101
|
Ftouh M, Kalboussi N, Abid N, Sfar S, Mignet N, Bahloul B. Contribution of Nanotechnologies to Vaccine Development and Drug Delivery against Respiratory Viruses. PPAR Res 2021; 2021:6741290. [PMID: 34721558 PMCID: PMC8550859 DOI: 10.1155/2021/6741290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
According to the Center for Disease Control and Prevention (CDC), the coronavirus disease 2019, a respiratory viral illness linked to significant morbidity, mortality, production loss, and severe economic depression, was the third-largest cause of death in 2020. Respiratory viruses such as influenza, respiratory syncytial virus, SARS-CoV-2, and adenovirus, are among the most common causes of respiratory illness in humans, spreading as pandemics or epidemics throughout all continents. Nanotechnologies are particles in the nanometer range made from various compositions. They can be lipid-based, polymer-based, protein-based, or inorganic in nature, but they are all bioinspired and virus-like. In this review, we aimed to present a short review of the different nanoparticles currently studied, in particular those which led to publications in the field of respiratory viruses. We evaluated those which could be beneficial for respiratory disease-based viruses; those which already have contributed, such as lipid nanoparticles in the context of COVID-19; and those which will contribute in the future either as vaccines or antiviral drug delivery systems. We present a short assessment based on a critical selection of evidence indicating nanotechnology's promise in the prevention and treatment of respiratory infections.
Collapse
Affiliation(s)
- Mahdi Ftouh
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Nesrine Kalboussi
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
- Sahloul University Hospital, Pharmacy Department, Sousse, Tunisia
| | - Nabil Abid
- Department of Biotechnology, High Institute of Biotechnology of Sidi Thabet, University of Manouba, BP-66, 2020 Ariana, Tunis, Tunisia
- Laboratory of Transmissible Diseases and Biological Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Rue Ibn Sina, 5000 Monastir, Tunisia
| | - Souad Sfar
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Nathalie Mignet
- University of Paris, INSERM, CNRS, UTCBS, Faculté de Pharmacie, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Badr Bahloul
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
| |
Collapse
|
102
|
Yang MH, Hu CC, Wong CH, Liang JJ, Ko HY, He MH, Lin YL, Lin NS, Hsu YH. Convenient Auto-Processing Vector Based on Bamboo Mosaic Virus for Presentation of Antigens Through Enzymatic Coupling. Front Immunol 2021; 12:739837. [PMID: 34721406 PMCID: PMC8551676 DOI: 10.3389/fimmu.2021.739837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/28/2021] [Indexed: 11/25/2022] Open
Abstract
We have developed a new binary epitope-presenting CVP platform based on bamboo mosaic virus (BaMV) by using the sortase A (SrtA)-mediated ligation technology. The reconstructed BaMV genome harbors two modifications: 1) a coat protein (CP) with N-terminal extension of the tobacco etch virus (TEV) protease recognition site plus 4 extra glycine (G) residues as the SrtA acceptor; and 2) a TEV protease coding region replacing that of the triple-gene-block proteins. Inoculation of such construct, pKB5G, on Nicotiana benthamiana resulted in the efficient production of filamentous CVPs ready for SrtA-mediated ligation with desired proteins. The second part of the binary platform includes an expression vector for the bacterial production of donor proteins. We demonstrated the applicability of the platform by using the recombinant envelope protein domain III (rEDIII) of Japanese encephalitis virus (JEV) as the antigen. Up to 40% of the BaMV CP subunits in each CVP were loaded with rEDIII proteins in 1 min. The rEDIII-presenting BaMV CVPs (BJLPET5G) could be purified using affinity chromatography. Immunization assays confirmed that BJLPET5G could induce the production of neutralizing antibodies against JEV infections. The binary platform could be adapted as a useful alternative for the development and mass production of vaccine candidates.
Collapse
MESH Headings
- Aminoacyltransferases/genetics
- Aminoacyltransferases/metabolism
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antigens, Viral/administration & dosage
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Bacterial Proteins/genetics
- Bacterial Proteins/metabolism
- Cell Line
- Cysteine Endopeptidases/genetics
- Cysteine Endopeptidases/metabolism
- Disease Models, Animal
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis, Japanese/blood
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/prevention & control
- Encephalitis, Japanese/virology
- Endopeptidases/genetics
- Endopeptidases/metabolism
- Escherichia coli/genetics
- Escherichia coli/immunology
- Escherichia coli/metabolism
- Female
- Genetic Vectors
- Immunogenicity, Vaccine
- Japanese Encephalitis Vaccines/administration & dosage
- Japanese Encephalitis Vaccines/genetics
- Japanese Encephalitis Vaccines/immunology
- Mice, Inbred BALB C
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/immunology
- Plants, Genetically Modified/metabolism
- Potexvirus/enzymology
- Potexvirus/genetics
- Potexvirus/immunology
- Nicotiana/genetics
- Nicotiana/immunology
- Nicotiana/metabolism
- Virion/enzymology
- Virion/genetics
- Virion/immunology
- Mice
Collapse
Affiliation(s)
- Ming-Hao Yang
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Chung-Chi Hu
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
- Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Chi-Hzeng Wong
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Ying Ko
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Meng-Hsun He
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Na-Sheng Lin
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Yau-Heiu Hsu
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
- Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
103
|
Dalton KP, Alvarado C, Reytor E, del Carmen Nuñez M, Podadera A, Martínez-Alonso D, Alonso JMM, Nicieza I, Gómez-Sebastián S, Dalton RM, Parra F, Escribano JM. Chimeric VLPs Bearing VP60 from Two Serotypes of Rabbit Haemorrhagic Disease Virus Are Protective against Both Viruses. Vaccines (Basel) 2021; 9:vaccines9091005. [PMID: 34579243 PMCID: PMC8472679 DOI: 10.3390/vaccines9091005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
The VP60 capsid protein from rabbit haemorrhagic disease virus (RHDV), the causative agent of one of the most economically important disease in rabbits worldwide, forms virus-like particles (VLPs) when expressed using heterologous protein expression systems such as recombinant baculovirus, yeasts, plants or mammalian cell cultures. To prevent RHDV dissemination, it would be beneficial to develop a bivalent vaccine including both RHDV GI.1- and RHDV GI.2-derived VLPs to achieve robust immunisation against both serotypes. In the present work, we developed a strategy of production of a dual-serving RHDV vaccine co-expressing the VP60 proteins from the two RHDV predominant serotypes using CrisBio technology, which uses Tricholusia ni insect pupae as natural bioreactors, which are programmed by recombinant baculovirus vectors. Co-infecting the insect pupae with two baculovirus vectors expressing the RHDV GI.1- and RHDV GI.2-derived VP60 proteins, we obtained chimeric VLPs incorporating both proteins as determined by using serotype-specific monoclonal antibodies. The resulting VLPs showed the typical size and shape of this calicivirus as determined by electron microscopy. Rabbits immunised with the chimeric VLPs were fully protected against a lethal challenge infection with the two RHDV serotypes. This study demonstrates that it is possible to generate a dual cost-effective vaccine against this virus using a single production and purification process, greatly simplifying vaccine manufacturing.
Collapse
Affiliation(s)
- Kevin P. Dalton
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Campus El Cristo, Universidad de Oviedo, Edificio Santiago Gascón, 33006 Oviedo, Spain; (K.P.D.); (A.P.); (J.M.M.A.); (I.N.); (F.P.)
| | - Carmen Alvarado
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
| | - Edel Reytor
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
| | - Maria del Carmen Nuñez
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
| | - Ana Podadera
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Campus El Cristo, Universidad de Oviedo, Edificio Santiago Gascón, 33006 Oviedo, Spain; (K.P.D.); (A.P.); (J.M.M.A.); (I.N.); (F.P.)
| | - Diego Martínez-Alonso
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
| | - Jose Manuel Martin Alonso
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Campus El Cristo, Universidad de Oviedo, Edificio Santiago Gascón, 33006 Oviedo, Spain; (K.P.D.); (A.P.); (J.M.M.A.); (I.N.); (F.P.)
| | - Ines Nicieza
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Campus El Cristo, Universidad de Oviedo, Edificio Santiago Gascón, 33006 Oviedo, Spain; (K.P.D.); (A.P.); (J.M.M.A.); (I.N.); (F.P.)
| | - Silvia Gómez-Sebastián
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
| | - Romy M. Dalton
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
| | - Francisco Parra
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Campus El Cristo, Universidad de Oviedo, Edificio Santiago Gascón, 33006 Oviedo, Spain; (K.P.D.); (A.P.); (J.M.M.A.); (I.N.); (F.P.)
| | - José M. Escribano
- Alternative Gene Expression S.L. Ronda de Poniente 14, Tres Cantos, 28760 Madrid, Spain; (C.A.); (E.R.); (M.d.C.N.); (D.M.-A.); (S.G.-S.); (R.M.D.)
