201
|
Roth GA, Picece VCTM, Ou BS, Luo W, Pulendran B, Appel EA. Designing spatial and temporal control of vaccine responses. NATURE REVIEWS. MATERIALS 2022; 7:174-195. [PMID: 34603749 PMCID: PMC8477997 DOI: 10.1038/s41578-021-00372-2] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 05/02/2023]
Abstract
Vaccines are the key technology to combat existing and emerging infectious diseases. However, increasing the potency, quality and durability of the vaccine response remains a challenge. As our knowledge of the immune system deepens, it becomes clear that vaccine components must be in the right place at the right time to orchestrate a potent and durable response. Material platforms, such as nanoparticles, hydrogels and microneedles, can be engineered to spatially and temporally control the interactions of vaccine components with immune cells. Materials-based vaccination strategies can augment the immune response by improving innate immune cell activation, creating local inflammatory niches, targeting lymph node delivery and controlling the time frame of vaccine delivery, with the goal of inducing enhanced memory immunity to protect against future infections. In this Review, we highlight the biological mechanisms underlying strong humoral and cell-mediated immune responses and explore materials design strategies to manipulate and control these mechanisms.
Collapse
Affiliation(s)
- Gillie A. Roth
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Vittoria C. T. M. Picece
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Wei Luo
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Paediatrics — Endocrinology, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
202
|
Carmona-Ribeiro AM, Mathiazzi BI, Pérez-Betancourt Y. Cationic Nanostructures as Adjuvants for Vaccines. Methods Mol Biol 2022; 2412:233-245. [PMID: 34918247 DOI: 10.1007/978-1-0716-1892-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Spherical or discoidal lipid polymer nanostructures bearing cationic charges successfully adsorb a variety of oppositely charged antigens (Ag) such as proteins, peptides, nucleic acids, or oligonucleotides. This report provides instructions for the preparation and physical characterization of four different cationic nanostructures able to combine and deliver antigens to the immune system: (1) dioctadecyl dimethylammonium bromide (DODAB) bilayer fragments (DODAB BF); (2) polystyrene sulfate (PSS) nanoparticles (NPs) covered with one cationic dioctadecyl dimethylammonium bromide bilayer (DODAB) named (PSS/DODAB); (3) cationic NPs of biocompatible polymer poly(methyl methacrylate) (PMMA) prepared by emulsion polymerization of the methyl methacrylate (MMA) monomer in the presence of DODAB BF (PMMA/DODAB NPs); (4) antigen NPs (NPs) where the cationic polymer poly(diallyl dimethyl ammonium chloride) (PDDA) directly combined at nontoxic and low dose with the antigen (Ag); when the oppositely charged model antigen is ovalbumin (OVA), NPs are named PDDA/OVA. These nanostructures provide adequate microenvironments for carrying and delivering antigens to the antigen-presenting cells of the immune system.
Collapse
Affiliation(s)
- Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil.
| | - Beatriz Ideriha Mathiazzi
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Yunys Pérez-Betancourt
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
203
|
Current view on novel vaccine technologies to combat human infectious diseases. Appl Microbiol Biotechnol 2022; 106:25-56. [PMID: 34889981 PMCID: PMC8661323 DOI: 10.1007/s00253-021-11713-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Inactivated and live attenuated vaccines have improved human life and significantly reduced morbidity and mortality of several human infectious diseases. However, these vaccines have faults, such as reactivity or suboptimal efficacy and expensive and time-consuming development and production. Additionally, despite the enormous efforts to develop vaccines against some infectious diseases, the traditional technologies have not been successful in achieving this. At the same time, the concerns about emerging and re-emerging diseases urge the need to develop technologies that can be rapidly applied to combat the new challenges. Within the last two decades, the research of vaccine technologies has taken several directions to achieve safe, efficient, and economic platforms or technologies for novel vaccines. This review will give a brief overview of the current state of the novel vaccine technologies, new vaccine candidates in clinical trial phases 1-3 (listed by European Medicines Agency (EMA) and Food and Drug Administration (FDA)), and vaccines based on the novel technologies which have already been commercially available (approved by EMA and FDA) with the special reference to pandemic COVID-19 vaccines. KEY POINTS: • Vaccines of the new generation follow the minimalist strategy. • Some infectious diseases remain a challenge for the vaccine development. • The number of new vaccine candidates in the late phase clinical trials remains low.
Collapse
|
204
|
Pei M, Li H, Zhu Y, Lu J, Zhang C. In vitro evidence of oncofetal antigen and TLR-9 agonist co-delivery by alginate nanovaccine for liver cancer immunotherapy. Biomater Sci 2022; 10:2865-2876. [DOI: 10.1039/d1bm02021h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Liver cancer is the most common malignant tumor and liver cancer immunotherapy has been one of the research hotspots. To induce antigen-specific antitumor immune responses against liver cancer, we developed...
Collapse
|
205
|
Carmona-Ribeiro AM. Supramolecular Nanostructures for Vaccines. Biomimetics (Basel) 2021; 7:6. [PMID: 35076466 PMCID: PMC8788484 DOI: 10.3390/biomimetics7010006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 12/31/2022] Open
Abstract
Although this is an era of pandemics and many devastating diseases, this is also a time when bionanotechnology flourishes, illuminating a multidisciplinary field where vaccines are quickly becoming a balsam and a prevention against insidious plagues. In this work, we tried to gain and also give a deeper understanding on nanovaccines and their way of acting to prevent or cure cancer, infectious diseases, and diseases caused by parasites. Major nanoadjuvants and nanovaccines are temptatively exemplified trying to contextualize our own work and its relative importance to the field. The main properties for novel adjuvants seem to be the nanosize, the cationic character, and the biocompatibility, even if it is achieved in a low dose-dependent manner.
Collapse
Affiliation(s)
- Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenida Professor Lineu Prestes, 748, Butantan, São Paulo CEP 05508-000, SP, Brazil
| |
Collapse
|
206
|
Geisshüsler S, Schineis P, Langer L, Wäckerle-Men Y, Leroux JC, Halin C, Vogel-Kindgen S, Johansen P, Gander B. Amphiphilic Cyclodextrin‐Based Nanoparticulate Vaccines Can Trigger T‐Cell Immune Responses. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Silvana Geisshüsler
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Philipp Schineis
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Lara Langer
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Ying Wäckerle-Men
- Department of Dermatology University of Zurich and University Hospital Zurich Gloriastrasse 31 8091 Zurich Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Sarah Vogel-Kindgen
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Pål Johansen
- Department of Dermatology University of Zurich and University Hospital Zurich Gloriastrasse 31 8091 Zurich Switzerland
| | - Bruno Gander
- Institute of Pharmaceutical Sciences ETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| |
Collapse
|
207
|
Siggins MK, Sriskandan S. Bacterial Lymphatic Metastasis in Infection and Immunity. Cells 2021; 11:33. [PMID: 35011595 PMCID: PMC8750085 DOI: 10.3390/cells11010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Lymphatic vessels permeate tissues around the body, returning fluid from interstitial spaces back to the blood after passage through the lymph nodes, which are important sites for adaptive responses to all types of pathogens. Involvement of the lymphatics in the pathogenesis of bacterial infections is not well studied. Despite offering an obvious conduit for pathogen spread, the lymphatic system has long been regarded to bar the onward progression of most bacteria. There is little direct data on live virulent bacteria, instead understanding is largely inferred from studies investigating immune responses to viruses or antigens in lymph nodes. Recently, we have demonstrated that extracellular bacterial lymphatic metastasis of virulent strains of Streptococcus pyogenes drives systemic infection. Accordingly, it is timely to reconsider the role of lymph nodes as absolute barriers to bacterial dissemination in the lymphatics. Here, we summarise the routes and mechanisms by which an increasing variety of bacteria are acknowledged to transit through the lymphatic system, including those that do not necessarily require internalisation by host cells. We discuss the anatomy of the lymphatics and other factors that influence bacterial dissemination, as well as the consequences of underappreciated bacterial lymphatic metastasis on disease and immunity.
Collapse
Affiliation(s)
- Matthew K. Siggins
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2DD, UK
| |
Collapse
|
208
|
Tsakiri M, Naziris N, Demetzos C. Innovative vaccine platforms against infectious diseases: Under the scope of the COVID-19 pandemic. Int J Pharm 2021; 610:121212. [PMID: 34687816 PMCID: PMC8527590 DOI: 10.1016/j.ijpharm.2021.121212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022]
Abstract
While classic vaccines have proved greatly efficacious in eliminating serious infectious diseases, innovative vaccine platforms open a new pathway to overcome dangerous pandemics via the development of safe and effective formulations. Such platforms play a key role either as antigen delivery systems or as immune-stimulators that induce both innate and adaptive immune responses. Liposomes or lipid nanoparticles, virus-like particles, nanoemulsions, polymeric or inorganic nanoparticles, as well as viral vectors, all belong to the nanoscale and are the main categories of innovative vaccines that are currently on the market or in clinical and preclinical phases. In this paper, we review the above formulations used in vaccinology and we discuss their connection with the development of safe and effective prophylactic vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Nikolaos Naziris
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece.
| |
Collapse
|
209
|
Mochizuki S, Miyamoto N, Sakurai K. Oligonucleotide delivery to antigen presenting cells by using schizophyllan. Drug Metab Pharmacokinet 2021; 42:100434. [PMID: 34896749 DOI: 10.1016/j.dmpk.2021.100434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
Schizophyllan (SPG), a member of the β-glucan family, can form novel complexes with homo-polynucleotides such as poly(dA) through hydrogen bonding between two main chain glucoses and the one nucleotide base. Dectin-1, one of the major receptors for β-glucans, is known to be expressed on antigen presenting cells (APCs) such as macrophages and dendritic cells. This suggests that the above-mentioned complexes could deliver bound functional oligonucleotides (ODNs) including antisense (AS)-ODNs, small interfering RNA, and CpG-ODNs to the APCs. Analysis using a quartz crystal microbalance revealed that a complex consisting of SPG and dA60 with a phosphorothioate backbone was recognized by recombinant Dectin-1 protein. Treatment with this complex containing an AS-ODN for tumor necrosis factor alpha protected mice against lipopolysaccharide-induced hepatitis at a very low AS-ODN dose. Moreover, immunization with CpG-ODN/SPG complex and antigenic proteins induced potent antigen specific immune responses. The present review also represents peptide delivery by conjugation with dA60 and the preparation of a nanogel using DNA-DNA hybridization. These findings indicate that the delivery of a specific ODN using β-glucans could be used for treating various diseases caused by APCs and for activating antigen specific immune responses.
