations. Nat Mater. 2019;18:1124–1132.
  • Ardolino L, Joshua A. Immune checkpoint inhibitors in malignancy. Aust Prescr. 2019;42:62–67.
  • Duan X, Chan C, Han W, et al. Immunostimulatory nanomedicines synergize with checkpoint blockade immunotherapy to eradicate colorectal tumors. Nat Commun. 2019;10:1899.
  • Fontana F, Fusciello M, Groeneveldt C, et al. Biohybrid vaccines for improved treatment of aggressive melanoma with checkpoint inhibitor. ACS Nano. 2019;13:6477–6490.
  • Alkie TN, Yitbarek A, Taha-Abdelaziz K, et al. Characterization of immunogenicity of avian influenza antigens encapsulated in PLGA nanoparticles following mucosal and subcutaneous delivery in chickens. PLoS One. 2018 Nov 1;13:e0206324.
  • Brignole C, Pastorino F, Marimpietri D, et al. Immune cell-mediated antitumor activities of GD2-targeted liposomal c-myb antisense oligonucleotides containing CpG motifs. J Natl Cancer Inst. 2004;96:1171–1180.
  • Wiedermann U, Wiltschke C, Jasinska J, et al. A virosomal formulated Her-2/neu multi-peptide vaccine induces Her-2/neu-specific immune responses in patients with metastatic breast cancer: a phase I study. Breast Cancer Res Treat. 2010;119:673–683.
  • GlaxoSmithKline Vaccine HPV-007 Study Group, Romanowski B, de Borba PC, et al. Sustained efficacy and immunogenicity of the human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine: analysis of a randomised placebo-controlled trial up to 6.4 years. Lancet. 2009;374:1975–1985.
  • Villa LL, Costa RL, Petta CA, et al. Prophylactic quadrivalent human papillomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in young women: a randomised double-blind placebo-controlled multicentre phase II efficacy trial. Lancet Oncol. 2005;6:271–278.
  • Xu Z, Ramishetti S, Tseng YC, et al. Multifunctional nanoparticles co-delivering Trp2 peptide and CpG adjuvant induce potent cytotoxic T-lymphocyte response against melanoma and its lung metastasis. J Control Release. 2013;172:259–265.
  • Zhong Z, Wei X, Qi B, et al. A novel liposomal vaccine improves humoral immunity and prevents tumor pulmonary metastasis in mice. Int J Pharm. 2010;399:156–162.
  • Jia C, Yang T, Liu Y, et al. A novel human papillomavirus 16 L1 pentamer-loaded hybrid particles vaccine system: influence of size on immune responses. ACS Appl Mater Interfaces. 2018;10:35745–35759.
  • Zhao Y, Wang H, Yang Y, et al. Mannose-modified liposome co-delivery of human papillomavirus type 16 E7 peptide and CpG oligodeoxynucleotide adjuvant enhances antitumor activity against established large TC-1 grafted tumors in mice. Int J Nanomedicine. 2020;15:9571–9586.
  • Liu L, Wang Y, Miao L, et al. Combination Immunotherapy of MUC1 mRNA nano-vaccine and CTLA-4 blockade effectively inhibits growth of triple negative breast cancer. Mol Ther. 2018;26:45–55.
  • Cheng R, Fontana F, Xiao J, et al. Recombination monophosphoryl lipid a-derived vacosome for the development of preventive cancer vaccines. ACS Appl Mater Interfaces. 2020;12:44554–44562.
  • Butts C, Maksymiuk A, Goss G, et al. Updated survival analysis in patients with stage IIIB or IV non-small-cell lung cancer receiving BLP25 liposome vaccine (L-BLP25): phase IIB randomized, multicenter, open-label trial. J Cancer Res Clin Oncol. 2011;137:1337–1342.
  • Gulla SK, Rao BR, Moku G, et al. In vivo targeting of DNA vaccines to dendritic cells using functionalized gold nanoparticles. Biomater Sci. 2019;7:773–788.
  • Aikins ME, Xu C, Moon JJ. Engineered nanoparticles for cancer vaccination and immunotherapy. Acc Chem Res. 2020;53:2094–2105.