- Correspondence:
| |
Collapse
|
104
|
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: 0.8] [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
|
105
|
Borkens Y. [Toxoplasma gondii-Current drugs and future vaccines against an underestimated protozoan infection]. Internist (Berl) 2021; 62:1123-1132. [PMID: 34467425 DOI: 10.1007/s00108-021-01155-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 12/01/2022]
Abstract
Toxoplasma gondii is a unicellular organism of the Apicomplexa that occurs worldwide and is therefore a close relative of the malaria pathogen Plasmodium. As T. gondii infests every warm-blooded vertebrate species as an intermediate host and has a very high prevalence worldwide, toxoplasmosis is one of the most important international foodborne diseases. Potential vaccines (human as well as veterinary) play a crucial role in controlling this disease.
Collapse
Affiliation(s)
- Yannick Borkens
- College of Public Health, Medical and Veterinary Science, James Cook University, 1 James Cook Drive, 4811, Townsville, Queensland, Australien.
| |
Collapse
|
106
|
Citiulo F, Crosatti C, Cattivelli L, Biselli C. Frontiers in the Standardization of the Plant Platform for High Scale Production of Vaccines. PLANTS (BASEL, SWITZERLAND) 2021; 10:1828. [PMID: 34579360 PMCID: PMC8467261 DOI: 10.3390/plants10091828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
The recent COVID-19 pandemic has highlighted the value of technologies that allow a fast setup and production of biopharmaceuticals in emergency situations. The plant factory system can provide a fast response to epidemics/pandemics. Thanks to their scalability and genome plasticity, plants represent advantageous platforms to produce vaccines. Plant systems imply less complicated production processes and quality controls with respect to mammalian and bacterial cells. The expression of vaccines in plants is based on transient or stable transformation systems and the recent progresses in genome editing techniques, based on the CRISPR/Cas method, allow the manipulation of DNA in an efficient, fast, and easy way by introducing specific modifications in specific sites of a genome. Nonetheless, CRISPR/Cas is far away from being fully exploited for vaccine expression in plants. In this review, an overview of the potential conjugation of the renewed vaccine technologies (i.e., virus-like particles-VLPs, and industrialization of the production process) with genome editing to produce vaccines in plants is reported, illustrating the potential advantages in the standardization of the plant platforms, with the overtaking of constancy of large-scale production challenges, facilitating regulatory requirements and expediting the release and commercialization of the vaccine products of genome edited plants.
Collapse
Affiliation(s)
- Francesco Citiulo
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy;
| | - Cristina Crosatti
- Council for Agricultural Research and Economics, Research Centre for Genomics and Bioinformatics, Via San Protaso 302, 29017 Fiorenzuola d’Arda, Italy; (C.C.); (L.C.)
| | - Luigi Cattivelli
- Council for Agricultural Research and Economics, Research Centre for Genomics and Bioinformatics, Via San Protaso 302, 29017 Fiorenzuola d’Arda, Italy; (C.C.); (L.C.)
| | - Chiara Biselli
- Council for Agricultural Research and Economics, Research Centre for Viticulture and Enology, Viale Santa Margherita 80, 52100 Arezzo, Italy
| |
Collapse
|
107
|
Gunathilake TMSU, Ching YC, Uyama H, Chuah CH. Nanotherapeutics for treating coronavirus diseases. J Drug Deliv Sci Technol 2021; 64:102634. [PMID: 34127930 PMCID: PMC8190278 DOI: 10.1016/j.jddst.2021.102634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Viral diseases have recently become a threat to human health and rapidly become a significant cause of mortality with a continually exacerbated unfavorable socio-economic impact. Coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome (MERS-CoV), and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), have threatened human life, with immense accompanying morbidity rates; the COVID-19 (caused by SARS-CoV-2) epidemic has become a severe threat to global public health. In addition, the design process of antiviral medications usually takes years before the treatments can be made readily available. Hence, it is necessary to invest scientifically and financially in a technology platform that can then be quickly repurposed on demand to be adequately positioned for this kind of pandemic situation through lessons learned from the previous pandemics. Nanomaterials/nanoformulations provide such platform technologies, and a proper investigation into their basic science and biological interactions would be of great benefit for potential vaccine and therapeutic development. In this respect, intelligent and advanced nano-based technologies provide specific physico-chemical properties, which can help fix the key issues related to the treatments of viral infections. This review aims to provide an overview of the latest research on the effective use of nanomaterials in the treatment of coronaviruses. Also raised are the problems, perspectives of antiviral nanoformulations, and the possibility of using nanomaterials effectively against current pandemic situations.
Collapse
Affiliation(s)
- Thennakoon M Sampath U Gunathilake
- Centre of Advanced Materials (CAM), Faculty of Engineering, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Department of Chemical Engineering, Faculty of Engineering, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yern Chee Ching
- Centre of Advanced Materials (CAM), Faculty of Engineering, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Department of Chemical Engineering, Faculty of Engineering, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hiroshi Uyama
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Cheng Hock Chuah
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
108
|
Song SJ, Shin GI, Noh J, Lee J, Kim DH, Ryu G, Ahn G, Jeon H, Diao HP, Park Y, Kim MG, Kim WY, Kim YJ, Sohn EJ, Song CS, Hwang I. Plant-based, adjuvant-free, potent multivalent vaccines for avian influenza virus via Lactococcus surface display. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2021; 63:1505-1520. [PMID: 34051041 DOI: 10.1111/jipb.13141] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/26/2021] [Indexed: 05/28/2023]
Abstract
Influenza epidemics frequently and unpredictably break out all over the world, and seriously affect the breeding industry and human activity. Inactivated and live attenuated viruses have been used as protective vaccines but exhibit high risks for biosafety. Subunit vaccines enjoy high biosafety and specificity but have a few weak points compared to inactivated virus or live attenuated virus vaccines, especially in low immunogenicity. In this study, we developed a new subunit vaccine platform for a potent, adjuvant-free, and multivalent vaccination. The ectodomains of hemagglutinins (HAs) of influenza viruses were expressed in plants as trimers (tHAs) to mimic their native forms. tHAs in plant extracts were directly used without purification for binding to inactivated Lactococcus (iLact) to produce iLact-tHAs, an antigen-carrying bacteria-like particle (BLP). tHAs BLP showed strong immune responses in mice and chickens without adjuvants. Moreover, simultaneous injection of two different antigens by two different formulas, tHAH5N6 + H9N2 BLP or a combination of tHAH5N6 BLP and tHAH9N2 BLP, led to strong immune responses to both antigens. Based on these results, we propose combinations of plant-based antigen production and BLP-based delivery as a highly potent and cost-effective platform for multivalent vaccination for subunit vaccines.