Collapse
Affiliation(s)
- Shinichi Mochizuki
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan.
| | - Noriko Miyamoto
- Department of Applied Chemistry, Aichi Institute of Technology, 1247, Yachigusa, Yakusacho, Toyota, Aichi, 470-0392, Japan
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan
| |
Collapse
|
210
|
Ding Y, Li Z, Jaklenec A, Hu Q. Vaccine delivery systems toward lymph nodes. Adv Drug Deliv Rev 2021; 179:113914. [PMID: 34363861 PMCID: PMC9418125 DOI: 10.1016/j.addr.2021.113914] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022]
Abstract
Strategies of improving vaccine targeting ability toward lymph nodes have been attracting considerable interest in recent years, though there are remaining delivery barriers based on the inherent properties of lymphatic systems and limited administration routes of vaccination. Recently, emerging vaccine delivery systems using various materials as carriers are widely developed to achieve efficient lymph node targeting and improve vaccine-triggered adaptive immune response. In this review, to further optimize the vaccine targeting ability for future research, the design principles of lymph node targeting vaccine delivery based on the anatomy of lymph nodes and vaccine administration routes are first summarized. Then different designs of lymph node targeting vaccine delivery systems, including vaccine delivery systems in clinical applications, are carefully surveyed. Also, the challenges and opportunities of current delivery systems for vaccines are concluded in the end.
Collapse
Affiliation(s)
- Yingyue Ding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Ana Jaklenec
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States.
| |
Collapse
|
211
|
Mast MP, Modh H, Champanhac C, Wang JW, Storm G, Krämer J, Mailänder V, Pastorin G, Wacker MG. Nanomedicine at the crossroads - A quick guide for IVIVC. Adv Drug Deliv Rev 2021; 179:113829. [PMID: 34174332 DOI: 10.1016/j.addr.2021.113829] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/17/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023]
Abstract
For many years, nanomedicine is pushing the boundaries of drug delivery. When applying these novel therapeutics, safety considerations are not only a key concern when entering clinical trials but also an important decision point in product development. Standing at the crossroads, nanomedicine may be able to escape the niche markets and achieve wider acceptance by the pharmaceutical industry. While there is a new generation of drug delivery systems, the extracellular vesicles, standing on the starting line, unresolved issues and new challenges emerge from their translation from bench to bedside. Some key features of injectable nanomedicines contribute to the predictability of the pharmacological and toxicological effects. So far, only a few of the physicochemical attributes of nanomedicines can be justified by a direct mathematical relationship between the in vitro and the in vivo responses. To further develop extracellular vesicles as drug carriers, we have to learn from more than 40 years of clinical experience in liposomal delivery and pass on this knowledge to the next generation. Our quick guide discusses relationships between physicochemical characteristics and the in vivo response, commonly referred to as in vitro-in vivo correlation. Further, we highlight the key role of computational methods, lay open current knowledge gaps, and question the established design strategies. Has the recent progress improved the predictability of targeted delivery or do we need another change in perspective?
Collapse
|
212
|
Zhang P, Narayanan E, Liu Q, Tsybovsky Y, Boswell K, Ding S, Hu Z, Follmann D, Lin Y, Miao H, Schmeisser H, Rogers D, Falcone S, Elbashir SM, Presnyak V, Bahl K, Prabhakaran M, Chen X, Sarfo EK, Ambrozak DR, Gautam R, Martin MA, Swerczek J, Herbert R, Weiss D, Misamore J, Ciaramella G, Himansu S, Stewart-Jones G, McDermott A, Koup RA, Mascola JR, Finzi A, Carfi A, Fauci AS, Lusso P. A multiclade env-gag VLP mRNA vaccine elicits tier-2 HIV-1-neutralizing antibodies and reduces the risk of heterologous SHIV infection in macaques. Nat Med 2021; 27:2234-2245. [PMID: 34887575 DOI: 10.1038/s41591-021-01574-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/06/2021] [Indexed: 12/16/2022]
Abstract
The development of a protective vaccine remains a top priority for the control of the HIV/AIDS pandemic. Here, we show that a messenger RNA (mRNA) vaccine co-expressing membrane-anchored HIV-1 envelope (Env) and simian immunodeficiency virus (SIV) Gag proteins to generate virus-like particles (VLPs) induces antibodies capable of broad neutralization and reduces the risk of infection in rhesus macaques. In mice, immunization with co-formulated env and gag mRNAs was superior to env mRNA alone in inducing neutralizing antibodies. Macaques were primed with a transmitted-founder clade-B env mRNA lacking the N276 glycan, followed by multiple booster immunizations with glycan-repaired autologous and subsequently bivalent heterologous envs (clades A and C). This regimen was highly immunogenic and elicited neutralizing antibodies against the most prevalent (tier-2) HIV-1 strains accompanied by robust anti-Env CD4+ T cell responses. Vaccinated animals had a 79% per-exposure risk reduction upon repeated low-dose mucosal challenges with heterologous tier-2 simian-human immunodeficiency virus (SHIV AD8). Thus, the multiclade env-gag VLP mRNA platform represents a promising approach for the development of an HIV-1 vaccine.
Collapse
Affiliation(s)
- Peng Zhang
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | | | - Qingbo Liu
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Yaroslav Tsybovsky
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Shilei Ding
- Université de Montreal, Montreal, Quebec, Canada
| | - Zonghui Hu
- Biostatistics Research Branch, NIAID, NIH, Bethesda, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, NIAID, NIH, Bethesda, MD, USA
| | - Yin Lin
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Huiyi Miao
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Hana Schmeisser
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Denise Rogers
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | | | | | | | | | | | - Xuejun Chen
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | | | | | - Rajeev Gautam
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Malcom A Martin
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Joanna Swerczek
- Experimental Primate Virology Section, NIAID, Poolesville, MD, USA
| | - Richard Herbert
- Experimental Primate Virology Section, NIAID, Poolesville, MD, USA
| | | | | | | | | | | | | | | | | | - Andrés Finzi
- Université de Montreal, Montreal, Quebec, Canada
| | | | - Anthony S Fauci
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Paolo Lusso
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA.
| |
Collapse
|
213
|
Andresen AMS, Gjøen T. Chitosan nanoparticle formulation attenuates poly (I:C) induced innate immune responses against inactivated virus vaccine in Atlantic salmon (Salmo salar). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2021; 40:100915. [PMID: 34634571 DOI: 10.1016/j.cbd.2021.100915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 06/13/2023]
Abstract
Many vaccine formulations, in particular vaccines based on inactivated virus, needs adjuvants to boost immunogenicity. In aquaculture, mineral and plant oil are used as adjuvant in commercial vaccines, and the advent of oil-adjuvanted vaccines was crucial to aquaculture development. Nevertheless, some of these approved vaccines display suboptimal performance in the field compared to experimental conditions. Therefore, there is a need to improve adjuvants and delivery methods for fish vaccines against viruses. We used RNA sequencing of Atlantic salmon head kidney to analyse the difference in gene expression 24 h after injection of different experimental vaccine formulations. We compared five different formulations in addition to a PBS control: inactivated virus alone (group V), soluble poly (I:C) (group P), nanoparticles containing poly (I:C) (group N), soluble poly (I:C) + inactivated virus (group PV) and finally nanoparticles containing poly (I:C) + inactivated virus (group NV). Our results showed poly (I:C)'s ability as adjuvant and its capacity influence innate immune genes expression in Atlantic salmon. Soluble poly (I:C) upregulated multiple immune related genes and was more effective compared to poly (I:C) formulated into chitosan nanoparticles (more than 10 fold increase in differentially expressed genes, DEGs). However, inclusion of inactivated ISA virus in the nanoparticle vaccine, increased the number of DEGs fivefold suggesting a synergistic effect of adjuvant and antigen. Our results indicate that the way poly (I:C) is formulated and the presence of antigen is important for the magnitude of the innate immune response in Atlantic salmon.
Collapse
Affiliation(s)
| | - Tor Gjøen
- Department of Pharmacy, Section for Pharmacology and Pharmaceutical Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
214
|
Rajput A, Pingale P, Telange D, Chalikwar S, Borse V. Lymphatic transport system to circumvent hepatic metabolism for oral delivery of lipid-based nanocarriers. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
215
|
Cai T, Liu H, Zhang S, Hu J, Zhang L. Delivery of nanovaccine towards lymphoid organs: recent strategies in enhancing cancer immunotherapy. J Nanobiotechnology 2021; 19:389. [PMID: 34823541 PMCID: PMC8620195 DOI: 10.1186/s12951-021-01146-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/14/2021] [Indexed: 01/15/2023] Open
Abstract
With the in-depth exploration on cancer therapeutic nanovaccines, increasing evidence shows that the poor delivery of nanovaccines to lymphoid organs has become the culprit limiting the rapid induction of anti-tumor immune response. Unlike the conventional prophylactic vaccines that mainly form a depot at the injection site to gradually trigger durable immune response, the rapid proliferation of tumors requires an efficient delivery of nanovaccines to lymphoid organs for rapid induction of anti-tumor immunity. Optimization of the physicochemical properties of nanovaccine (e.g., size, shape, charge, colloidal stability and surface ligands) is an effective strategy to enhance their accumulation in lymphoid organs, and nanovaccines with dynamic structures are also designed for precise targeted delivery of lymphoid organs or their subregions. The recent progress of these nanovaccine delivery strategies is highlighted in this review, and the challenges and future direction are also discussed. ![]()
Collapse
Affiliation(s)
- Ting Cai
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China
| | - Huina Liu
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China
| | - Shun Zhang
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China
| | - Jing Hu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China. .,Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 211200, China.