  • Jeyanathan M, Afkhami S, Smaill F, et al. Immunological considerations for COVID-19 vaccine strategies. Nat Rev Immunol. 2020;20:615–632.
  • Ni L, Ye F, Cheng ML, et al. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. 2020;52:971–977.
  • Mantovani A, Netea MG. Trained innate immunity, epigenetics, and Covid-19. N Engl J Med. 2020;383:1078–1080.
  • Zhu M, Wang R, Nie G. Applications of nanomaterials as vaccine adjuvants. Hum Vaccin Immunother. 2014;10:2761–2774.
  • Ghiringhelli F, Apetoh L, Tesniere A, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β–dependent adaptive immunity against tumors. Nat Med. 2009;15:1170–1178.
  • Orr MT, Khandhar AP, Seydoux E, et al. Reprogramming the adjuvant properties of aluminum oxyhydroxide with nanoparticle technology. NPJ Vaccines. 2019;4:1.
  • 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.
  • Wang N, Wei C, Zhang Z, et al. Aluminum nanoparticles acting as a pulmonary vaccine adjuvant-delivery system (VADS) able to safely elicit robust systemic and mucosal immunity. J Inorg Organomet Polym Mater. 2020;9:1–15.
  • Ferrando RM, Lay L, Polito L. Gold nanoparticle-based platforms for vaccine development. Drug Discov Today. 2021. DOI:https://doi.org/10.1016/j.ddtec.2021.02.001
  • Talamini L, Violatto MB, Cai Q, et al. Influence of size and shape on the anatomical distribution of endotoxin-free gold nanoparticles. ACS Nano. 2017;11:5519–5529.
  • Almeida JPM, Lin AY, Figueroa ER, et al. In vivo gold nanoparticle delivery of peptide vaccine induces anti-tumor immune response in prophylactic and therapeutic tumor models. Small. 2015;11:1453–1459.
  • Ballou B, Ernst LA, Andreko S, et al. Sentinel lymph node imaging using quantum dots in mouse tumor models. Bioconjug Chem. 2007;18:389–396.
  • Lai SK, Hida K, Man ST, et al. Privileged delivery of polymer nanoparticles to the perinuclear region of live cells via a non-clathrin, non-degradative pathway. Biomaterials. 2007;28:2876–2884.
  • Kwon YJ, James E, Shastri N, et al. In vivo targeting of dendritic cells for activation of cellular immunity using vaccine carriers based on pH-responsive microparticles. Proc Natl Acad Sci U S A. 2005;102:18264–18268.
  • Novio F. Design of targeted nanostructured coordination polymers (NCPs) for cancer therapy. Molecules. 2020;25:3449.
  • Poon C, Patel AA. Organic and inorganic nanoparticle vaccines for prevention of infectious diseases. Nano Ex. 2020;1:012001.
  • Moon JJ, Suh H, Li AV, et al. Enhancing humoral responses to a malaria antigen with nanoparticle vaccines that expand Tfh cells and promote germinal center induction. Proc Natl Acad Sci U S A. 2012;109:1080–1085.
  • Fan Y, Stronsky SM, Xu Y, et al. Multilamellar vaccine particle elicits potent immune activation with protein antigens and protects mice against ebola virus infection. ACS Nano. 2019;13:11087–11096.
  • Garinot M, Piras-Douce F, Probeck P, et al. A potent novel vaccine adjuvant based on straight polyacrylate. Int J Pharm X. 2020;2:100054.
  • Wegmann F, Moghaddam AE, Schiffner T, et al. The carbomer-lecithin adjuvant adjuplex has potent immunoactivating properties and elicits protective adaptive immunity against influenza virus challenge in mice. Clin Vaccine Immunol. 2015;22:1004–1012.
  • Pavot V, Bisceglia H, Guillaume F, et al. A novel vaccine adjuvant based on straight polyacrylate potentiates vaccine-induced humoral and cellular immunity in cynomolgus macaques. Hum Vaccin Immunother. 2021;17: 2336-2348.