Collapse
Affiliation(s)
- Shi-Jian Song
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Gyeong-Im Shin
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, 660-701, Korea
| | | | - Jiho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Korea
| | - Deok-Hwan Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Korea
| | - Gyeongryul Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, 660-701, Korea
| | - Gyeongik Ahn
- Division of Applied Life Science (BK21 PLUS), Institute of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Korea
| | - Hyungmin Jeon
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Hai-Ping Diao
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Youngmin Park
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
- Bioapp, Inc., Pohang Technopark Complex, Pohang, 37668, Korea
| | - Min Gab Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju, 660-701, Korea
| | - Woe-Yeon Kim
- Division of Applied Life Science (BK21 PLUS), Institute of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Korea
| | - Young-Jin Kim
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Eun-Ju Sohn
- Bioapp, Inc., Pohang Technopark Complex, Pohang, 37668, Korea
| | - Chang Seon Song
- KCAV Inc., Gwangjin-gu, 05029, Korea
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Korea
| | - Inhwan Hwang
- Department of Life Science, Pohang University of Science and Technology, Pohang, 37673, Korea
| |
Collapse
|
109
|
Liu ZH, Xu HL, Han GW, Tao LN, Lu Y, Zheng SY, Fang WH, He F. A self-assembling nanoparticle: Implications for the development of thermostable vaccine candidates. Int J Biol Macromol 2021; 183:2162-2173. [PMID: 34102236 DOI: 10.1016/j.ijbiomac.2021.06.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022]
Abstract
Effective controls on viral infections rely on the continuous development in vaccine technology. Nanoparticle (NP) antigens are highly immunogenic based on their unique physicochemical properties, making them molecular scaffolds to present soluble vaccine antigens. Here, viral targets (113-354 aas) were genetically fused to N terminal of mi3, a protein that self-assembles into nanoparticles composed of 60 subunits. With transmission electron microscopy, it was confirmed that target-mi3 fusion proteins which have insertions of up to 354 aas in N terminal form intact NPs. Moreover, viral targets are surface-displayed on NPs as indicated in dynamic light scattering. NPs exhibit perfect stability after long-term storage at room temperature. Moreover, SP-E2-mi3 NPs enhance antigen uptake and maturation in dendritic cells (DCs) via up-regulating marker molecules and immunostimulatory cytokines. Importantly, in a mouse model, SP-E2-mi3 nanovaccines against Classical swine fever virus (CSFV) remarkably improved CSFV-specific neutralizing antibodies (NAbs) and cellular immunity related cytokines (IFN-γ and IL-4) as compared to monomeric E2. Specially, improved NAb response with more than tenfold increase in NAb titer against both CSFV Shimen and HZ-08 strains indicated better cross-protection against different genotypes. Collectively, this structure-based, self-assembling NP provides an attractive platform to improve the potency of subunit vaccine for emerging pathogens.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/pharmacology
- Cells, Cultured
- Classical Swine Fever/blood
- Classical Swine Fever/immunology
- Classical Swine Fever/prevention & control
- Classical Swine Fever/virology
- Classical Swine Fever Virus/immunology
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Disease Models, Animal
- Drug Stability
- Female
- Immunogenicity, Vaccine
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nanoparticles
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- Swine
- Temperature
- Vaccines, Subunit/immunology
- Vaccines, Subunit/pharmacology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/pharmacology
- Viral Vaccines/immunology
- Viral Vaccines/pharmacology
- Mice
Collapse
Affiliation(s)
- Ze-Hui Liu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Hui-Ling Xu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Guang-Wei Han
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Li-Na Tao
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Ying Lu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Su-Ya Zheng
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Wei-Huan Fang
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China.
| | - Fang He
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China.
| |
Collapse
|
110
|
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: 88] [Impact Index Per Article: 22.0] [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
|
111
|
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: 0.5] [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
|
112
|
Lim J, Cheong Y, Kim YS, Chae W, Hwang BJ, Lee J, Jang YH, Roh YH, Seo SU, Seong BL. RNA-dependent assembly of chimeric antigen nanoparticles as an efficient H5N1 pre-pandemic vaccine platform. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102438. [PMID: 34256061 DOI: 10.1016/j.nano.2021.102438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022]
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) pose a significant threat to human health, with high mortality rates, and require effective vaccines. We showed that, harnessed with novel RNA-mediated chaperone function, hemagglutinin (HA) of H5N1 HPAIV could be displayed as an immunologically relevant conformation on self-assembled chimeric nanoparticles (cNP). A tri-partite monomeric antigen was designed including: i) an RNA-interaction domain (RID) as a docking tag for RNA to enable chaperna function (chaperna: chaperone + RNA), ii) globular head domain (gd) of HA as a target antigen, and iii) ferritin as a scaffold for 24 mer-assembly. The immunization of mice with the nanoparticles (~46 nm) induced a 25-30 fold higher neutralizing capacity of the antibody and provided cross-protection from homologous and heterologous lethal challenges. This study suggests that cNP assembly is conducive to eliciting antibodies against the conserved region in HA, providing potent and broad protective efficacy.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Birds/virology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/therapeutic use
- Humans
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/chemistry
- Influenza Vaccines/immunology
- Influenza Vaccines/therapeutic use
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Mice
- Nanoparticles/chemistry
- Nanoparticles/therapeutic use
- Pandemics
- RNA/genetics
- RNA/immunology
- RNA/therapeutic use
Collapse
Affiliation(s)
- Jongkwan Lim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yucheol Cheong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Seok Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Wonil Chae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Beom Jeung Hwang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jinhee Lee
- Department of Integrated OMICS for Biomedical Science, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yo Han Jang
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Biotechnology, Andong National University, Andong, Republic of Korea
| | - Young Hoon Roh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Baik L Seong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Yonsei University, Seoul, Republic of Korea; Vaccine Innovative Technology Alliance-Korea, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
113
|
Ma J, Wang H, Zheng X, Wu H, Yang S, Xia X. Western equine encephalitis virus virus-like particles from an insect cell-baculovirus system elicit the strong immune responses in mice. Biotechnol J 2021; 16:e2100008. [PMID: 34176228 DOI: 10.1002/biot.202100008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/07/2022]
Abstract
Western equine encephalitis virus (WEEV) causes lethal encephalitis in humans and equines, and it poses a serious public health threat in many countries. Therefore, the development of an efficient vaccine remains an important challenge for the prevention of WEEV infection. This study presents the first description of WEEV virus-like particles (VLPs) generated from insect cells using recombinant baculoviruses. WEEV VLPs with 206 adjuvant could trigger a strong cellular immune response; increase the levels of IL-2, IL-4 and IFN-γ; and induce a high level of neutralizing antibodies against WEEV in mice. These data showed that the insect cell-baculovirus system is suitable for the production of WEEV VLPs and that these VLPs could elicit the strong immunogenicity in mice. These results suggest a new, nonreplicating, and effective vaccine candidate against WEEV infection.
Collapse
Affiliation(s)
- JinZhu Ma
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China.,College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - HuaLei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - XueXing Zheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - HongXia Wu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - SongTao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - XianZhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| |
Collapse
|
114
|
Raman R, Patel KJ, Ranjan K. COVID-19: Unmasking Emerging SARS-CoV-2 Variants, Vaccines and Therapeutic Strategies. Biomolecules 2021; 11:993. [PMID: 34356617 PMCID: PMC8301790 DOI: 10.3390/biom11070993] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of the coronavirus disease 2019 (COVID-19) pandemic, which has been a topic of major concern for global human health. The challenge to restrain the COVID-19 pandemic is further compounded by the emergence of several SARS-CoV-2 variants viz. B.1.1.7 (Alpha), B.1.351 (Beta), P1 (Gamma) and B.1.617.2 (Delta), which show increased transmissibility and resistance towards vaccines and therapies. Importantly, there is convincing evidence of increased susceptibility to SARS-CoV-2 infection among individuals with dysregulated immune response and comorbidities. Herein, we provide a comprehensive perspective regarding vulnerability of SARS-CoV-2 infection in patients with underlying medical comorbidities. We discuss ongoing vaccine (mRNA, protein-based, viral vector-based, etc.) and therapeutic (monoclonal antibodies, small molecules, plasma therapy, etc.) modalities designed to curb the COVID-19 pandemic. We also discuss in detail, the challenges posed by different SARS-CoV-2 variants of concern (VOC) identified across the globe and their effects on therapeutic and prophylactic interventions.