| | - Lingxiao Zhang
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China. .,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China. .,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China. .,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
216
|
Liu B, Yang Y, Chao Y, Xiao Z, Xu J, Wang C, Dong Z, Hou L, Li Q, Liu Z. Equipping Cancer Cell Membrane Vesicles with Functional DNA as a Targeted Vaccine for Cancer Immunotherapy. NANO LETTERS 2021; 21:9410-9418. [PMID: 34730968 DOI: 10.1021/acs.nanolett.1c02582] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
By inducing tumor-specific immune responses, tumor vaccines have recently aroused great research interest. Herein, we design a targeted nanovaccine by equipping cell membrane vesicles (CMVs) harvested from tumor cells with functional DNA including CpG oligonucleotide, an agonist for toll-like receptor 9, as well as an aptamer targeting the dendritic cell (DC)-specific intercellular adhesion molecule (ICAM)-3 grabbing nonintegrin (DC-SIGN) receptor overexpressed on DCs. Such DNA-modified CMVs could target DCs and further stimulate their maturation. Notably, our nanovaccines could trigger robust antitumor immune responses to effective delay the tumor growth. Moreover, the combination of CMV-based nanovaccines with an immune checkpoint blockade could result in improved therapeutic responses by eliminating the majority of the tumors as well as long-term immune memory to prevent tumor recurrence. Therefore, by simply assembling functional DNA on CMVs harvested from tumor cells, we propose a general platform of DC-targeted personalized cancer vaccines for effective and specific cancer immunotherapy.
Collapse
Affiliation(s)
- Bo Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Chao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhisheng Xiao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chunjie Wang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ziliang Dong
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Linqian Hou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qiaofeng Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau Special Administrative Region 999078, China
| |
Collapse
|
217
|
Kim J, Archer PA, Thomas SN. Innovations in lymph node targeting nanocarriers. Semin Immunol 2021; 56:101534. [PMID: 34836772 DOI: 10.1016/j.smim.2021.101534] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022]
Abstract
Lymph nodes are secondary lymphoid tissues in the body that facilitate the co-mingling of immune cells to enable and regulate the adaptive immune response. They are also tissues implicated in a variety of diseases, including but not limited to malignancy. The ability to access lymph nodes is thus attractive for a variety of therapeutic and diagnostic applications. As nanotechnologies are now well established for their potential in translational biomedical applications, their high relevance to applications that involve lymph nodes is highlighted. Herein, established paradigms of nanocarrier design to enable delivery to lymph nodes are discussed, considering the unique lymph node tissue structure as well as lymphatic system physiology. The influence of delivery mechanism on how nanocarrier systems distribute to different compartments and cells that reside within lymph nodes is also elaborated. Finally, current advanced nanoparticle technologies that have been developed to enable lymph node delivery are discussed.
Collapse
Affiliation(s)
- Jihoon Kim
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Emory University, 201 Dowman Drive, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, USA.
| |
Collapse
|
218
|
Meena J, Goswami DG, Anish C, Panda AK. Cellular uptake of polylactide particles induces size dependent cytoskeletal remodeling in antigen presenting cells. Biomater Sci 2021; 9:7962-7976. [PMID: 34704986 DOI: 10.1039/d1bm01312b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Phagocytosis of particulate vaccine delivery systems is a critical immune mechanism involved in antigen capture and processing by macrophages and dendritic cells. The internalization and degradation of the particles involve a complex sequence of events. This process coordinates lipids, signaling proteins, and the cytoskeleton. Dynamic changes in the actin cytoskeleton are essential for phagocytosis and antigen presentation. Knowledge regarding the correlation of surface properties, attached ligand density and geometric size of particles with the efficiency of phagocytosis may facilitate their design and application. To investigate this, polylactide biodegradable particles with different diameters (2-4 μm and 200-300 nm) were exposed to murine macrophages and dendritic cells and the effect of size on a series of cellular responses was evaluated. Cellular uptake studies using microscopy and flow cytometry showed size dependent internalization of particles, with nanoparticles accumulating in cells at a faster rate. The particles induced homoaggregation of cells and also showed cytoskeletal remodeling that could be inhibited by cytochalasin-D. Scanning electron microscopy images showed the time dependent formation of phagocytic cups and invaginations that promote particle uptake. The particles were observed to co-localized with the endo-lysosomal compartments after phagocyotosis. In our experiments, particle mediated immunoactivation, antigen processing and cytokine secretion have shown a good correlation with the uptake process. These findings would allow a better understanding of the process of particle uptake and may be instrumental in the rational design of optimal vaccine delivery systems.
Collapse
Affiliation(s)
- Jairam Meena
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi - 110067, India.
| | - Dinesh Giri Goswami
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi - 110067, India.
| | - Chakkumkal Anish
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi - 110067, India.
| | - Amulya Kumar Panda
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi - 110067, India.
| |
Collapse
|
219
|
Braz Gomes K, D’Sa S, Allotey-Babington GL, Kang SM, D’Souza MJ. Transdermal Vaccination with the Matrix-2 Protein Virus-like Particle (M2e VLP) Induces Immunity in Mice against Influenza A Virus. Vaccines (Basel) 2021; 9:1324. [PMID: 34835255 PMCID: PMC8619166 DOI: 10.3390/vaccines9111324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 01/01/2023] Open
Abstract
In this study, our goal was to utilize the extracellular domain matrix-2 protein virus-like particle (M2e VLP) that has been found to be highly conserved amongst all strains of influenza and could serve as a potential vaccine candidate against influenza. Previous studies have demonstrated that the VLP of the M2e showed increased activation of innate and adaptive immune responses. Therefore, to further explore its level of efficacy and protection, this vaccine was administered transdermally and tested in a pre-clinical mouse model. The M2e VLP was encapsulated into a polymeric matrix with the addition of Alhydrogel® and Monophosphoryl Lipid-A (MPL-A®), together referred to as AS04. The M2e VLP formulations induced IgG titers, with increased levels of IgG1 in the M2e VLP MP groups and further elevated levels of IgG2a were found specifically in the M2e VLP MP Adjuvant group. This trend in humoral immunity was also observed from a cell-mediated standpoint, where M2e VLP MP groups showed increased expression in CD4+ T cells in the spleen and the lymph node and high levels of CD8+ T cells in the lymph node. Taken together, the results illustrate the immunogenic potential of the matrix-2 protein virus-like particle (M2e VLP) vaccine.
Collapse
Affiliation(s)
- Kimberly Braz Gomes
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (G.L.A.-B.); (M.J.D.)
| | - Sucheta D’Sa
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (G.L.A.-B.); (M.J.D.)
| | - Grace Lovia Allotey-Babington
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (G.L.A.-B.); (M.J.D.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA;
| | - Martin J. D’Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (G.L.A.-B.); (M.J.D.)
| |
Collapse
|
220
|
Lu Y, Shi Y, You J. Strategy and clinical application of up-regulating cross presentation by DCs in anti-tumor therapy. J Control Release 2021; 341:184-205. [PMID: 34774890 DOI: 10.1016/j.jconrel.2021.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/20/2022]
Abstract
The cross presentation of exogenous antigen (Ag) by dendritic cells (DCs) facilitates a diversified mode of T-cell activation, orchestrates specific humoral and cellular immunity, and contributes to an efficient anti-tumor immune response. DCs-mediated cross presentation is subject to both intrinsic and extrinsic factors, including the homing and phenotype of DCs, the spatiotemporal trafficking and degradation kinetics of Ag, and multiple microenvironmental clues, with many details largely unexplored. Here, we systemically review the current mechanistic understanding and regulation strategies of cross presentation by heterogeneous DC populations. We also provide insights into the future exploitation of DCs cross presentation for a better clinical efficacy in anti-tumor therapy.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
221
|
Dangkoub F, Sankian M, Tafaghodi M, Jaafari MR, Badiee A. The impact of nanocarriers in the induction of antigen-specific immunotolerance in autoimmune diseases. J Control Release 2021; 339:274-283. [PMID: 34600024 DOI: 10.1016/j.jconrel.2021.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/22/2022]
Abstract
Immunotolerance induction in an antigen-specific manner is the long-term goal of immunotherapy to treat autoimmune diseases. Nanocarriers (NCs) can be designed as a new generation of delivery systems to modulate the immune responses through targeted delivery of antigens and immunomodulators to antigen presenting cells (APCs). In this manuscript, several formulation factors in the preparation of NCs which affect their uptake using APCs and generation of tolerance have been reviewed. The physicochemical properties and composition of NCs have been shown to play essential roles in achieving the desired immunological outcome. Also, targeting of dendritic cells and macrophages as APCs and direct targeting of the autoreactive lymphocytes have been presented as two main ways for induction of antigen-specific tolerance by these tolerogenic nanocarriers (tNCs). These particles herald a promising approach to treat or even prevent unwanted immune reactions in humans specifically.
Collapse
Affiliation(s)
- Faezeh Dangkoub
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
222
|
Pérez-Betancourt Y, Araujo PM, Távora BDCLF, Pereira DR, Faquim-Mauro EL, Carmona-Ribeiro AM. Cationic and Biocompatible Polymer/Lipid Nanoparticles as Immunoadjuvants. Pharmaceutics 2021; 13:pharmaceutics13111859. [PMID: 34834275 PMCID: PMC8621050 DOI: 10.3390/pharmaceutics13111859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/01/2022] Open
Abstract
Nanostructures have been of paramount importance for developing immunoadjuvants. They must be cationic and non-cytotoxic, easily assembling with usually oppositely charged antigens such as proteins, haptens or nucleic acids for use in vaccines. We obtained optimal hybrid nanoparticles (NPs) from the biocompatible polymer poly(methyl methacrylate) (PMMA) and the cationic lipid dioctadecyl dimethyl ammonium bromide (DODAB) by emulsion polymerization of methyl methacrylate (MMA) in the presence of DODAB. NPs adsorbed ovalbumin (OVA) as a model antigen and we determined their adjuvant properties. Interestingly, they elicited high double immune responses of the cellular and humoral types overcoming the poor biocompatibility of DODAB-based adjuvants of the bilayer type. The results suggested that the novel adjuvant would be possibly of use in a variety of vaccines.