  • Amin MK, Boateng JS. Comparison and process optimization of PLGA, chitosan and silica nanoparticles for potential oral vaccine delivery. Ther Deliv. 2019;10:493–514.
  • Xu Y, Yuen PW, Lam JK. Intranasal DNA vaccine for protection against respiratory infectious diseases: the delivery perspectives. Pharmaceutics. 2014;6:378–415.
  • MacLaughlin FC, Mumper RJ, Wang J, et al. Chitosan and depolymerized chitosan oligomers as condensing carriers for in vivo plasmid delivery. J Control Release. 1998;56:259–272.
  • AbdelAllah NH, Gaber Y, Rashed ME, et al. Alginate-coated chitosan nanoparticles act as effective adjuvant for hepatitis A vaccine in mice. Int J Biol Macromol. 2020;152:904–912.
  • Mumper RJ, Rolland A. 8. Chitosan and chitosan oligomers for nucleic acid delivery: original research article: chitosan and depolymerized chitosan oligomers as condensing carriers for in vivo plasmid delivery, 1998. J Control Release. 2014;190:46–48.
  • Abouelmagd SA, Ku YJ, Yeo Y. Low molecular weight chitosan-coated polymeric nanoparticles for sustained and pH-sensitive delivery of paclitaxel. J Drug Target. 2015;23:725–735.
  • Debache K, Kropf C, Schütz CA, et al. Vaccination of mice with chitosan nanogel-associated recombinant NcPDI against challenge infection with Neospora caninum tachyzoites. Parasite Immunol. 2011;33:81–94.
  • Şenel S, Yüksel S. Chitosan-based particulate systems for drug and vaccine delivery in the treatment and prevention of neglected tropical diseases. Drug Deliv Transl Res. 2020;10:1644–1674.
  • Pawar D, Mangal S, Goswami R, et al. Development and characterization of surface modified PLGA nanoparticles for nasal vaccine delivery: effect of mucoadhesive coating on antigen uptake and immune adjuvant activity. Eur J Pharm Biopharm. 2013;85:550–559.
  • Slütter 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;28:6282–6291.
  • Hojatizade M, Soleymani M, Tafaghodi M, et al. Chitosan nanoparticles loaded with whole and soluble Leishmania antigens, and evaluation of their immunogenecity in a mouse model of leishmaniasis. Iran J Immunol. 2018;15:281–293.
  • Kasturi SP, Kozlowski PA, Nakaya HI, et al. Adjuvanting a simian immunodeficiency virus vaccine with toll-like receptor ligands encapsulated in nanoparticles induces persistent antibody responses and enhanced protection in trim5α restrictive macaques. J Virol. 2017;91:e01844–16.
  • Tukhvatulin A, Dzharullaeva A, Erokhova A, et al. Adjuvantation of an influenza hemagglutinin antigen with TLR4 and NOD2 agonists encapsulated in poly(D,L-Lactide-Co-Glycolide) nanoparticles enhances immunogenicity and protection against lethal influenza virus infection in mice. Vaccines (Basel). 2020;8:519.
  • Silva AL, Soema PC, Slütter B, et al. PLGA particulate delivery systems for subunit vaccines: linking particle properties to immunogenicity. Hum Vaccin Immunother. 2016;12:1056–1069.
  • Zupančič E, Curato C, Kim JS, et al. Nanoparticulate vaccine inhibits tumor growth via improved T cell recruitment into melanoma and huHER2 breast cancer. Nanomedicine. 2018;14:835–847.
  • Kim H, Griffith TS, Panyam J. Poly(d,l-lactide-co-glycolide) nanoparticles as delivery platforms for TLR7/8 agonist-based cancer vaccine. J Pharmacol Exp Ther. 2019;370:715–724.
  • Luo Z, Wang C, Yi H, et al. Nanovaccine loaded with poly I:C and STAT3 siRNA robustly elicits anti-tumor immune responses through modulating tumor-associated dendritic cells in vivo. Biomaterials. 2015;38:50–60.
  • Adams JR, Haughney SL, Mallapragada SK. Effective polymer adjuvants for sustained delivery of protein subunit vaccines. Acta Biomater. 2015;14:104–114.