Collapse
Affiliation(s)
- Renuka Raman
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Krishna J. Patel
- Mount Sinai Innovation Partners, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kishu Ranjan
- School of Medicine, Yale University, New Haven, CT 06519, USA
| |
Collapse
|
115
|
Ram Kumar PS, Rencilin CF, Sundar K. Emerging nanomaterials for cancer immunotherapy. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy is a unique approach to treat cancer that targets tumours besides triggering the immune cells. It attempts to harness the supremacy and specificity of immune cells for the regression of malignancy. The key strategy of immunotherapy is that it boosts the natural defence and manipulates the immune system at both cellular and molecular levels. Long-lasting anti-tumour response, reduced metastasis, and recurrence can be achieved with immunotherapy than conventional treatments. For example, targeting cytotoxic T-lymphocyte antigen-4 (CTLA4) by monoclonal antibody is reported as an effective strategy against cancer progression in vivo and chimeric antigen receptor (CAR) modified T-cells are known to express a stronger anti-tumour activity. CTLA4 and CAR are, therefore, beneficial in cancer immunotherapy; however, in clinical settings, both are expensive and cause adverse side effects. Nanomaterials have augmented advantages in cancer immunotherapy, besides their utility in effective delivery and diagnostics. In particular, materials based on lipids, polymers, and metals have been sought-after for delivery technologies. Moreover, the surface of nanomaterials can be engineered using ligands, antigens, and antibodies to target immune cells. In this sense, checkpoint inhibitors, cytokines, agonistic antibodies, surface receptors, and engineered T-cells are promising to regulate the immune system against tumours. Therefore, emerging nanomaterials that can be used for the treatment of cancer is the prime focus of this review. The correlation of mode of administration and biodistribution of various nanomaterials is reviewed here. Besides, the acute and chronic side effects and outcome of clinical trials in the context of cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Pandian Sureshbabu Ram Kumar
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Clayton Fernando Rencilin
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Krishnan Sundar
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| |
Collapse
|
116
|
Seyfoori A, Shokrollahi Barough M, Mokarram P, Ahmadi M, Mehrbod P, Sheidary A, Madrakian T, Kiumarsi M, Walsh T, McAlinden KD, Ghosh CC, Sharma P, Zeki AA, Ghavami S, Akbari M. Emerging Advances of Nanotechnology in Drug and Vaccine Delivery against Viral Associated Respiratory Infectious Diseases (VARID). Int J Mol Sci 2021; 22:6937. [PMID: 34203268 PMCID: PMC8269337 DOI: 10.3390/ijms22136937] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/19/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022] Open
Abstract
Viral-associated respiratory infectious diseases are one of the most prominent subsets of respiratory failures, known as viral respiratory infections (VRI). VRIs are proceeded by an infection caused by viruses infecting the respiratory system. For the past 100 years, viral associated respiratory epidemics have been the most common cause of infectious disease worldwide. Due to several drawbacks of the current anti-viral treatments, such as drug resistance generation and non-targeting of viral proteins, the development of novel nanotherapeutic or nano-vaccine strategies can be considered essential. Due to their specific physical and biological properties, nanoparticles hold promising opportunities for both anti-viral treatments and vaccines against viral infections. Besides the specific physiological properties of the respiratory system, there is a significant demand for utilizing nano-designs in the production of vaccines or antiviral agents for airway-localized administration. SARS-CoV-2, as an immediate example of respiratory viruses, is an enveloped, positive-sense, single-stranded RNA virus belonging to the coronaviridae family. COVID-19 can lead to acute respiratory distress syndrome, similarly to other members of the coronaviridae. Hence, reviewing the current and past emerging nanotechnology-based medications on similar respiratory viral diseases can identify pathways towards generating novel SARS-CoV-2 nanotherapeutics and/or nano-vaccines.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Mahdieh Shokrollahi Barough
- Department of Immunology, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Pooneh Mokarram
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran;
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran 1316943551, Iran;
| | - Alireza Sheidary
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Tavia Walsh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
| | - Kielan D. McAlinden
- Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
| | - Chandra C. Ghosh
- Roger Williams Medical Center, Immuno-Oncology Institute (Ix2), Providence, RI 02908, USA;
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Amir A. Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, U.C. Davis Lung Center, Davis School of Medicine, University of California, Davis, CA 95817, USA;
- Veterans Affairs Medical Center, Mather, CA 95817, USA
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
117
|
Kardani K, Sadat SM, Kardani M, Bolhassani A. The next generation of HCV vaccines: a focus on novel adjuvant development. Expert Rev Vaccines 2021; 20:839-855. [PMID: 34114513 DOI: 10.1080/14760584.2021.1941895] [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] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Considerable efforts have been made to treat and prevent acute and chronic infections caused by the hepatitis C virus (HCV). Current treatments are unable to protect people from reinfection. Hence, there is a need for development of both preventive and therapeutic HCV vaccines. Many vaccine candidates are in development to fight against HCV, but their efficacy has so far proven limited partly due to low immunogenicity. AREAS COVERED We explore development of novel and powerful adjuvants to achieve an effective HCV vaccine. The basis for developing strong adjuvants is to understand the innate immunity pathway, which subsequently stimulates humoral and cellular immune responses. We have also investigated immunogenicity of developed adjuvants that have been used in recent studies available in online databases such as PubMed, PMC, ScienceDirect, Google Scholar, etc. EXPERT OPINION Adjuvants are used as a part of vaccine formulation to boost vaccine immunogenicity and antigen delivery. Several FDA-approved adjuvants are used in licensed human vaccines. Unfortunately, no adjuvant has yet been proven to boost HCV immune responses to the extent needed for an effective vaccine. One of the promising approaches for developing an effective adjuvant is the combination of various adjuvants to trigger several innate immune responses, leading to activation of adaptive immunity.[Figure: see text].
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Mona Kardani
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
118
|
Mardanova ES, Ravin NV. Transient expression of recombinant proteins in plants using potato virus X based vectors. Methods Enzymol 2021; 660:205-222. [PMID: 34742389 DOI: 10.1016/bs.mie.2021.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Plants become a promising biofactory for the large-scale production of recombinant proteins due to low cost, scalability, and safety. Agroinfiltration of plant leaves with a plant viral vector carrying a gene of interest is a rapid and efficient method for protein production in plants. Currently this method is in use for producing a wide range of proteins for multiple applications, including vaccine antigens, antibodies, and protein nanoparticles such as virus-like particles. A number of pharmaceutical proteins produced by transient expression are currently in clinical development. Here, we describe potato virus X based vector pEff-GFP enabling fast and high-level expression of recombinant proteins in Nicotiana benthamiana plants. The pEff vector provides green fluorescent protein expression levels of up to 30% of total soluble protein (about 1mg per g of fresh leaf tissue) and was successfully applied for the production of the immunogens of potential clinical interest.
Collapse
Affiliation(s)
- Eugenia S Mardanova
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Nikolai V Ravin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
119
|
Wang Z, Zhou C, Gao F, Zhu Q, Jiang Y, Ma X, Hu Y, Shi L, Wang X, Zhang C, Liu B, Shen L, Mao Q, Liu G. Preclinical evaluation of recombinant HFMD vaccine based on enterovirus 71 (EV71) virus-like particles (VLP): Immunogenicity, efficacy and toxicology. Vaccine 2021; 39:4296-4305. [PMID: 34167837 DOI: 10.1016/j.vaccine.2021.06.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/09/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Enterovirus 71 (EV71) is one of the major causative agents for hand, foot and mouth disease (HFMD) in children. Currently, three inactivated EV71 vaccines have been approved by Chinese government. We previously demonstrated that recombinant EV71 virus-like particles (VLP) produced in Pichia pastoris can be produced at a high yield with a simple manufacturing process, and the candidate vaccine elicited protective humoral immune responses in mice. In present study, the nonclinical immunogenicity, efficacy and toxicity of the EV71 vaccine was comprehensively evaluated in rodents and non-human primates. The immunogenicity assessment showed that EV71 VLPs vaccine elicited high and persistent neutralizing antibody responses, which could be comparable with a licensed inactivated vaccine in animals. The immune sera of vaccinated mice also exhibited cross-neutralization activities to the heterologous subtypes of EV71. Both passive and maternal antigen specific antibodies protected the neonatal mice against the lethal EV71 challenge. Furthermore, nonclinical safety assessment of EV71 VLP vaccine showed no signs of systemic toxicity in animals. Therefore, the excellent immunogenicity, efficacy and toxicology data supported further evaluation of the VLP-based EV71 vaccine in humans.
Collapse
Affiliation(s)
- Ziyan Wang
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| | | | - Fan Gao
- National Institutes for Food and Drug Control (NIFDC), Beijing, China.
| | - Qianjun Zhu
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| | | | - Xinxing Ma
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| | - Yalin Hu
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| | - Likang Shi
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| | | | - Chao Zhang
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| | - Baofeng Liu
- Shandong Xinbo Pharmaceutical R&D Co. Ltd., Dezhou, Shandong, China.
| | - Lianzhong Shen
- Shandong Xinbo Pharmaceutical R&D Co. Ltd., Dezhou, Shandong, China.
| | - Qunying Mao
- National Institutes for Food and Drug Control (NIFDC), Beijing, China.
| | - Ge Liu
- Shanghai Zerun Biotech Co., Ltd., Shanghai, China.
| |
Collapse
|
120
|
Luo J, Huo C, Qin H, Hu J, Lei L, Pan Z. Chimeric enterovirus 71 virus-like particle displaying conserved coxsackievirus A16 epitopes elicits potent immune responses and protects mice against lethal EV71 and CA16 infection. Vaccine 2021; 39:4135-4143. [PMID: 34116877 DOI: 10.1016/j.vaccine.2021.05.093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022]
Abstract
Hand-foot-and-mouth disease (HFMD) is an infectious disease of infants and young children frequently caused by the enterovirus A species, mainly enterovirus 71 (EV71) and coxsackievirus A16 (CA16). In this study, we prepared the EV71 virus-like particle (EV71-VLP) and its chimeras using recombinant baculovirus (Bac-P1-3CD) co-expressing EV71 P1 (under polyhedrin promoter) and 3CD (under CMV-IE promoter) proteins in Sf9 cells. EV71-VLP chimera ChiEV71(1E)-VLP or ChiEV71(4E)-VLP displayed single CA16 PEP71 epitope in VP1 or four conserved CA16 neutralizing epitopes (PEP71 in VP1, aa136-150 in VP2, aa176-190 in VP3 and aa48-62 in VP4) by substitution of the corresponding regions of EV71 structure proteins, respectively. In mice, EV71-VLP and its chimeras elicited similar EV71-specific IgG and neutralizing antibody (NAb) titers compared to inactivated EV71. Expectedly, vaccination of ChiEV71(1E)-VLP or ChiEV71(4E)-VLP resulted in significantly increased CA16-specific IgG and NAb production and improved cross-protection against CA16 infection compared to EV71-VLP. Interestingly, the VLPs induced potent cellular immune responses and significantly decreased Th2 type (IL-4 and IL-10) cytokines secretion in the splenocytes of immunized mice compared to inactivated EV71 or inactivated CA16. Neonatal mice born to dams immunized with the chimeric VLPs or neonatal mice passively transferred with sera of immunized mice were completely protected from lethal EV71 challenge and partially protected from lethal CA16 infection. Our study provides a novel bivalent or multivalent vaccine strategy to prevent EV71 and related-enterovirus infections.