Collapse
Affiliation(s)
- Yunys Pérez-Betancourt
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenida Professor Lineu Prestes, 748 Butantan, São Paulo 05508-000, Brazil; (Y.P.-B.); (P.M.A.)
| | - Péricles Marques Araujo
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenida Professor Lineu Prestes, 748 Butantan, São Paulo 05508-000, Brazil; (Y.P.-B.); (P.M.A.)
| | - Bianca de Carvalho Lins Fernandes Távora
- Immunopathology Laboratory, Butantan Institute, Avenida Vital Brasil, 1500 Butantan, São Paulo 05503-900, Brazil; (B.d.C.L.F.T.); (D.R.P.); (E.L.F.-M.)
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Avenida Professor Lineu Prestes, 1730 Butantan, São Paulo 05508-000, Brazil
| | - Daniele Rodrigues Pereira
- Immunopathology Laboratory, Butantan Institute, Avenida Vital Brasil, 1500 Butantan, São Paulo 05503-900, Brazil; (B.d.C.L.F.T.); (D.R.P.); (E.L.F.-M.)
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Avenida Professor Lineu Prestes, 1730 Butantan, São Paulo 05508-000, Brazil
| | - Eliana Lima Faquim-Mauro
- Immunopathology Laboratory, Butantan Institute, Avenida Vital Brasil, 1500 Butantan, São Paulo 05503-900, Brazil; (B.d.C.L.F.T.); (D.R.P.); (E.L.F.-M.)
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Avenida Professor Lineu Prestes, 1730 Butantan, São Paulo 05508-000, Brazil
| | - Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenida Professor Lineu Prestes, 748 Butantan, São Paulo 05508-000, Brazil; (Y.P.-B.); (P.M.A.)
- Correspondence:
| |
Collapse
|
223
|
Mayorga C, Perez‐Inestrosa E, Rojo J, Ferrer M, Montañez MI. Role of nanostructures in allergy: Diagnostics, treatments and safety. Allergy 2021; 76:3292-3306. [PMID: 33559903 DOI: 10.1111/all.14764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 01/08/2023]
Abstract
Nanotechnology is science, engineering and technology conducted at the nanoscale, which is about 1-100 nm. It has led to the development of nanomaterials, which behave very differently from materials with larger scales and can have a wide range of applications in biomedicine. The physical and chemical properties of materials of such small compounds depend mainly on the size, shape, composition and functionalization of the system. Nanoparticles, carbon nanotubes, liposomes, polymers, dendrimers and nanogels, among others, can be nanoengineeried for controlling all parameters, including their functionalization with ligands, which provide the desired interaction with the immunological system, that is dendritic cell receptors to activate and/or modulate the response, as well as specific IgE, or effector cell receptors. However, undesired issues related to toxicity and hypersensitivity responses can also happen and would need evaluation. There are wide panels of accessible structures, and controlling their physico-chemical properties would permit obtaining safer and more efficient compounds for clinical applications goals, either in diagnosis or treatment. The application of dendrimeric antigens, nanoallergens and nanoparticles in allergy diagnosis is very promising since it can improve sensitivity by increasing specific IgE binding, mimicking carrier proteins or enhancing signal detection. Additionally, in the case of immunotherapy, glycodendrimers, liposomes, polymers and nanoparticles have shown interest, behaving as platforms of allergenic structures, adjuvants or protectors of allergen from degradation or having a depot capacity. Taken together, the application of nanotechnology to allergy shows promising facts facing important goals related to the improvement of diagnosis as well as specific immunotherapy.
Collapse
Affiliation(s)
- Cristobalina Mayorga
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA Málaga Spain
- Allergy Unit Hospital Regional Universitario de Málaga Málaga Spain
- Andalusian Centre for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
| | - Ezequiel Perez‐Inestrosa
- Andalusian Centre for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
- Departamento de Química Orgánica, and the Biomimetic Dendrimers and Photonic Laboratory Instituto de Investigación Biomédica de Málaga‐IBIMAUniversidad de Málaga Málaga Spain
| | - Javier Rojo
- Glycosystems Laboratory Instituto de Investigaciones Químicas (IIQ)CSIC—Universidad de Sevilla Sevilla Spain
| | - Marta Ferrer
- Department of Allergy and Clinical Immunology Clínica Universidad de NavarraInstituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona Spain
| | - Maria Isabel Montañez
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA Málaga Spain
- Andalusian Centre for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
| |
Collapse
|
224
|
Zhu M. Immunological perspectives on spatial and temporal vaccine delivery. Adv Drug Deliv Rev 2021; 178:113966. [PMID: 34506868 DOI: 10.1016/j.addr.2021.113966] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/22/2021] [Accepted: 09/05/2021] [Indexed: 12/19/2022]
Abstract
The so-called rational design of vaccines has been a very attractive concept and also an important direction for vaccine research and development. However, the underlying rationales, especially on the immunological aspect, remain less systemically and deeply understood. Given the critical role of lymph nodes (LNs) in the induction of B and T cell responses upon vaccination, LN targeting has been a popular strategy in vaccine design. The LN is a highly organized structure; induction of adaptive immune response is highly orchestrated by various types of LN stromal cells and hematopoietic immune cells both spatially and temporally. Thus, not only LN targeting, but also cellular targeting and even subcellular compartment targeting should be considered for specifically enhanced vaccine efficacy. Moreover, temporal control of vaccine antigen and adjuvant delivery may also optimize the immune response.
Collapse
|
225
|
Garofalo C, De Marco C, Cristiani CM. NK Cells in the Tumor Microenvironment as New Potential Players Mediating Chemotherapy Effects in Metastatic Melanoma. Front Oncol 2021; 11:754541. [PMID: 34712615 PMCID: PMC8547654 DOI: 10.3389/fonc.2021.754541] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Until the last decade, chemotherapy was the standard treatment for metastatic cutaneous melanoma, even with poor results. The introduction of immune checkpoints inhibitors (ICIs) radically changed the outcome, increasing 5-year survival from 5% to 60%. However, there is still a large portion of unresponsive patients that would need further therapies. NK cells are skin-resident innate cytotoxic lymphocytes that recognize and kill virus-infected as well as cancer cells thanks to a balance between inhibitory and activating signals delivered by surface molecules expressed by the target. Since NK cells are equipped with cytotoxic machinery but lack of antigen restriction and needing to be primed, they are nowadays gaining attention as an alternative to T cells to be exploited in immunotherapy. However, their usage suffers of the same limitations reported for T cells, that is the loss of immunogenicity by target cells and the difficulty to penetrate and be activated in the suppressive tumor microenvironment (TME). Several evidence showed that chemotherapy used in metastatic melanoma therapy possess immunomodulatory properties that may restore NK cells functions within TME. Here, we will discuss the capability of such chemotherapeutics to: i) up-regulate melanoma cells susceptibility to NK cell-mediated killing, ii) promote NK cells infiltration within TME, iii) target other immune cell subsets that affect NK cells activities. Alongside traditional systemic melanoma chemotherapy, a new pharmacological strategy based on nanocarriers loaded with chemotherapeutics is developing. The use of nanotechnologies represents a very promising approach to improve drug tolerability and effectiveness thanks to the targeted delivery of the therapeutic molecules. Here, we will also discuss the recent developments in using nanocarriers to deliver anti-cancer drugs within the melanoma microenvironment in order to improve chemotherapeutics effects. Overall, we highlight the possibility to use standard chemotherapeutics, possibly delivered by nanosystems, to enhance NK cells anti-tumor cytotoxicity. Combined with immunotherapies targeting NK cells, this may represent a valuable alternative approach to treat those patients that do not respond to current ICIs.
Collapse
Affiliation(s)
- Cinzia Garofalo
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Carmela De Marco
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Costanza Maria Cristiani
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
226
|
Liu Z, Xu N, Zhao L, Yu J, Zhang P. Bifunctional lipids in tumor vaccines: An outstanding delivery carrier and promising immune stimulator. Int J Pharm 2021; 608:121078. [PMID: 34500059 DOI: 10.1016/j.ijpharm.2021.121078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/28/2021] [Accepted: 09/02/2021] [Indexed: 12/18/2022]
Abstract
Cancer is still a major threat for human life, and the cancer immunotherapy can be more optimized to prolong life. However, the effect of immunotherapy is not encouraging. In order to achieve outstanding immune effect, it is necessary to strengthen antigens uptake of antigen presenting cells. Adjuvants were added to vaccines to achieve this purpose, which could be divided into two types: as an immunostimulatory molecule, the innate immunities of the body were triggered; or as a delivery carrier, and antigens were cross-delivery through the "cytoplasmic pathway" and released at a specific location. This paper reviewed the relevant research status of tumor vaccine immune adjuvants in recent years. Among the review, the function, combination strategies and derivatives of lipid A were discussed in detail. In addition, some suggestions on the existing problems and research direction of lipids as tumor vaccine adjuvants were put forward.