  • Kasturi SP, Skountzou I, Albrecht RA, et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature. 2011;470:543–547.
  • Yousefi M, Narmani A, Jafari SM. Dendrimers as efficient nanocarriers for the protection and delivery of bioactive phytochemicals. Adv Colloid Interface Sci. 2020;278:102125.
  • Sherje AP, Jadhav M, Dravyakar BR, et al. Dendrimers: a versatile nanocarrier for drug delivery and targeting. Int J Pharm. 2018;548:707–720.
  • Zaman M, Skwarczynski M, Malcolm JM, et al. Self-adjuvanting polyacrylic nanoparticulate delivery system for group A streptococcus (GAS) vaccine. Nanomedicine. 2011;7:168–173.
  • Xu J, Wang H, Xu L, et al. Nanovaccine based on a protein-delivering dendrimer for effective antigen cross-presentation and cancer immunotherapy. Biomaterials. 2019;207:1–9.
  • Setaro F, Brasch M, Hahn U, et al. Generation-dependent templated self-assembly of biohybrid protein nanoparticles around photosensitizer dendrimers. Nano Lett. 2015;15:1245–1251.
  • Mecke A, Majoros IJ, Patri AK, et al. Lipid bilayer disruption by polycationic polymers: the roles of size and chemical functional group. Langmuir. 2005;21:10348–10354.
  • Chis AA, Dobrea C, Morgovan C, et al. Applications and limitations of dendrimers in biomedicine. Molecules. 2020;25:3982.
  • Hood JL, Wickline SA. A systematic approach to exosome-based translational nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2012;4:458–467.
  • Admyre C, Johansson SM, Paulie S, et al. Direct exosome stimulation of peripheral human T cells detected by ELISPOT. Eur J Immunol. 2006;36:1772–1781.
  • Cheung AS, Koshy ST, Stafford AG, et al. Adjuvant-loaded subcellular vesicles derived from disrupted cancer cells for cancer vaccination. Small. 2016;12:2321–2333.
  • Beauvillain C, Juste MO, Dion S, et al. Exosomes are an effective vaccine against congenital toxoplasmosis in mice. Vaccine. 2009;27:1750–1757.
  • Elashiry M, Elashiry MM, Elsayed R, et al. Dendritic cell derived exosomes loaded with immunoregulatory cargo reprogram local immune responses and inhibit degenerative bone disease in vivo. J Extracell Vesicles. 2020;9:1795362.
  • Whiteside TL, Mandapathil M, Szczepanski M, et al. Mechanisms of tumor escape from the immune system: adenosine-producing Treg, exosomes and tumor-associated TLRs. Bull Cancer. 2011;98:E25–E31.
  • Chen G, Huang AC, Zhang W, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560:382–386.
  • Xu Z, Zeng S, Gong Z, et al. Exosome-based immunotherapy: a promising approach for cancer treatment. Mol Cancer. 2020;19:160.
  • Yan W, Jiang S. Immune cell-derived exosomes in the cancer-immunity cycle. Trends Cancer. 2020;6:506–517.
  • Naseri M, Bozorgmehr M, Zöller M, et al. Tumor-derived exosomes: the next generation of promising cell-free vaccines in cancer immunotherapy. Oncoimmunology. 2020;9:1779991.
  • Hiltbrunner S, Larssen P, Eldh M, et al. Exosomal cancer immunotherapy is independent of MHC molecules on exosomes. Oncotarget. 2016;7:38707–38717.
  • Morse MA, Garst J, Osada T, et al. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med. 2005;3:9.
  • Shenoda BB, Ajit SK. Modulation of immune responses by exosomes derived from antigen-presenting cells. Clin Med Insights Pathol. 2016;9:1–8.
  • Patel JM, Vartabedian VF, Bozeman EN, et al. Plasma membrane vesicles decorated with glycolipid-anchored antigens and adjuvants via protein transfer as an antigen delivery platform for inhibition of tumor growth. Biomaterials. 2016;74:231–244.
  • Donaldson B, Lateef Z, Walker GF, et al. Virus-like particle vaccines: immunology and formulation for clinical translation. Expert Rev Vaccines. 2018;17:833–849.