Collapse
Affiliation(s)
- Jin Luo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chunling Huo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huan Qin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Junhong Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lei Lei
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
121
|
Deletion of the SARS-CoV-2 Spike Cytoplasmic Tail Increases Infectivity in Pseudovirus Neutralization Assays. J Virol 2021; 95:JVI.00044-21. [PMID: 33727331 PMCID: PMC8139703 DOI: 10.1128/jvi.00044-21] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudotyped viruses are valuable tools for studying virulent or lethal viral pathogens that need to be handled in biosafety level 3 (BSL-3) or higher facilities. With the explosive spread of the coronavirus disease 2019 (COVID-19) pandemic, the establishment of a BSL-2 adapted SARS-CoV-2 pseudovirus neutralization assay is needed to facilitate the development of countermeasures. Here we describe an approach to generate a single-round lentiviral vector-based SARS-CoV-2 pseudovirus, which produced a signal more than 2 logs above background. Specifically, a SARS-CoV-2 spike variant with a cytoplasmic tail deletion of 13 amino acids, termed SΔCT13, conferred enhanced spike incorporation into pseudovirions and increased viral entry into cells as compared with full-length spike (S). We further compared S and SΔCT13 in terms of their sensitivity to vaccine sera, purified convalescent IgG, hACE2-mIgG, and the virus entry inhibitor BafA1. We developed a SΔCT13-based pseudovirus neutralization assay and defined key assay characteristics, including linearity, limit of detection, and intra- and intermediate-assay precision. Our data demonstrate that the SΔCT13-based pseudovirus shows enhanced infectivity in target cells, which will facilitate the assessment of humoral immunity to SARS-CoV-2 infection, antibody therapeutics, and vaccination. This pseudovirus neutralization assay can also be readily adapted to SARS-CoV-2 variants that emerge.IMPORTANCESARS-CoV-2 is the etiologic agent of the COVID-19 pandemic. The development of a high throughput pseudovirus neutralization assay is critical for the development of vaccines and immune-based therapeutics. In this study, we show that deletion of the cytoplasmic tail of the SARS-CoV-2 spike leads to pseudoviruses with enhanced infectivity. This SΔCT13-based pseudovirus neutralization assay should be broadly useful for the field.
Collapse
|
122
|
Passive Immunity and Antibody Response Induced by Toxoplasma gondii VLP Immunization. Vaccines (Basel) 2021; 9:vaccines9050425. [PMID: 33922808 PMCID: PMC8146287 DOI: 10.3390/vaccines9050425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/29/2022] Open
Abstract
Passive immunity can provide immediate protection against infectious pathogens. To date, only a few studies have investigated the effect of passive immunization against Toxoplasma gondii, and the use of immune sera acquired from VLP-vaccinated mice for passive immunity assessment remains unreported. In this study, immune sera were produced by a single immunization with virus-like particles (VLPs) expressing the inner membrane complex (IMC), rhoptry protein 18 (ROP18), and microneme protein 8 (MIC8) of Toxoplasma gondii, with or without a CpG-ODN adjuvant. The passive immunization of immune sera conferred protection in mice, as indicated by their potent parasite-specific antibody response, lessened brain cyst counts, lower bodyweight loss, and enhanced survival. In order to confirm that the immune sera of the VLP-immunized mice were truly protective, the antibody responses and other immunological parameters were measured in the VLP-immunized mice. We found that VLP immunization induced higher levels of parasite-specific IgG, IgG subclass, and IgM antibody responses in the sera and intestines than in the controls. Enhanced Th1 and Th2-associated cytokines in the spleen, diminished brain cyst counts, and lessened body weight loss were found following T. gondii ME49 challenge infection. These results suggest that passive immunization with the immune sera acquired from VLP-vaccinated mice can confer adequate protection against T. gondii infection.
Collapse
|
123
|
Chu KB, Quan FS. Advances in Toxoplasma gondii Vaccines: Current Strategies and Challenges for Vaccine Development. Vaccines (Basel) 2021; 9:vaccines9050413. [PMID: 33919060 PMCID: PMC8143161 DOI: 10.3390/vaccines9050413] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Toxoplasmosis, caused by the apicomplexan parasite Toxoplasma gondii, is one of the most damaging parasite-borne zoonotic diseases of global importance. While approximately one-third of the entire world’s population is estimated to be infected with T. gondii, an effective vaccine for human use remains unavailable. Global efforts in pursuit of developing a T. gondii vaccine have been ongoing for decades, and novel innovative approaches have been introduced to aid this process. A wide array of vaccination strategies have been conducted to date including, but not limited to, nucleic acids, protein subunits, attenuated vaccines, and nanoparticles, which have been assessed in rodents with promising results. Yet, translation of these in vivo results into clinical studies remains a major obstacle that needs to be overcome. In this review, we will aim to summarize the current advances in T. gondii vaccine strategies and address the challenges hindering vaccine development.
Collapse
Affiliation(s)
- Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
124
|
Jin Y, Zhuang Y, Dong X, Liu M. Development of CpG oligodeoxynucleotide TLR9 agonists in anti-cancer therapy. Expert Rev Anticancer Ther 2021; 21:841-851. [PMID: 33831324 DOI: 10.1080/14737140.2021.1915136] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Toll-like receptor-9(TLR9) can recognize the foreign unmethylated CpG DNA, and thus intrigue a strong Th1 response which plays a crucial role in the innate and adaptive immune responses. To date, CpG oligodeoxynucleotide (ODN)-based TLR9 agonists have undergone four generations. Each generations' breakthroughs in immune activation, safety profiles and pharmacokinetic properties were confirmed by both preclinical and clinical studies. AREAS COVERED We reviewed the development and major clinical trials of TLR9 agonists and summarized the optimization strategies of each generation. The applications, limitations and prospects of TLR9 agonists in cancer immunotherapy are also discussed. EXPERT OPINION Clinical trials of CpG ODN TLR9 agonists as a single agent demonstrated insufficient efficacy to reverse the immunosuppressive status of majority of patients with high tumor burden. Therefore, more efforts are now been carried out in combination with chemotherapy, radiotherapy and immunotherapy maintenance therapy as well as vaccine adjuvant. Importantly, the synergistic and complementary effect of TLR9 agonists and tumor immune checkpoint inhibitor therapy is expected to exert greater potential. On the other hand, the double-edged sword effect of TLR9 activation in tumor and toxic effect reported in combination therapies should be noted and further studies required.
Collapse
Affiliation(s)
- Yizhen Jin
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Yuxin Zhuang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Mei Liu
- Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, P.R. China
| |
Collapse
|
125
|
Derakhshan MA, Amani A, Faridi-Majidi R. State-of-the-Art of Nanodiagnostics and Nanotherapeutics against SARS-CoV-2. ACS APPLIED MATERIALS & INTERFACES 2021; 13:14816-14843. [PMID: 33779135 PMCID: PMC8028022 DOI: 10.1021/acsami.0c22381] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/17/2021] [Indexed: 05/02/2023]
Abstract
The pandemic outbreak of SARS-CoV-2, with millions of infected patients worldwide, has severely challenged all aspects of public health. In this regard, early and rapid detection of infected cases and providing effective therapeutics against the virus are in urgent demand. Along with conventional clinical protocols, nanomaterial-based diagnostics and therapeutics hold a great potential against coronavirus disease 2019 (COVID-19). Indeed, nanoparticles with their outstanding characteristics would render additional advantages to the current approaches for rapid and accurate diagnosis and also developing prophylactic vaccines or antiviral therapeutics. In this review, besides presenting an overview of the coronaviruses and SARS-CoV-2, we discuss the introduced nanomaterial-based detection assays and devices and also antiviral formulations and vaccines for coronaviruses.