Collapse
Affiliation(s)
- Zhiling Liu
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Na Xu
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Lin Zhao
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jia Yu
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
227
|
He Y, de Araújo Júnior RF, Cruz LJ, Eich C. Functionalized Nanoparticles Targeting Tumor-Associated Macrophages as Cancer Therapy. Pharmaceutics 2021; 13:1670. [PMID: 34683963 PMCID: PMC8540805 DOI: 10.3390/pharmaceutics13101670] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays a central role in regulating antitumor immune responses. As an important part of the TME, alternatively activated type 2 (M2) macrophages drive the development of primary and secondary tumors by promoting tumor cell proliferation, tumor angiogenesis, extracellular matrix remodeling and overall immunosuppression. Immunotherapy approaches targeting tumor-associated macrophages (TAMs) in order to reduce the immunosuppressive state in the TME have received great attention. Although these methods hold great potential for the treatment of several cancers, they also face some limitations, such as the fast degradation rate of drugs and drug-induced cytotoxicity of organs and tissues. Nanomedicine formulations that prevent TAM signaling and recruitment to the TME or deplete M2 TAMs to reduce tumor growth and metastasis represent encouraging novel strategies in cancer therapy. They allow the specific delivery of antitumor drugs to the tumor area, thereby reducing side effects associated with systemic application. In this review, we give an overview of TAM biology and the current state of nanomedicines that target M2 macrophages in the course of cancer immunotherapy, with a specific focus on nanoparticles (NPs). We summarize how different types of NPs target M2 TAMs, and how the physicochemical properties of NPs (size, shape, charge and targeting ligands) influence NP uptake by TAMs in vitro and in vivo in the TME. Furthermore, we provide a comparative analysis of passive and active NP-based TAM-targeting strategies and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Yuanyuan He
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| | - Raimundo Fernandes de Araújo Júnior
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal 59064-720, Brazil
- Cancer and Inflammation Research Laboratory (LAICI), Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59064-720, Brazil
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| | - Christina Eich
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| |
Collapse
|
228
|
Abstract
INTRODUCTION Vaccination is so far the most effective way of eradicating infections. Rapidly emerging drug resistance against infectious diseases and chemotherapy-related toxicities in cancer warrant immediate vaccine development to save mankind. Subunit vaccines alone, however, fail to elicit sufficiently strong and long-lasting protective immunity against deadly pathogens. Nanoparticle (NP)-based delivery vehicles like microemulsions, liposomes, virosomes, nanogels, micelles and dendrimers offer promising strategies to overcome limitations of traditional vaccine adjuvants. Nanovaccines can improve targeted delivery, antigen presentation, stimulation of body's innate immunity, strong T cell response combined with safety to combat infectious diseases and cancers. Further, nanovaccines can be highly beneficial to generate effective immutherapeutic formulations against cancer. AREAS COVERED This review summarizes the emerging nanoparticle strategies highlighting their success and challenges in preclinical and clinical trials in infectious diseases and cancer. It provides a concise overview of current nanoparticle-based vaccines, their adjuvant potential and their cellular delivery mechanisms. EXPERT OPINION The nanovaccines (50-250 nm in size) are most efficient in terms of tissue targeting, prolonged circulation and preferential uptake by the professional APCs chiefly due to their small size. More rational designing, improved antigen loading, extensive functionalization and targeted delivery are some of the future goals of ideal nanovaccines.
Collapse
Affiliation(s)
- Amrita Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
229
|
Aktar N, Chen T, Moudud A, Xu S, Zhou X. Tolerogenic vehicles of antigens in the antigen-specific immunotherapy for autoimmunity. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
230
|
Ye T, Li F, Ma G, Wei W. Enhancing therapeutic performance of personalized cancer vaccine via delivery vectors. Adv Drug Deliv Rev 2021; 177:113927. [PMID: 34403752 DOI: 10.1016/j.addr.2021.113927] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/29/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022]
Abstract
In recent years, personalized cancer vaccines have gained increasing attention as emerging immunotherapies with the capability to overcome interindividual differences and show great benefits for individual patients in the clinic due to the highly tailored vaccine formulations. A large number of materials have been studied as delivery vectors to enhance the therapeutic performance of personalized cancer vaccines, including artificial materials, engineered microorganisms, cells and cell derivatives. These delivery vectors with distinct features are employed to change antigen biodistributions and to facilitate antigen uptake, processing and presentation, improving the strength, velocity, and duration of the immune response when delivered by different strategies. Here, we provide an overview of personalized cancer vaccine delivery vectors, describing their materials, physicochemical properties, delivery strategies and challenges for clinical transformation.
Collapse
|
231
|
O'Neill CL, Shrimali PC, Clapacs ZE, Files MA, Rudra JS. Peptide-based supramolecular vaccine systems. Acta Biomater 2021; 133:153-167. [PMID: 34010691 PMCID: PMC8497425 DOI: 10.1016/j.actbio.2021.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022]
Abstract
Currently approved replication-competent and inactivated vaccines are limited by excessive reactogenicity and poor safety profiles, while subunit vaccines are often insufficiently immunogenic without co-administering exogenous adjuvants. Self-assembling peptide-, peptidomimetic-, and protein-based biomaterials offer a means to overcome these challenges through their inherent modularity, multivalency, and biocompatibility. As these scaffolds are biologically derived and present antigenic arrays reminiscent of natural viruses, they are prone to immune recognition and are uniquely capable of functioning as self-adjuvanting vaccine delivery vehicles that improve humoral and cellular responses. Beyond this intrinsic immunological advantage, the wide range of available amino acids allows for facile de novo design or straightforward modifications to existing sequences. This has permitted the development of vaccines and immunotherapies tailored to specific disease models, as well as generalizable platforms that have been successfully applied to prevent or treat numerous infectious and non-infectious diseases. In this review, we briefly introduce the immune system, discuss the structural determinants of coiled coils, β-sheets, peptide amphiphiles, and protein subunit nanoparticles, and highlight the utility of these materials using notable examples of their innate and adaptive immunomodulatory capacity. STATEMENT OF SIGNIFICANCE: Subunit vaccines have recently gained considerable attention due to their favorable safety profiles relative to traditional whole-cell vaccines; however, their reduced efficacy requires co-administration of reactogenic adjuvants to boost immune responses. This has led to collaborative efforts between engineers and immunologists to develop nanomaterial-based vaccination platforms that can elicit protection without deleterious side effects. Self-assembling peptidic biomaterials are a particularly attractive approach to this problem, as their structure and function can be controlled through primary sequence design and their capacity for multivalent presentation of antigens grants them intrinsic self-adjuvanticity. This review introduces the various architectures adopted by self-assembling peptides and discusses their application as modulators of innate and adaptive immunity.
Collapse
Affiliation(s)
- Conor L O'Neill
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Paresh C Shrimali
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Zoe E Clapacs
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Megan A Files
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States.
| | - Jai S Rudra
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| |
Collapse
|
232
|
Gracia, Cao E, Kochappan R, Jh Porter C, Pr Johnston A, Trevaskis NL. Association of a vaccine adjuvant with endogenous HDL increases lymph uptake and dendritic cell activation. Eur J Pharm Biopharm 2021; 172:240-252. [PMID: 34571191 DOI: 10.1016/j.ejpb.2021.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/02/2021] [Accepted: 09/11/2021] [Indexed: 11/28/2022]
Abstract
Vaccines are a powerful health intervention but there is still an unmet need for effective preventative and therapeutic vaccines for many diseases such as cancer and infections. Interstitial (e.g. subcutaneous (SC)) injection in nano-sized carriers such as high density lipoproteins (HDLs) can improve the access of vaccine subunit antigens or adjuvants to target immune cells in the lymphatics and potentiate vaccination responses such as cytotoxic T lymphocyte (CTL) responses[1-3]. Here we examined how cholesterol conjugation to the vaccine adjuvant CpG, and incorporation into HDL, changes lymphatic absorption and association with, and processing by, dendritic cells (DCs), ultimately influencing adjuvant efficacy. We investigated the lymphatic disposition of cholesterol conjugated CpG incorporated into HDL (HDL(Chol-CpG-Cy5)) relative to cholesterol conjugated (Chol-CpG-Cy5) and unconjugated CpG (free CpG-Cy5) alone after SC administration into rats and mice. HDL (Chol-CpG-Cy5) and Chol-CpG-Cy5 differentially altered CpG absorption into lymph vs. blood, but surprisingly resulted in similarly higher LN accumulation relative to free CpG. The mechanism of access of Chol-CpG-Cy5 into lymph might be partly due to association with endogenous HDL at the injection site followed by transport into lymph in association with the HDL. To measure CpG association with and processing by DCs and the strength of the immune response, mice were vaccinated with free ovalbumin (OVA) co-administered with the different CpG constructs. There were significant changes in DC activation that were reflective of the trend in LN accumulation at 24h post-vaccination. However, T cell response at 24h and 7 days post-vaccination were not significantly different across the CpG groups although the response was less variable for Chol-CpG-Cy5 compared to free CpG Cy5 and also HDL(Chol-CpG-Cy5) - despite similar LN accumulation with the latter. Overall, our data indicate that cholesterol conjugation and incorporation into HDL increases adjuvant lymph disposition and DC activation.
Collapse
Affiliation(s)
- Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Ruby Kochappan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Christopher Jh Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Angus Pr Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia.
| |
Collapse
|
233
|
Lv S, Sylvestre M, Prossnitz AN, Yang LF, Pun SH. Design of Polymeric Carriers for Intracellular Peptide Delivery in Oncology Applications. Chem Rev 2021; 121:11653-11698. [PMID: 33566580 DOI: 10.1021/acs.chemrev.0c00963] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent decades, peptides, which can possess high potency, excellent selectivity, and low toxicity, have emerged as promising therapeutics for cancer applications. Combined with an improved understanding of tumor biology and immuno-oncology, peptides have demonstrated robust antitumor efficacy in preclinical tumor models. However, the translation of peptides with intracellular targets into clinical therapies has been severely hindered by limitations in their intrinsic structure, such as low systemic stability, rapid clearance, and poor membrane permeability, that impede intracellular delivery. In this Review, we summarize recent advances in polymer-mediated intracellular delivery of peptides for cancer therapy, including both therapeutic peptides and peptide antigens. We highlight strategies to engineer polymeric materials to increase peptide delivery efficiency, especially cytosolic delivery, which plays a crucial role in potentiating peptide-based therapies. Finally, we discuss future opportunities for peptides in cancer treatment, with an emphasis on the design of polymer nanocarriers for optimized peptide delivery.