  • Coleman CM, Venkataraman T, Liu YV, et al. MERS-CoV spike nanoparticles protect mice from MERS-CoV infection. Vaccine. 2017;35:1586–1589.
  • Medhi R, Srinoi P, Ngo N, et al. Nanoparticle-based strategies to combat COVID-19. ACS Appl Nano Mater. 2020;3:acsanm.0c01978.
  • Bungener L, Huckriede A, De Mare A, et al. Virosome-mediated delivery of protein antigens in vivo: efficient induction of class I MHC-restricted cytotoxic T lymphocyte activity. Vaccine. 2005;23:1232–1241.
  • Genito CJ, Batty CJ, Bachelder EM, et al. Considerations for size, surface charge, polymer degradation, co-delivery, and manufacturability in the development of polymeric particle vaccines for infectious diseases. Adv Nanobiomed Res. 2021;18:2000041.
  • Moser C, Müller M, Kaeser MD, et al. Influenza virosomes as vaccine adjuvant and carrier system. Expert Rev Vaccines. 2013;12:779–791.
  • Blom RA, Erni ST, Krempaská K, et al. A triple co-culture model of the human respiratory tract to study immune-modulatory effects of liposomes and virosomes. PLoS One. 2016;11:e0163539.
  • Manolova V, Flace A, Bauer M, et al. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol. 2008;38:1404–1413.
  • Carrasco YR, Batista FD. B cells acquire particulate antigen in a macrophage-rich area at the boundary between the follicle and the subcapsular sinus of the lymph node. Immunity. 2007;27:160–171.
  • Raghuwanshi D, Mishra V, Das D, et al. Dendritic cell targeted chitosan nanoparticles for nasal DNA immunization against SARS CoV nucleocapsid protein. Mol Pharm. 2012;9:946–956.
  • Genton B, Pluschke G, Degen L, et al. A randomized placebo-controlled phase Ia malaria vaccine trial of two virosome-formulated synthetic peptides in healthy adult volunteers. PLoS One. 2007;2:e1018.
  • Cech PG, Aebi T, Abdallah MS, et al. Virosome-formulated Plasmodium falciparum AMA-1 & CSP derived peptides as malaria vaccine: randomized phase 1b trial in semi-immune adults & children. PLoS One. 2011;6:e22273.
  • Okitsu SL, Silvie O, Westerfeld N, et al. A virosomal malaria peptide vaccine elicits a long-lasting sporozoite-inhibitory antibody response in a phase 1a clinical trial. PLoS One. 2007;2:e1278.
  • Amacker M, Smardon C, Mason L, et al. New GMP manufacturing processes to obtain thermostable HIV-1 gp41 virosomes under solid forms for various mucosal vaccination routes. NPJ Vaccines. 2020;5:41.
  • Tao W, Gill HS. M2e-immobilized gold nanoparticles as influenza A vaccine: role of soluble M2e and longevity of protection. Vaccine. 2015;33:2307–2315.
  • Climent N, García I, Marradi M, et al. Loading dendritic cells with gold nanoparticles (GNPs) bearing HIV-peptides and mannosides enhance HIV-specific T cell responses. Nanomedicine. 2018;14:339–351.
  • Kumar R, Ray PC, Datta D, et al. Nanovaccines for malaria using Plasmodium falciparum antigen Pfs25 attached gold nanoparticles. Vaccine. 2015;33:5064–5071.
  • Bento D, Staats HF, Gonçalves T, et al. Development of a novel adjuvanted nasal vaccine: C48/80 associated with chitosan nanoparticles as a path to enhance mucosal immunity. Eur J Pharm Biopharm. 2015;93:149–164.
  • Malik A, Gupta M, Mani R, et al. Single-dose Ag85B-ESAT6-loaded poly(lactic-co-glycolic acid) nanoparticles confer protective immunity against tuberculosis. Int J Nanomedicine. 2019;14:3129–3143.