Collapse
Affiliation(s)
- Mohammad Ali Derakhshan
- Department
of Medical Nanotechnology, School of Advanced Medical Sciences and
Technologies, Shiraz University of Medical
Sciences, Shiraz, Iran
- Nanomedicine
and Nanobiology Research Center, Shiraz
University of Medical Sciences, Shiraz Iran
| | - Amir Amani
- Natural
Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Faridi-Majidi
- Department
of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
126
|
Lipid nanovesicles for biomedical applications: 'What is in a name'? Prog Lipid Res 2021; 82:101096. [PMID: 33831455 DOI: 10.1016/j.plipres.2021.101096] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/28/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022]
Abstract
Vesicles, generally defined as self-assembled structures formed by single or multiple concentric bilayers that surround an aqueous core, have been widely used for biomedical applications. They can either occur naturally (e.g. exosomes) or be produced artificially and range from the micrometric scale to the nanoscale. One the most well-known vesicle is the liposome, largely employed as a drug delivery nanocarrier. Liposomes have been modified along the years to improve physicochemical and biological features, resulting in long-circulating, ligand-targeted and stimuli-responsive liposomes, among others. In this process, new nomenclatures were reported in an extensive literature. In many instances, the new names suggest the emergence of a new nanocarrier, which have caused confusion as to whether the vesicles are indeed new entities or could simply be considered modified liposomes. Herein, we discussed the extensive nomenclature of vesicles based on the suffix "some" that are employed for drug delivery and composed of various types and proportions of lipids and others amphiphilic compounds. New names have most often been selected based on changes of vesicle lipid composition, but the payload, structural complexity (e.g. multicompartment) and new/improved proprieties (e.g. elasticity) have also inspired new vesicle names. Based on this discussion, we suggested a rational classification for vesicles.
Collapse
|
127
|
Abstract
To this day, the coronavirus disease 2019 (COVID-19) pandemic has not shown signs of abating. Moreover, the virus responsible for the pandemic, severe acute respiratory syndrome coronavirus 2, has evolved into three different variants. This phenomenon highlights an even greater need to develop drugs and vaccines to control the rate of infection and spread of the disease. As of July 7, 2020, at least 160 vaccine candidates, 21 of which have entered the clinical trial phase, have been developed. This article describes the latest advances in development, reliable platforms, strategies used, and challenges that remain in developing COVID-19 vaccines.
Collapse
Affiliation(s)
- Riyadi Sumirtanurdin
- Pharmacist Profession Education, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Biotechnology Pharmacy Laboratory, Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Melisa Intan Barliana
- Biotechnology Pharmacy Laboratory, Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
128
|
Ho W, Gao M, Li F, Li Z, Zhang X, Xu X. Next-Generation Vaccines: Nanoparticle-Mediated DNA and mRNA Delivery. Adv Healthc Mater 2021; 10:e2001812. [PMID: 33458958 PMCID: PMC7995055 DOI: 10.1002/adhm.202001812] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/06/2020] [Indexed: 01/07/2023]
Abstract
Nucleic acid vaccines are a method of immunization aiming to elicit immune responses akin to live attenuated vaccines. In this method, DNA or messenger RNA (mRNA) sequences are delivered to the body to generate proteins, which mimic disease antigens to stimulate the immune response. Advantages of nucleic acid vaccines include stimulation of both cell-mediated and humoral immunity, ease of design, rapid adaptability to changing pathogen strains, and customizable multiantigen vaccines. To combat the SARS-CoV-2 pandemic, and many other diseases, nucleic acid vaccines appear to be a promising method. However, aid is needed in delivering the fragile DNA/mRNA payload. Many delivery strategies have been developed to elicit effective immune stimulation, yet no nucleic acid vaccine has been FDA-approved for human use. Nanoparticles (NPs) are one of the top candidates to mediate successful DNA/mRNA vaccine delivery due to their unique properties, including unlimited possibilities for formulations, protective capacity, simultaneous loading, and delivery potential of multiple DNA/mRNA vaccines. This review will summarize the many varieties of novel NP formulations for DNA and mRNA vaccine delivery as well as give the reader a brief synopsis of NP vaccine clinical trials. Finally, the future perspectives and challenges for NP-mediated nucleic acid vaccines will be explored.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Mingzhu Gao
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Fengqiao Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Zhongyu Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
- Department of Biomedical EngineeringNew Jersey Institute of Technology323 Dr Martin Luther King Jr BlvdNewarkNJ07102USA
| |
Collapse
|
129
|
Machhi J, Shahjin F, Das S, Patel M, Abdelmoaty MM, Cohen JD, Singh PA, Baldi A, Bajwa N, Kumar R, Vora LK, Patel TA, Oleynikov MD, Soni D, Yeapuri P, Mukadam I, Chakraborty R, Saksena CG, Herskovitz J, Hasan M, Oupicky D, Das S, Donnelly RF, Hettie KS, Chang L, Gendelman HE, Kevadiya BD. Nanocarrier vaccines for SARS-CoV-2. Adv Drug Deliv Rev 2021; 171:215-239. [PMID: 33428995 PMCID: PMC7794055 DOI: 10.1016/j.addr.2021.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/18/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Srijanee Das
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Preet Amol Singh
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Ashish Baldi
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Neha Bajwa
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lalit K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Tapan A Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology (CHARUSAT), Changa, Anand 388421, Gujarat, India
| | - Maxim D Oleynikov
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Dhruvkumar Soni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Rajashree Chakraborty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Caroline G Saksena
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Jonathan Herskovitz
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Suvarthi Das
- Department of Medicine, Stanford Medical School, Stanford University, Palo Alto, CA 94304, USA
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Department of Otolaryngology - Head & Neck Surgery, Stanford University, Palo Alto, CA 94304, USA
| | - Linda Chang
- Departments of Diagnostic Radiology & Nuclear Medicine, and Neurology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| |
Collapse
|
130
|
Marin A, Chowdhury A, Valencia SM, Zacharia A, Kirnbauer R, Roden RBS, Pinto LA, Shoemaker RH, Marshall JD, Andrianov AK. Next generation polyphosphazene immunoadjuvant: Synthesis, self-assembly and in vivo potency with human papillomavirus VLPs-based vaccine. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 33:102359. [PMID: 33476764 PMCID: PMC8184581 DOI: 10.1016/j.nano.2021.102359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/15/2020] [Accepted: 01/04/2021] [Indexed: 10/22/2022]
Abstract
Poly[di(carboxylatomethylphenoxy)phosphazene] (PCMP), a new member of polyphosphazene immunoadjuvant family, is synthesized. In vitro assessment of a new macromolecule revealed hydrolytic degradation profile and immunostimulatory activity comparable to its clinical stage homologue PCPP; however, PCMP was characterized by a beneficial reduced sensitivity to the ionic environment. In vivo evaluation of PCMP potency was conducted with human papillomavirus (HPV) virus-like particles (VLPs) based RG1-VLPs vaccine. In contrast with previously reported self-assembly of polyphosphazene adjuvants with proteins, which typically results in the formation of complexes with multimeric display of antigens, PCMP surface modified VLPs in a composition dependent pattern, which at a high polymer-to VLPs ratio led to stabilization of antigenic particles. Immunization experiments in mice demonstrated that PCMP adjuvanted RG1-VLPs vaccine induced potent humoral immune responses, in particular, on the level of highly desirable protective cross-neutralizing antibodies, and outperformed PCPP and Alhydrogel adjuvanted formulations.
Collapse
Affiliation(s)
- Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States
| | - Ananda Chowdhury
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States
| | - Sarah M Valencia
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Athina Zacharia
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Reinhard Kirnbauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, , United States
| | - Ligia A Pinto
- HPV Immunology laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, United States
| | - Jason D Marshall
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States.
| |
Collapse
|
131
|
Müller C, Hrynkiewicz R, Bębnowska D, Maldonado J, Baratelli M, Köllner B, Niedźwiedzka-Rystwej P. Immunity against Lagovirus europaeus and the Impact of the Immunological Studies on Vaccination. Vaccines (Basel) 2021; 9:vaccines9030255. [PMID: 33805607 PMCID: PMC8002203 DOI: 10.3390/vaccines9030255] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
In the early 1980s, a highly contagious viral hemorrhagic fever in rabbits (Oryctolagus cuniculus) emerged, causing a very high rate of mortality in these animals. Since the initial occurrence of the rabbit hemorrhagic disease virus (RHDV), several hundred million rabbits have died after infection. The emergence of genetically-different virus variants (RHDV GI.1 and GI.2) indicated the very high variability of RHDV. Moreover, with these variants, the host range broadened to hare species (Lepus). The circulation of RHDV genotypes displays different virulences and a limited induction of cross-protective immunity. Interestingly, juvenile rabbits (<9 weeks of age) with an immature immune system display a general resistance to RHDV GI.1, and a limited resistance to RHDV GI.2 strains, whereas less than 3% of adult rabbits survive an infection by either RHDV GI.1. or GI.2. Several not-yet fully understood phenomena characterize the RHD. A very low infection dose followed by an extremely rapid viral replication could be simplified to the induction of a disseminated intravascular coagulopathy (DIC), a severe loss of lymphocytes—especially T-cells—and death within 36 to 72 h post infection. On the other hand, in animals surviving the infection or after vaccination, very high titers of RHDV-neutralizing antibodies were induced. Several studies have been conducted in order to deepen the knowledge about the virus’ genetics, epidemiology, RHDV-induced pathology, and the anti-RHDV immune responses of rabbits in order to understand the phenomenon of the juvenile resistance to this virus. Moreover, several approaches have been used to produce efficient vaccines in order to prevent an infection with RHDV. In this review, we discuss the current knowledge about anti-RHDV resistance and immunity, RHDV vaccination, and the further need to establish rationally-based RHDV vaccines.