Collapse
Affiliation(s)
| | | | - Alexander N Prossnitz
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | | | | |
Collapse
|
234
|
Lucas ED, Schafer JB, Matsuda J, Kraus M, Burchill MA, Tamburini BAJ. PD-L1 Reverse Signaling in Dermal Dendritic Cells Promotes Dendritic Cell Migration Required for Skin Immunity. Cell Rep 2021; 33:108258. [PMID: 33053342 PMCID: PMC7688291 DOI: 10.1016/j.celrep.2020.108258] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/06/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Although the function of the extracellular region of programmed death ligand 1 (PD-L1) through its interactions with PD-1 on T cells is well studied, little is understood regarding the intracellular domain of PD-L1. Here, we outline a major role for PD-L1 intracellular signaling in the control of dendritic cell (DC) migration from the skin to the draining lymph node (dLN). Using a mutant mouse model, we identify a TSS signaling motif within the intracellular domain of PD-L1. The TSS motif proves critical for chemokine-mediated DC migration to the dLN during inflammation. This loss of DC migration, in the PD-L1 TSS mutant, leads to a significant decline in T cell priming when DC trafficking is required for antigen delivery to the dLN. Finally, the TSS motif is required for chemokine receptor signaling downstream of the Gα subunit of the heterotrimeric G protein complex, ERK phosphorylation, and actin polymerization in DCs. Lucas et al. define three residues within the cytoplasmic tail of PD-L1 that are required for proper dendritic cell migration from the skin to the lymph node. These three-amino-acid residues promote chemokine signaling in dendritic cells and productive T cell responses to skin infections.
Collapse
Affiliation(s)
- Erin D Lucas
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | | | - Madison Kraus
- Gates Summer Research Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Matthew A Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA.
| |
Collapse
|
235
|
Wilks LR, Joshi G, Grisham MR, Gill HS. Tyrosine-Based Cross-Linking of Peptide Antigens to Generate Nanoclusters with Enhanced Immunogenicity: Demonstration Using the Conserved M2e Peptide of Influenza A. ACS Infect Dis 2021; 7:2723-2735. [PMID: 34432416 DOI: 10.1021/acsinfecdis.1c00219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A method of creating nanoclusters (NCs) from soluble peptide molecules is described utilizing an approach based on a tyrosine-tyrosine cross-linking reaction. A reactive tag comprising histidine and tyrosine residues was introduced at the termini of the peptide molecules. The cross-linking reaction led to the creation of dityrosine bonds within the tag, which allowed for the generation of peptide NCs. We show that it is essential for the reactive tag to be present at both the "N" and "C" termini of the peptide for cluster formation to occur. Additionally, the cross-linking reaction was systematically characterized to show the importance of reaction conditions on final cluster diameter, allowing us to generate NCs of various sizes. To demonstrate the immunogenic potential of the peptide clusters, we chose to study the conserved influenza peptide, M2e, as the antigen. M2e NCs were formulated using the cross-linking reaction. We show the ability of the clusters to generate protective immunity in a dose, size, and frequency dependent manner against a lethal influenza A challenge in BALB/c mice. Taken together, the data presented suggest this new cluster formation technique can generate highly immunogenic peptide NCs in a simple and controllable manner.
Collapse
Affiliation(s)
- Logan R. Wilks
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Megan R. Grisham
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
236
|
Sun Z, Qiao D, Shi Y, Barz M, Liu L, Chen Y. Precision Wormlike Nanoadjuvant Governs Potency of Vaccination. NANO LETTERS 2021; 21:7236-7243. [PMID: 34459617 DOI: 10.1021/acs.nanolett.1c02274] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
It remains unclear how the precise length of one-dimensional nanovehicles influences the characters of vaccination. Here, a unimolecular nanovehicle with tailored size and aspect ratio (AR) is applied to deliver CpG oligodeoxynucleotide, a Toll-like receptor (TLR) 9 agonist, as an adjuvant of recombinant hepatitis B virus surface antigen (rHBsAg), for treating chronic hepatitis B (CHB). Cationic nanovehicles with fixed width (ca. 45 nm) but varied length (46 nm-180 nm), AR from 1 to 4, are prepared through controlled polymerization and are loaded with CpG by electrostatic interaction. We reveal that the nanoadjuvant with AR = 2 shows the highest retention in proximal lymph nodes. Importantly, it is more easily internalized into antigen-presenting cells and accumulates in the late endosome, where TLR9 is located. Such a nanoadjuvant exhibits the strongest immune response with rHBsAg to clear the hepatitis B virus in the CHB mouse model, showing that the AR of nanovehicles governs the efficiency of vaccination.
Collapse
Affiliation(s)
- Ziyang Sun
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Dongdong Qiao
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Yi Shi
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Matthias Barz
- Leiden Academic Center for Drug Research, Division of Biotherapeutics, Laboratory for Biotherapeutic Delivery, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Department Chemie, Johannes Gutenberg University, Duesbergweg 10-14, 55099 Mainz, Germany
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
237
|
Virus-Like Particle Vaccines Against Respiratory Viruses and Protozoan Parasites. Curr Top Microbiol Immunol 2021; 433:77-106. [PMID: 33650036 DOI: 10.1007/82_2021_232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The field of vaccinology underwent massive advances over the past decades with the introduction of virus-like particles (VLPs), a supra-molecular nanoparticle vaccine platform that resembles viral structures without the ability to replicate in hosts. This innovative approach has been remarkably effective, as evidenced by its profound immunogenicity and safety. These highly desirable intrinsic properties enabled their further development as vaccines against a multitude of diseases. To date, several VLP-based vaccines have already been commercialized and many more are undergoing clinical evaluation prior to FDA approval. However, efficacious vaccines against a plethora of pathogens are still lacking, which imposes a tremendous socioeconomic burden and continues to threaten public health throughout the globe. This is especially the case for several respiratory pathogens and protozoan parasites. In this review, we briefly describe the fundamentals of VLP vaccines and the unique properties that enable these to be such valuable vaccine candidates and summarize current advances in VLP-based vaccines targeting respiratory and parasitic diseases of global importance.
Collapse
|
238
|
Pilkington EH, Suys EJA, Trevaskis NL, Wheatley AK, Zukancic D, Algarni A, Al-Wassiti H, Davis TP, Pouton CW, Kent SJ, Truong NP. From influenza to COVID-19: Lipid nanoparticle mRNA vaccines at the frontiers of infectious diseases. Acta Biomater 2021; 131:16-40. [PMID: 34153512 PMCID: PMC8272596 DOI: 10.1016/j.actbio.2021.06.023] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023]
Abstract
Vaccination represents the best line of defense against infectious diseases and is crucial in curtailing pandemic spread of emerging pathogens to which a population has limited immunity. In recent years, mRNA vaccines have been proposed as the new frontier in vaccination, owing to their facile and rapid development while providing a safer alternative to traditional vaccine technologies such as live or attenuated viruses. Recent breakthroughs in mRNA vaccination have been through formulation with lipid nanoparticles (LNPs), which provide both protection and enhanced delivery of mRNA vaccines in vivo. In this review, current paradigms and state-of-the-art in mRNA-LNP vaccine development are explored through first highlighting advantages posed by mRNA vaccines, establishing LNPs as a biocompatible delivery system, and finally exploring the use of mRNA-LNP vaccines in vivo against infectious disease towards translation to the clinic. Furthermore, we highlight the progress of mRNA-LNP vaccine candidates against COVID-19 currently in clinical trials, with the current status and approval timelines, before discussing their future outlook and challenges that need to be overcome towards establishing mRNA-LNPs as next-generation vaccines. STATEMENT OF SIGNIFICANCE: With the recent success of mRNA vaccines developed by Moderna and BioNTech/Pfizer against COVID-19, mRNA technology and lipid nanoparticles (LNP) have never received more attention. This manuscript timely reviews the most advanced mRNA-LNP vaccines that have just been approved for emergency use and are in clinical trials, with a focus on the remarkable development of several COVID-19 vaccines, faster than any other vaccine in history. We aim to give a comprehensive introduction of mRNA and LNP technology to the field of biomaterials science and increase accessibility to readers with a new interest in mRNA-LNP vaccines. We also highlight current limitations and future outlook of the mRNA vaccine technology that need further efforts of biomaterials scientists to address.
Collapse
Affiliation(s)
- Emily H Pilkington
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Estelle J A Suys
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Danijela Zukancic
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Azizah Algarni
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Hareth Al-Wassiti
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Colin W Pouton
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Nghia P Truong
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia.
| |
Collapse
|
239
|
Liu G, Zhu M, Zhao X, Nie G. Nanotechnology-empowered vaccine delivery for enhancing CD8 + T cells-mediated cellular immunity. Adv Drug Deliv Rev 2021; 176:113889. [PMID: 34364931 DOI: 10.1016/j.addr.2021.113889] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/17/2021] [Accepted: 07/18/2021] [Indexed: 12/18/2022]
Abstract
After centuries of development, using vaccination to stimulate immunity has become an effective method for prevention and treatment of a variety of diseases including infective diseases and cancers. However, the tailor-made efficient delivery system for specific antigens is still urgently needed due to the low immunogenicity and stability of antigens, especially for vaccines to induce CD8+ T cells-mediated cellular immunity. Unlike B cells-mediated humoral immunity, CD8+ T cells-mediated cellular immunity mainly aims at the intracellular antigens from microorganism in virus-infected cells or genetic mutations in tumor cells. Therefore, the vaccines for stimulating CD8+ T cells-mediated cellular immunity should deliver the antigens efficiently into the cytoplasm of antigen presenting cells (APCs) to form major histocompatibility complex I (MHCI)-antigen complex through cross-presentation, followed by activating CD8+ T cells for immune protection and clearance. Importantly, nanotechnology has been emerged as a powerful tool to facilitate these multiple processes specifically, allowing not only enhanced antigen immunogenicity and stability but also APCs-targeted delivery and elevated cross-presentation. This review summarizes the process of CD8+ T cells-mediated cellular immunity induced by vaccines and the technical advantages of nanotechnology implementation in general, then provides an overview of the whole spectrum of nanocarriers studied so far and the recent development of delivery nanotechnology in vaccines against infectious diseases and cancer. Finally, we look forward to the future development of nanotechnology for the next generation of vaccines to induce CD8+ T cells-mediated cellular immunity.