  • Tosyali OA, Allahverdiyev A, Bagirova M, et al. Nano-co-delivery of lipophosphoglycan with soluble and autoclaved leishmania antigens into PLGA nanoparticles: evaluation of in vitro and in vivo immunostimulatory effects against visceral leishmaniasis. Mater Sci Eng C Mater Biol Appl. 2021;120:111684.
  • Moon JJ, Suh H, Polhemus ME, et al. Antigen-displaying lipid-enveloped PLGA nanoparticles as delivery agents for a Plasmodium vivax malaria vaccine. PLoS One. 2012;7:e31472.
  • Barry MA, Wang Q, Jones KM, et al. A therapeutic nanoparticle vaccine against Trypanosoma cruzi in a BALB/c mouse model of Chagas disease. Hum Vaccin Immunother. 2016;12:976–987.
  • Gao Y, Ji H, Peng L, et al. Development of PLGA-PEG-PLGA hydrogel delivery system for enhanced immunoreaction and efficacy of newcastle disease virus DNA vaccine. Molecules. 2020 May 28;25:2505.
  • Cecílio P, Pérez-Cabezas B, Fernández L, et al. Pre-clinical antigenicity studies of an innovative multivalent vaccine for human visceral leishmaniasis. PLoS Negl Trop Dis. 2017;11:e0005951.
  • Singh K, Marasini B, Chen X, et al. A bivalent, spherical virus-like particle vaccine enhances breadth of immune responses against pathogenic ebola viruses in rhesus macaques. J Virol. 2020;94(9):e01884–19.
  • Branco LM, Grove JN, Geske FJ, et al. Lassa virus-like particles displaying all major immunological determinants as a vaccine candidate for Lassa hemorrhagic fever. Virol J. 2010;7:279.
  • Fries L, Cho I, Krähling V, et al. Randomized, blinded, dose-ranging trial of an ebola virus glycoprotein nanoparticle vaccine with matrix-M adjuvant in healthy adults. J Infect Dis. 2020;222:572–582.
  • Kanekiyo M, Wei CJ, Yassine HM, et al. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature. 2013;499:102–106.
  • Bazzill JD, Stronsky SM, Kalinyak LC, et al. Vaccine nanoparticles displaying recombinant Ebola virus glycoprotein for induction of potent antibody and polyfunctional T cell responses. Nanomedicine. 2019;18:414–425.
  • Lo MK, Spengler JR, Welch SR, et al. Evaluation of a single-dose nucleoside-modified messenger RNA vaccine encoding hendra virus-soluble glycoprotein against lethal nipah virus challenge in syrian hamsters. J Infect Dis. 2020;221(Supplement_4):S493–S498.
  • Sabur A, Bhowmick S, Chhajer R, et al. Liposomal elongation Factor-1α triggers effector CD4 and CD8 T cells for induction of long-lasting protective immunity against visceral leishmaniasis. Front Immunol. 2018;9:18.
  • Nisini R, Poerio N, Mariotti S, et al. The multirole of liposomes in therapy and prevention of infectious diseases. Front Immunol. 2018;9:155.
  • Mazumdar T, Anam K, Ali N. Influence of phospholipid composition on the adjuvanticity and protective efficacy of liposome-encapsulated Leishmania donovani antigens. J Parasitol. 2005;91:269–274.
  • 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.
  • Bernasconi V, Norling K, Bally M, et al. Mucosal vaccine development based on liposome technology. J Immunol Res. 2016;2016:5482087.
  • Bulbake U, Doppalapudi S, Kommineni N, et al. Liposomal formulations in clinical use: an updated review. Pharmaceutics. 2017;9:12.
  • Bhowmick S, Mazumdar T, Sinha R, et al. Comparison of liposome based antigen delivery systems for protection against Leishmania donovani. J Control Release. 2010;141:199–207.
  • De Serrano LO, Burkhart DJ. Liposomal vaccine formulations as prophylactic agents: design considerations for modern vaccines. J Nanobiotechnology. 2017;15:83.
  • Wui SR, Ko A, Ryu JI, et al. The effect of a TLR4 agonist/cationic liposome adjuvant on varicella-zoster virus glycoprotein e vaccine efficacy: antigen presentation, uptake, and delivery to lymph nodes. Pharmaceutics. 2021;13:390.