Collapse
Affiliation(s)
- Claudia Müller
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, 17493 Greifswald-Insel Riems, Germany;
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (R.H.); (D.B.)
| | - Dominika Bębnowska
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (R.H.); (D.B.)
| | | | | | - Bernd Köllner
- Institute of Immunology, Friedrich-Loeffler-Institute, 17493 Greifswald-Insel Riems, Germany
- Correspondence: (B.K.); (P.N.-R.)
| | - Paulina Niedźwiedzka-Rystwej
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (R.H.); (D.B.)
- Correspondence: (B.K.); (P.N.-R.)
| |
Collapse
|
132
|
Kumari S, Chatterjee K. Biomaterials-based formulations and surfaces to combat viral infectious diseases. APL Bioeng 2021; 5:011503. [PMID: 33598595 PMCID: PMC7881627 DOI: 10.1063/5.0029486] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Rapidly growing viral infections are potent risks to public health worldwide. Accessible virus-specific antiviral vaccines and drugs are therapeutically inert to emerging viruses, such as Zika, Ebola, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, discovering ways to prevent and control viral infections is among the foremost medical challenge of our time. Recently, innovative technologies are emerging that involve the development of new biomaterial-based formulations and surfaces endowed with broad-spectrum antiviral properties. Here, we review emerging biomaterials technologies for controlling viral infections. Relevant advances in biomaterials employed with nanotechnology to inactivate viruses or to inhibit virus replication and further their translation in safe and effective antiviral formulations in clinical trials are discussed. We have included antiviral approaches based on both organic and inorganic nanoparticles (NPs), which offer many advantages over molecular medicine. An insight into the development of immunomodulatory scaffolds in designing new platforms for personalized vaccines is also considered. Substantial research on natural products and herbal medicines and their potential in novel antiviral drugs are discussed. Furthermore, to control contagious viral infections, i.e., to reduce the viral load on surfaces, current strategies focusing on biomimetic anti-adhesive surfaces through nanostructured topography and hydrophobic surface modification techniques are introduced. Biomaterial surfaces functionalized with antimicrobial polymers and nanoparticles against viral infections are also discussed. We recognize the importance of research on antiviral biomaterials and present potential strategies for future directions in applying these biomaterial-based approaches to control viral infections and SARS-CoV-2.
Collapse
Affiliation(s)
- Sushma Kumari
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
133
|
Hepatitis B core-based virus-like particles: A platform for vaccine development in plants. ACTA ACUST UNITED AC 2021; 29:e00605. [PMID: 33732633 PMCID: PMC7937989 DOI: 10.1016/j.btre.2021.e00605] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023]
Abstract
Virus-like particles (VLPs) are a class of structures formed by the self-assembly of viral capsid protein subunits and contain no infective viral genetic material. The Hepatitis B core (HBc) antigen is capable of assembling into VLPs that can elicit strong immune responses and has been licensed as a commercial vaccine against Hepatitis B. The HBc VLPs have also been employed as a platform for the presentation of foreign epitopes to the immune system and have been used to develop vaccines against, for example, influenza A and Foot-and-mouth disease. Plant expression systems are rapid, scalable and safe, and are capable of providing correct post-translational modifications and reducing upstream production costs. The production of HBc-based virus-like particles in plants would thus greatly increase the efficiency of vaccine production. This review investigates the application of plant-based HBc VLP as a platform for vaccine production.
Collapse
|
134
|
Li W, Jing Z, Wang S, Li Q, Xing Y, Shi H, Li S, Hong Z. P22 virus-like particles as an effective antigen delivery nanoplatform for cancer immunotherapy. Biomaterials 2021; 271:120726. [PMID: 33636548 DOI: 10.1016/j.biomaterials.2021.120726] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
As a new strategy for cancer immunotherapy, therapeutic cancer vaccines have been greatly improved in recent years. However, addressing the needs to quickly and efficiently elicit a high-intensity immune response against neoantigen peptides, especially to induce an effective cytotoxic lymphocyte (CTL) reaction, remain challenges in this field. In this study, virus-like particles (VLPs) derived from the phage P22 were adopted to load peptide antigens on the surface, to test whether VLP technology can be used as a platform for efficient peptide antigen delivery by therapeutic cancer vaccines. The B and T epitopes (OVAB peptide and OVAT peptide) of ovalbumin (OVA) were used here as model antigens and fused individually at the C terminus of the coat protein (CP), which allowed display on the surface of P22 particles to form two types of vaccine particles (VLP-OVAB and VLP-OVAT). Subsequent experiments showed that VLP-OVAB induced an antibody titer against the peptide antigen as high as 5.0 × 105 and that VLP-OVAT induced highly effective cross-presentation and then strongly activated a T epitope-specific CTL response. Mouse tumor model experiments showed that VLP-OVAT could significantly inhibit tumor growth by increasing the proportions of CD4+ T cells, CD8+ T cells and effector memory T cells (TEM cells) and lowering the proportion of myeloid-derived suppressor cells (MDSCs) among tumor-infiltrating lymphocytes and splenocytes. Compared with other chemically synthesized nanomaterials, VLPs have obvious advantages as vaccine carriers due to their clear chemical composition, fixed spatial structure, excellent biocompatibility, and relatively high potential for clinical translation. Therefore, this platform may lay a solid foundation for the design and preparation of personalized therapeutic vaccines based on neoantigen peptides.
Collapse
Affiliation(s)
- Wenjing Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Zhe Jing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shuqing Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Qiyu Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Yutong Xing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Haobo Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shuang Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
135
|
Zhang Y, Yuan Y, Zhang LH, Zhu D, Wang L, Wei LP, Fan WS, Zhao CR, Su YJ, Liao JQ, Yong L, Wei TC, Wei P, Mo ML. Construction and Immunogenicity Comparison of Three Virus-Like Particles Carrying Different Combinations of Structural Proteins of Avian Coronavirus Infectious Bronchitis Virus. Vaccines (Basel) 2021; 9:vaccines9020146. [PMID: 33670249 PMCID: PMC7918244 DOI: 10.3390/vaccines9020146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/27/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Infectious bronchitis virus (IBV) poses massive economic losses in the global poultry industry. Here, we firstly report the construction and immunogenicity comparison of virus-like particles (VLPs) carrying the S, M and E proteins (SME-VLPs); VLPs carrying the S and M proteins (SM-VLPs); and VLPs carrying the M and E proteins (ME-VLPs) from the dominant serotype representative strain GX-YL5 in China. The neutralizing antibody response induced by the SME-VLPs was similar to that induced by the inactivated oil vaccine (OEV) of GX-YL5, and higher than those induced by the SM-VLPs, ME-VLPs and commercial live vaccine H120. More importantly, the SME-VLPs elicited higher percentages of CD4+ and CD8+ T lymphocytes than the SM-VLPs, ME-VLPs and OEV of GX-YL5. Compared with the OEV of GX-YL5, higher levels of IL-4 and IFN-γ were also induced by the SME-VLPs. Moreover, the mucosal immune response (sIgA) induced by the SME-VLPs in the tear and oral swabs was comparable to that induced by the H120 vaccine and higher than that induced by the OEV of GX-YL5. In the challenge experiment, the SME-VLPs resulted in significantly lower viral RNA levels in the trachea and higher protection scores than the OEV of GX-YL5 and H120 vaccines, and induced comparable viral RNA levels in the kidneys, and tear and oral swabs to the OEV of GX-YL5. In summary, among the three VLPs, the SME-VLPs carrying the S, M and E proteins of IBV could stimulate the strongest humoral, cellular and mucosal immune responses and provide effective protection, indicating that it would be an attractive vaccine candidate for IB.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Ping Wei
- Correspondence: (P.W.); (M.-L.M.); Tel.: +86-771-323-5638 (P.W.); +86-771-323-5635 (M.-L.M.)