Collapse
Affiliation(s)
- Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; The GBA National Institute for Nanotechnology Innovation, Guangdong 510700, China.
| |
Collapse
|
240
|
Shute T, Amiel E, Alam N, Yates JL, Mohrs K, Dudley E, Salas B, Mesa C, Serrata A, Angel D, Vincent BK, Weyers A, Lanthier PA, Vomhof-Dekrey E, Fromme R, Laughlin M, Durham O, Miao J, Shipp D, Linhardt RJ, Nash K, Leadbetter EA. Glycolipid-Containing Nanoparticle Vaccine Engages Invariant NKT Cells to Enhance Humoral Protection against Systemic Bacterial Infection but Abrogates T-Independent Vaccine Responses. THE JOURNAL OF IMMUNOLOGY 2021; 206:1806-1816. [PMID: 33811104 DOI: 10.4049/jimmunol.2001283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
CD4+ T cells enable the critical B cell humoral immune protection afforded by most effective vaccines. We and others have recently identified an alternative source of help for B cells in mice, invariant NK T (iNKT) cells. iNKT cells are innate glycolipid-specific T cells restricted to the nonpolymorphic Ag-presenting molecule CD1d. As such, iNKT cells respond to glycolipids equally well in all people, making them an appealing adjuvant for universal vaccines. We tested the potential for the iNKT glycolipid agonist, α-galactosylceramide (αGC), to serve as an adjuvant for a known human protective epitope by creating a nanoparticle that delivers αGC plus antigenic polysaccharides from Streptococcus pneumoniae αGC-embedded nanoparticles activate murine iNKT cells and B cells in vitro and in vivo, facilitate significant dose sparing, and avoid iNKT anergy. Nanoparticles containing αGC plus S. pneumoniae polysaccharides elicits robust IgM and IgG in vivo and protect mice against lethal systemic S. pneumoniae However, codelivery of αGC via nanoparticles actually eliminated Ab protection elicited by a T-independent S. pneumoniae vaccine. This is consistent with previous studies demonstrating iNKT cell help for B cells following acute activation, but negative regulation of B cells during chronic inflammation. αGC-containing nanoparticles represent a viable platform for broadly efficacious vaccines against deadly human pathogens, but their potential for eliminating B cells under certain conditions suggests further clarity on iNKT cell interactions with B cells is warranted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Daniel Angel
- Department of Astronomy and Physics, The University of Texas at San Antonio, San Antonio, TX
| | - Brandy K Vincent
- Department of Astronomy and Physics, The University of Texas at San Antonio, San Antonio, TX
| | | | | | | | - Rachel Fromme
- Center for Advanced Material Processing, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699
| | - Mitchell Laughlin
- Center for Advanced Material Processing, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699
| | - Olivia Durham
- Center for Advanced Material Processing, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699
| | | | - Devon Shipp
- Center for Advanced Material Processing, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699
| | | | - Kelly Nash
- Department of Astronomy and Physics, The University of Texas at San Antonio, San Antonio, TX
| | | |
Collapse
|
241
|
Mancini F, Micoli F, Necchi F, Pizza M, Berlanda Scorza F, Rossi O. GMMA-Based Vaccines: The Known and The Unknown. Front Immunol 2021; 12:715393. [PMID: 34413858 PMCID: PMC8368434 DOI: 10.3389/fimmu.2021.715393] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/19/2021] [Indexed: 11/20/2022] Open
Abstract
Generalized Modules for Membrane Antigens (GMMA) are outer membrane vesicles derived from Gram-negative bacteria engineered to provide an over-vesiculating phenotype, which represent an attractive platform for the design of affordable vaccines. GMMA can be further genetically manipulated to modulate the risk of systemic reactogenicity and to act as delivery system for heterologous polysaccharide or protein antigens. GMMA are able to induce strong immunogenicity and protection in animal challenge models, and to be well-tolerated and immunogenic in clinical studies. The high immunogenicity could be ascribed to their particulate size, to their ability to present to the immune system multiple antigens in a natural conformation which mimics the bacterial environment, as well as to their intrinsic self-adjuvanticity. However, GMMA mechanism of action and the role in adjuvanticity are still unclear and need further investigation. In this review, we discuss progresses in the development of the GMMA vaccine platform, highlighting successful applications and identifying knowledge gaps and potential challenges.
Collapse
Affiliation(s)
- Francesca Mancini
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Francesca Micoli
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Francesca Necchi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Mariagrazia Pizza
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | - Omar Rossi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health (GVGH), Siena, Italy
| |
Collapse
|
242
|
Dou L, Meng X, Yang H, Dong H. Advances in technology and applications of nanoimmunotherapy for cancer. Biomark Res 2021; 9:63. [PMID: 34419164 PMCID: PMC8379775 DOI: 10.1186/s40364-021-00321-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/06/2021] [Indexed: 01/01/2023] Open
Abstract
Host-tumor immune interactions play critical roles in the natural history of tumors, including oncogenesis, progress and metastasis. On the one hand, neoantigens have the potential to drive a tumor-specific immune response. In tumors, immunogenic cell death (ICD) triggered by various inducers can initiate a strong host anti-immune response. On the other hand, the tolerogenic tumor immune microenvironment suppresses host immune responses that eradicate tumor cells and impair the effect of tumor therapy. Therefore, a deeper understanding and more effective manipulation of the intricate host-tumor immune interaction involving the host, tumor cells and the corresponding tumor immune microenvironment are required. Despite the encouraging breakthroughs resulting from tumor immunotherapy, no single strategy has elicited sufficient or sustained antitumor immune responses in most patients with specific malignancies due to limited activation of specific antitumor immune responses and inadequate remodeling of the tolerogenic tumor immune microenvironment. However, nanotechnology provides a unique paradigm to simultaneously tackle all these challenges, including effective “targeted” delivery of tumor antigens, sustained ICD mediation, and “cold” tumor microenvironment remodeling. In this review, we focus on several key concepts in host-tumor immune interactions and discuss the corresponding therapeutic strategy based on the application of nanoparticles.
Collapse
Affiliation(s)
- Lei Dou
- Department of Gerontology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Surgery, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiangdan Meng
- Research Center for Bioengineering and Sensing Technology, University of Science & Technology Beijing, Beijing, 100083, China
| | - Huiyuan Yang
- Department of Surgery, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haifeng Dong
- Research Center for Bioengineering and Sensing Technology, University of Science & Technology Beijing, Beijing, 100083, China. .,School of Biomedical Engineering, Health Science Centre, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
243
|
Passeri L, Marta F, Bassi V, Gregori S. Tolerogenic Dendritic Cell-Based Approaches in Autoimmunity. Int J Mol Sci 2021; 22:8415. [PMID: 34445143 PMCID: PMC8395087 DOI: 10.3390/ijms22168415] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) dictate the outcomes of tissue-specific immune responses. In the context of autoimmune diseases, DCs instruct T cells to respond to antigens (Ags), including self-Ags, leading to organ damage, or to becoming regulatory T cells (Tregs) promoting and perpetuating immune tolerance. DCs can acquire tolerogenic properties in vitro and in vivo in response to several stimuli, a feature that opens the possibility to generate or to target DCs to restore tolerance in autoimmune settings. We present an overview of the different subsets of human DCs and of the regulatory mechanisms associated with tolerogenic (tol)DC functions. We review the role of DCs in the induction of tissue-specific autoimmunity and the current approaches exploiting tolDC-based therapies or targeting DCs in vivo for the treatment of autoimmune diseases. Finally, we discuss limitations and propose future investigations for improving the knowledge on tolDCs for future clinical assessment to revert and prevent autoimmunity. The continuous expansion of tolDC research areas will lead to improving the understanding of the role that DCs play in the development and treatment of autoimmunity.
Collapse
Affiliation(s)
- Laura Passeri
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
- San Raffaele Scientific Institute IRCCS, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Fortunato Marta
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
| | - Virginia Bassi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
- San Raffaele Scientific Institute IRCCS, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
| |
Collapse
|
244
|
Cid R, Bolívar J. Platforms for Production of Protein-Based Vaccines: From Classical to Next-Generation Strategies. Biomolecules 2021; 11:1072. [PMID: 34439738 PMCID: PMC8394948 DOI: 10.3390/biom11081072] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
To date, vaccination has become one of the most effective strategies to control and reduce infectious diseases, preventing millions of deaths worldwide. The earliest vaccines were developed as live-attenuated or inactivated pathogens, and, although they still represent the most extended human vaccine types, they also face some issues, such as the potential to revert to a pathogenic form of live-attenuated formulations or the weaker immune response associated with inactivated vaccines. Advances in genetic engineering have enabled improvements in vaccine design and strategies, such as recombinant subunit vaccines, have emerged, expanding the number of diseases that can be prevented. Moreover, antigen display systems such as VLPs or those designed by nanotechnology have improved the efficacy of subunit vaccines. Platforms for the production of recombinant vaccines have also evolved from the first hosts, Escherichia coli and Saccharomyces cerevisiae, to insect or mammalian cells. Traditional bacterial and yeast systems have been improved by engineering and new systems based on plants or insect larvae have emerged as alternative, low-cost platforms. Vaccine development is still time-consuming and costly, and alternative systems that can offer cost-effective and faster processes are demanding to address infectious diseases that still do not have a treatment and to face possible future pandemics.