  • Maji M, Mazumder S, Bhattacharya S, et al. A lipid based antigen delivery system efficiently facilitates mhc class-i antigen presentation in dendritic cells to stimulate CD8(+) T cells. Sci Rep. 2016;6:27206.
  • Das A, Asad M, Sabur A, et al. Monophosphoryl lipid a based cationic liposome facilitates vaccine induced expansion of polyfunctional t cell immune responses against visceral leishmaniasis. ACS Appl Bio Mater. 2018;1:999–1018.
  • Lonez C, Vandenbranden M, Ruysschaert JM. Cationic lipids activate intracellular signaling pathways. Adv Drug Deliv Rev. 2012;64:1749–1758.
  • Das A, Ali N. Vaccine development against Leishmania donovani. Front Immunol. 2012;3:99.
  • Moerk SK, Donia M, Kringelum JV, et al. Pilot study on the feasibility, safety and immunogenicity of a personalized neoantigen-targeted immunotherapy (NeoPepVac) in combination with anti-PD-1 or anti-PD-L1 in advanced solid tumors. Ann Oncol. 2019;30. DOI:https://doi.org/10.1093/annonc/mdz451.019
  • Bayyurt B, Tincer G, Almacioglu K, et al. Encapsulation of two different TLR ligands into liposomes confer protective immunity and prevent tumor development. J Control Release. 2017;247:134–144.
  • Bhowmick S, Mazumdar T, Ali N. Vaccination route that induces transforming growth factor beta production fails to elicit protective immunity against Leishmania donovani infection. Infect Immun. 2009;77:1514–1523.
  • Vosika GJ, Barr C, Gilbertson D. Phase-I study of intravenous modified lipid A. Cancer Immunol Immunother. 1984;18:107–112.
  • Boks MA, Bruijns SCM, Ambrosini M, et al. In situ delivery of tumor antigen- and adjuvant-loaded liposomes boosts antigen-specific T-cell responses by human dermal dendritic cells. J Invest Dermatol. 2015;135:2697–2704.
  • Mazumder S, Maji M, Ali N. Potentiating effects of MPL on DSPC bearing cationic liposomes promote recombinant GP63 vaccine efficacy: high immunogenicity and protection. PLoS Negl Trop Dis. 2011;5:e1429.
  • Das A, Ali N. Combining cationic liposomal delivery with MPL-TDM for cysteine protease cocktail vaccination against Leishmania donovani: evidence for antigen synergy and protection. PLoS Negl Trop Dis. 2015 Oct;9:e0004185.
  • Deshpande PP, Biswas S, Torchilin VP. Current trends in the use of liposomes for tumor targeting. Nanomedicine (Lond). 2013;8:1509–1528.
  • Gu Z, Da Silva CG, Van Der Maaden K, et al. Liposome-based drug delivery systems in cancer immunotherapy. Pharmaceutics. 2020;12:1054.
  • Yuan Y, Zhang L, Cao H, et al. A polyethylenimine-containing and transferrin-conjugated lipid nanoparticle system for antisense oligonucleotide delivery to AML. Biomed Res Int. 2016;2016:1287128.
  • Kang MH, Yoo HJ, Kwon YH, et al. Design of multifunctional liposomal nanocarriers for folate receptor-specific intracellular drug delivery. Mol Pharm. 2015;12:4200–4213.
  • Tie Y, Zheng H, He Z, et al. Targeting folate receptor β positive tumor-associated macrophages in lung cancer with a folate-modified liposomal complex. Signal Transduct Target Ther. 2020;5:6.
  • Fu S, Xu X, Ma Y, et al. RGD peptide-based non-viral gene delivery vectors targeting integrin αvβ3 for cancer therapy. J Drug Target. 2019;27:1–11.
  • Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2 - preliminary report. N Engl J Med. 2020;383:1920–1931.
  • Guevara ML, Persano F, Persano S. Advances in lipid nanoparticles for mRNA-based cancer immunotherapy. Front Chem. 2020;8:589959.
  • Danhier F. To exploit the tumor microenvironment: since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release. 2016;244:108–121.
  • Shah S, Dhawan V, Holm R, et al. Liposomes: advancements and innovation in the manufacturing process. Adv Drug Deliv Rev. 2020;154-155:102–122.
  • Liu C, Zhang L, Zhu W, et al. Barriers and strategies of cationic liposomes for cancer gene therapy. Mol Ther Methods Clin Dev. 2020;18:751–764.
  • Scioli Montoto S, Muraca G, Ruiz ME. Solid lipid nanoparticles for drug delivery: pharmacological and biopharmaceutical aspects. Front Mol Biosci. 2020;7:587997.
  • Müller RH, Mehnert W, Lucks JS, et al. Solid lipid nanoparticles (SLN) — an alternative colloidal carrier system for controlled drug delivery. Eur J Pharm Biopharm. 1995;41:62–69.
  • Doroud D, Vatanara A, Zahedifard F, et al. Cationic solid lipid nanoparticles loaded by cystein proteinase genes as a novel anti-leishmaniasis DNA vaccine delivery system: characterization and in vitro evaluations. J Control Release. 2010;148:e105–6.
  • Li S, Yang Y, Lin X, et al. Biocompatible cationic solid lipid nanoparticles as adjuvants effectively improve humoral and T cell immune response of foot and mouth disease vaccines. Vaccine. 2020;38:2478–2486.
  • Thi TTH, Suys EJA, Lee JS, et al. Lipid-based nanoparticles in the clinic and clinical trials: from cancer nanomedicine to COVID-19 vac cines. Vaccines (Basel). 2021;9:359.
  • Dianzani C, Zara GP, Maina G, et al. Drug delivery nanoparticles in skin cancers. Biomed Res Int. 2014;2014:895986.
  • Milane L, Amiji M. Clinical approval of nanotechnology-based SARS-CoV-2 mRNA vaccines: impact on translational nanomedicine. Drug Deliv Transl Res. 2021;1–7.
  • Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384:403–416.
  • Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383:2603–2615.
  • Li B, Zhang X, Dong Y. Nanoscale platforms for messenger RNA delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11:e1530.
  • Wu Z, Li T. Nanoparticle-mediated cytoplasmic delivery of messenger rna vaccines: challenges and future perspectives. Pharm Res. 2021;38:473–478.
  • Kulkarni JA, Thomson SB, Zaifman J, et al. Spontaneous, solvent-free entrapment of siRNA within lipid nanoparticles. Nanoscale. 2020;12:23959–23966.
  • Blumenthal KG, Robinson LB, Camargo CA, et al. Acute allergic Reactions to mRNA COVID-19 Vaccines. JAMA. 2021;325:1562–1565.
  • Kounis NG, Koniari I, De Gregorio C, et al. Allergic reactions to current available COVID-19 Vaccinations: pathophysiology, causality, and therapeutic considerations. Vaccines (Basel). 2021;9:221.
  • Baz Morelli A, Becher D, Koernig S, et al. ISCOMATRIX: a novel adjuvant for use in prophylactic and therapeutic vaccines against infectious diseases. J Med Microbiol. 2012;61:935–943.
  • Guo Y, Tang L. A magnetic nanovaccine enhances cancer immunotherapy. ACS Cent Sci. 2019;5:747–749.
  • Lei Y, Zhang J, Schiavon CR, et al. SARS-CoV-2 spike protein impairs endothelial function via downregulation of ACE 2. Circ Res. 2021;128:1323–1326.
  • Dan JM, Mateus J, Kato Y, et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science. 2021;371:eabf4063.
  • Turner JS, O’Halloran JA, Kalaidina E, et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature. 2021;596:109–113.
  • Goel RR, Apostolidis SA, Painter MM, et al. Distinct antibody and memory B cell responses in SARS-CoV-2 naïve and recovered individuals following mRNA vaccination. Sci Immunol. 2021;6:eabi6950.
  • Painter MM, Mathew D, Goel RR, et al. Rapid induction of antigen-specific CD4+ T cells is associated with coordinated humoral and cellular immune responses to SARS-CoV-2 mRNA vaccination. Immunity. 2021;54:2133–2142.e3.

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.