Collapse
Affiliation(s)
- Raquel Cid
- ADL Bionatur Solutions S.A., Av. del Desarrollo Tecnológico 11, 11591 Jerez de la Frontera, Spain
| | - Jorge Bolívar
- Department of Biomedicine, Biotechnology and Public Health-Biochemistry and Molecular Biology, Campus Universitario de Puerto Real, University of Cadiz, 11510 Puerto Real, Spain
| |
Collapse
|
245
|
Azhar A, Hassan N, Singh M, Al-Hosaini K, Kamal MA. Synopsis on Pharmotechnological Approaches in Diagnostic to Management Strategies in Fighting Against COVID-19. Curr Pharm Des 2021; 27:4086-4099. [PMID: 34269664 DOI: 10.2174/1381612827666210715154004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
Nanoparticles (NPs) are projected to play a significant role in fighting against coronavirus disease (COVID-19). The various properties of NPs like magnetic and optical can be exploited to build diagnostic test kits. The unembellished morphological and physiochemical resemblances of SARS-CoV-2 with synthetic NPs make them a potent tool for mediation. Nanoparticles can be analytically functionalized with different proteins, polymers, and functional groups to perform specific inhibitory functions while also serving as delivery vehicles . Moreover, NPs can also be employed to prepare broad-spectrum respiratory drugs and vaccines that can guard seasonal flu and prepare the human race for the pandemic in the future. The present review outlines the role of NPs in detection, diagnostic and therapeutic against members of the coronavirus family. We emphasize nanomaterial-based approaches to address coronaviruses in general and SARS-CoV-2 in particular. We discuss NPs based detection systems like graphene (G-FET), biosensors, and plasmonic photothermal associated sensors. Inorganic, organic virus-like & self-assembly protein (VLP), and photodynamic inactivation of SARS-CoV-2 are also presented as therapeutic approaches exploiting NPs.
Collapse
Affiliation(s)
- Asim Azhar
- Aligarh College of Education, Aligarh Uttar Pradesh, India
| | - Nazia Hassan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Manvi Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Khaled Al-Hosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451. Saudi Arabia
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
246
|
Fries CN, Chen JL, Dennis ML, Votaw NL, Eudailey J, Watts BE, Hainline KM, Cain DW, Barfield R, Chan C, Moody MA, Haynes BF, Saunders KO, Permar SR, Fouda GG, Collier JH. HIV envelope antigen valency on peptide nanofibers modulates antibody magnitude and binding breadth. Sci Rep 2021; 11:14494. [PMID: 34262096 PMCID: PMC8280189 DOI: 10.1038/s41598-021-93702-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/23/2021] [Indexed: 01/02/2023] Open
Abstract
A major challenge in developing an effective vaccine against HIV-1 is the genetic diversity of its viral envelope. Because of the broad range of sequences exhibited by HIV-1 strains, protective antibodies must be able to bind and neutralize a widely mutated viral envelope protein. No vaccine has yet been designed which induces broadly neutralizing or protective immune responses against HIV in humans. Nanomaterial-based vaccines have shown the ability to generate antibody and cellular immune responses of increased breadth and neutralization potency. Thus, we have developed supramolecular nanofiber-based immunogens bearing the HIV gp120 envelope glycoprotein. These immunogens generated antibody responses that had increased magnitude and binding breadth compared to soluble gp120. By varying gp120 density on nanofibers, we determined that increased antigen valency was associated with increased antibody magnitude and germinal center responses. This study presents a proof-of-concept for a nanofiber vaccine platform generating broad, high binding antibody responses against the HIV-1 envelope glycoprotein.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maria L Dennis
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Joshua Eudailey
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brian E Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin O Saunders
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medicine, New York, NY, 10065, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
247
|
Hassett KJ, Higgins J, Woods A, Levy B, Xia Y, Hsiao CJ, Acosta E, Almarsson Ö, Moore MJ, Brito LA. Impact of lipid nanoparticle size on mRNA vaccine immunogenicity. J Control Release 2021; 335:237-246. [PMID: 34019945 DOI: 10.1016/j.jconrel.2021.05.021] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 01/03/2023]
Abstract
Lipid nanoparticles (LNP) are effective delivery vehicles for messenger RNA (mRNA) and have shown promise for vaccine applications. Yet there are no published reports detailing how LNP biophysical properties can impact vaccine performance. In our hands, a retrospective analysis of mRNA LNP vaccine in vivo studies revealed a relationship between LNP particle size and immunogenicity in mice using LNPs of various compositions. To further investigate this, we designed a series of studies to systematically change LNP particle size without altering lipid composition and evaluated biophysical properties and immunogenicity of the resulting LNPs. While small diameter LNPs were substantially less immunogenic in mice, all particle sizes tested yielded a robust immune response in non-human primates (NHP).
Collapse
Affiliation(s)
- Kimberly J Hassett
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Jaclyn Higgins
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Angela Woods
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Becca Levy
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Yan Xia
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Chiaowen Joyce Hsiao
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Edward Acosta
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Örn Almarsson
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Melissa J Moore
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America
| | - Luis A Brito
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, United States of America.
| |
Collapse
|
248
|
Wu H, Fu X, Zhai Y, Gao S, Yang X, Zhai G. Development of Effective Tumor Vaccine Strategies Based on Immune Response Cascade Reactions. Adv Healthc Mater 2021; 10:e2100299. [PMID: 34021717 DOI: 10.1002/adhm.202100299] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/11/2021] [Indexed: 12/13/2022]
Abstract
To solve the problems of high toxicity and poor efficacy of existing tumor treatment methods, researchers have developed a variety of tumor immunotherapies. Among them, tumor vaccines activate antigen-presenting cells and T lymphocytes upstream of the cancer-immunity cycle are considered the most promising therapy to activate the immune system. Nanocarriers are considered the most promising tumor vaccine delivery vehicles, including polymer nanocarriers, lipid nanocarriers, inorganic nanocarriers, and biomimetic nanocarriers that have been developed for vaccine delivery. Based on the cascade reaction for tumor vaccines to exert their effects, this review summarizes the four key factors for the design and construction of nano-tumor vaccines. The composition and functional characteristics of the corresponding preferred nanocarriers are illustrated to provide a reference for the development of effective tumor vaccines. Finally, potential challenges and perspectives are illustrated in the hope of improving the efficacy of tumor vaccine immunotherapy and accelerating the clinical transformation of next-generation tumor vaccines.
Collapse
Affiliation(s)
- Hang Wu
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Xianglei Fu
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry University of Utah Salt Lake City UT 84124 USA
| | - Shan Gao
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Xiaoye Yang
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences Key Laboratory of Chemical Biology Ministry of Education Shandong University Jinan 250012 China
| |
Collapse
|
249
|
An W, Defaus S, Andreu D, Rivera-Gil P. In Vivo Sustained Release of Peptide Vaccine Mediated by Dendritic Mesoporous Silica Nanocarriers. Front Immunol 2021; 12:684612. [PMID: 34220835 PMCID: PMC8244784 DOI: 10.3389/fimmu.2021.684612] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
Mesoporous silica nanoparticles have drawn increasing attention as promising candidates in vaccine delivery. Previous studies evaluating silica-based vaccine delivery systems concentrated largely on macromolecular antigens, such as inactivated whole viruses. In this study, we synthesized dendritic mesoporous silica nanoparticles (DMSNs), and we evaluated their effectiveness as delivery platforms for peptide-based subunit vaccines. We encapsulated and tested in vivo an earlier reported foot-and-mouth disease virus (FMDV) peptide vaccine (B2T). The B2T@DMSNs formulation contained the peptide vaccine and the DMSNs without further need of other compounds neither adjuvants nor emulsions. We measured in vitro a sustained release up to 930 h. B2T@DMSNs-57 and B2T@DMSNs-156 released 23.7% (135 µg) and 22.8% (132 µg) of the total B2T. The formation of a corona of serum proteins around the DMSNs increased the B2T release up to 61% (348 µg/mg) and 80% (464 µg/mg) for B2T@DMSNs-57 and B2T@DMSNs-156. In vitro results point out to a longer sustained release, assisted by the formation of a protein corona around DMSNs, compared to the reference formulation (i.e., B2T emulsified in Montanide). We further confirmed in vivo immunogenicity of B2T@DMSNs in a particle size-dependent manner. Since B2T@DMSNs elicited specific immune responses in mice with high IgG production like the reference B2T@Montanide™, self-adjuvant properties of the DMSNs could be ascribed. Our results display DMSNs as efficacious nanocarriers for peptide-based vaccine administration.
Collapse
Affiliation(s)
- Weiteng An
- Integrative Biomedical Materials and Nanomedicine Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sira Defaus
- Proteomics and Protein Chemistry Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Andreu
- Proteomics and Protein Chemistry Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Pilar Rivera-Gil
- Integrative Biomedical Materials and Nanomedicine Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
250
|
Kumar M, Kumari N, Thakur N, Bhatia SK, Saratale GD, Ghodake G, Mistry BM, Alavilli H, Kishor DS, Du X, Chung SM. A Comprehensive Overview on the Production of Vaccines in Plant-Based Expression Systems and the Scope of Plant Biotechnology to Combat against SARS-CoV-2 Virus Pandemics. PLANTS (BASEL, SWITZERLAND) 2021; 10:1213. [PMID: 34203729 PMCID: PMC8232254 DOI: 10.3390/plants10061213] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 06/12/2021] [Indexed: 12/23/2022]
Abstract
Many pathogenic viral pandemics have caused threats to global health; the COVID-19 pandemic is the latest. Its transmission is growing exponentially all around the globe, putting constraints on the health system worldwide. A novel coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), causes this pandemic. Many candidate vaccines are available at this time for COVID-19, and there is a massive international race underway to procure as many vaccines as possible for each country. However, due to heavy global demand, there are strains in global vaccine production. The use of a plant biotechnology-based expression system for vaccine production also represents one part of this international effort, which is to develop plant-based heterologous expression systems, virus-like particles (VLPs)-vaccines, antiviral drugs, and a rapid supply of antigen-antibodies for detecting kits and plant origin bioactive compounds that boost the immunity and provide tolerance to fight against the virus infection. This review will look at the plant biotechnology platform that can provide the best fight against this global pandemic.
Collapse
Affiliation(s)
- Manu Kumar
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| | - Nisha Kumari
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Nishant Thakur
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea;
| | - Shashi Kant Bhatia
- Department of Biological Engineering, College of Engineering, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea;
| | - Ganesh Dattatraya Saratale
- Department of Food Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (G.D.S.); (B.M.M.)
| | - Gajanan Ghodake
- Department of Biological and Environmental Science, Dongguk University, Seoul 10326, Korea;
| | - Bhupendra M. Mistry
- Department of Food Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (G.D.S.); (B.M.M.)
| | - Hemasundar Alavilli
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea;
| | - D. S. Kishor
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| | - Xueshi Du
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| | - Sang-Min Chung
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| |
Collapse
|