| | - Mei-Lan Mo
- Correspondence: (P.W.); (M.-L.M.); Tel.: +86-771-323-5638 (P.W.); +86-771-323-5635 (M.-L.M.)
| |
Collapse
|
136
|
Immunogenicity and Protective Activity of Pigeon Circovirus Recombinant Capsid Protein Virus-Like Particles (PiCV rCap-VLPs) in Pigeons ( Columba livia) Experimentally Infected with PiCV. Vaccines (Basel) 2021; 9:vaccines9020098. [PMID: 33525416 PMCID: PMC7912323 DOI: 10.3390/vaccines9020098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/24/2021] [Accepted: 01/24/2021] [Indexed: 12/11/2022] Open
Abstract
Pigeon circovirus (PiCV) is the most recurrent virus diagnosed in pigeons and is among the major causative agents of young pigeon disease syndrome (YPDS). Due to the lack of an established laboratory protocol for PiCV cultivation, development of prophylaxis is hampered. Alternatively, virus-like particles (VLPs), which closely resemble native viruses but lack the viral genetic material, can be generated using a wide range of expression systems and are shown to have strong immunogenicity. Therefore, the use of VLPs provides a promising prospect for vaccine development. In this study, transfected human embryonic kidney (HEK-293) cells, a mammalian expression system, were used to express the PiCV capsid protein (Cap), which is a major component of PiCV and believed to contain antibody epitopes, to obtain self-assembled VLPs. The VLPs were observed to have a spherical morphology with diameters ranging from 12 to 26 nm. Subcutaneous immunization of pigeons with 100 µg PiCV rCap-VLPs supplemented with water-in-oil-in-water (W/O/W) adjuvant induced specific antibodies against PiCV. Observations of the cytokine expression and T-cell proliferation levels in spleen samples showed significantly higher T-cell proliferation and IFN- γ expression in pigeons immunized with VLPs compared to the controls (p < 0.05). Experimentally infected pigeons that were vaccinated with VLPs also showed no detectable viral titer. The results of the current study demonstrated the potential use of PiCV rCap-VLPs as an effective vaccine candidate against PiCV.
Collapse
|
137
|
The Use of Nanobiotechnology in Immunology and Vaccination. Vaccines (Basel) 2021; 9:vaccines9020074. [PMID: 33494441 PMCID: PMC7910821 DOI: 10.3390/vaccines9020074] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 01/07/2023] Open
Abstract
Nanotechnology uses the unique properties of nanostructures with a size of 1 to 200 nanometers. Different nanoparticles have shown great promise for the production of new vaccines and drugs. Nanostructures can be used to deliver immunological compounds more effectively than microstructures to target sites. Different nanostructures can be applied to form a new generation of vaccines, adjuvants, and immune system drugs. The goal of nanotechnology is to better respond to a wide range of infectious and non-infectious diseases.
Collapse
|
138
|
Hwang BJ, Jang Y, Kwon SB, Yu JE, Lim J, Roh YH, Seong BL. RNA-assisted self-assembly of monomeric antigens into virus-like particles as a recombinant vaccine platform. Biomaterials 2021; 269:120650. [PMID: 33465537 DOI: 10.1016/j.biomaterials.2021.120650] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022]
Abstract
Representing highly ordered repetitive structures of antigen macromolecular assemblies, virus-like particles (VLPs) serve as a high-priority vaccine platform against emerging viral infections, as alternatives to traditional cell culture-based vaccines. RNAs can function as chaperones (Chaperna) and are extremely effective in promoting protein folding. Beyond their canonical function as translational adaptors, tRNAs may moonlight as chaperones for the kinetic control of macromolecular antigen assembly. Capitalizing on genomic RNA co-assembly in infectious virions, we present the first report of a biomimetic assembly of viral capsids that was assisted by non-viral host RNAs into genome-free, non-infectious empty particles. Here, we demonstrate the assembly of bacterially-produced soluble norovirus VP1 forming VLPs (n = 180) in vitro. A tRNA-interacting domain (tRID) was genetically fused with the VP1 capsid protein, as a tRNA docking tag, in the bacterial host to transduce chaperna function for de novo viral antigen folding. tRID/tRNA removal prompted the in vitro assembly of monomeric antigens into highly ordered repetitive structures that elicited robust protective immune responses after immunization. The chaperna-based assembly of monomeric antigens will impact the development and deployment of VLP vaccines for emerging and re-emerging viral infections.
Collapse
Affiliation(s)
- Beom Jeung Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Vaccine Innovative Technology Alliance-Korea, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yohan Jang
- Department of Biological Sciences and Biotechnology Major in Bio-Vaccine Engineering, Andong National University, Andong, South Korea
| | - Soon Bin Kwon
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Ji Eun Yu
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jongkwan Lim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Hoon Roh
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Baik L Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Vaccine Innovative Technology Alliance-Korea, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
139
|
Fearon SH, Dennis SJ, Hitzeroth II, Rybicki EP, Meyers AE. Humoral and cell-mediated immune responses to plant-produced African horse sickness virus VP7 quasi-crystals. Virus Res 2021; 294:198284. [PMID: 33421520 DOI: 10.1016/j.virusres.2020.198284] [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: 08/20/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 11/26/2022]
Abstract
African horse sickness (AHS) is a devastating viral disease affecting equines and has resulted in many disastrous epizootics. To date, no successful therapeutic treatment exists for AHS, and commercially used live-attenuated vaccines have various undesirable side effects. Previous studies have shown that mice inoculated with insoluble African horse sickness virus (AHSV) VP7 crystals are protected from live challenge with a lethal dose of AHSV. This study investigates the humoral and cell-mediated immune responses in guinea-pigs to a safer monovalent vaccine alternative based on AHSV-5 VP7 quasi-crystals produced in plants. Guinea-pigs received prime- and boost-inoculations of between 10 and 50 μg of purified plant-produced AHSV VP7. Western immunoblot analysis of the humoral response showed stimulation of high titres of anti-VP7 antibodies 28 days after the boost-inoculation in sera from three of the five experimental animals. In addition, RNA-seq transcriptome profiling of guinea-pig spleen-derived RNA highlighted thirty significantly (q ≤ 0.05) differentially expressed genes involved in innate and adaptive immunity. Differential expression of genes involved in Th1, Th2 and Th17 cell differentiation suggest a cell-mediated immune response to AHSV-5 VP7. Upregulation of several important cytokines and cytokine receptors were noted, including TNFSF14, CX3CR1, IFNLR1 and IL17RA. Upregulation of IL17RA suggests a Th17 response which has been reported as a key component in AHSV immunity. While further investigation is needed to validate these findings, these results suggest that AHSV-5 VP7 quasi-crystals produced in N. benthamiana are immunogenic and induce both humoral and cell-mediated responses.
Collapse
Affiliation(s)
- Shelley H Fearon
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Susan J Dennis
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Inga I Hitzeroth
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Edward P Rybicki
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town 7700, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Ann E Meyers
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town 7700, South Africa.
| |
Collapse
|
140
|
Fukase K. Glycoconjugates for Adjuvants and Self-Adjuvanting Vaccines. COMPREHENSIVE GLYCOSCIENCE 2021:166-184. [DOI: 10.1016/b978-0-12-819475-1.00099-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
141
|
Expression of the rabies virus nucleoprotein and matrix protein in a prokaryotic system at high-levels: An efficacious production method. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
142
|
Yadav T, Srivastava N, Mishra G, Dhama K, Kumar S, Puri B, Saxena SK. Recombinant vaccines for COVID-19. Hum Vaccin Immunother 2020; 16:2905-2912. [PMID: 33232211 PMCID: PMC7711739 DOI: 10.1080/21645515.2020.1820808] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
SARS-CoV-2, the causative agent of COVID-19, has imposed a major public health threat, which needs effective therapeutics and vaccination strategies. Several potential candidate vaccines being rapidly developed are in clinical evaluation. Considering the crucial role of SARS-CoV-2 spike (S) glycoprotein in virus attachment, entry, and induction of neutralizing antibodies, S protein is being widely used as a target for vaccine development. Based on advances in techniques for vaccine design, inactivated, live-vectored, nucleic acid, and recombinant COVID-19 vaccines are being developed and tested for their efficacy. Phase3 clinical trials are underway or will soon begin for several of these vaccines. Assuming that clinical efficacy is shown for one or more vaccines, safety is a major aspect to be considered before deploying such vaccines to the public. The current review focuses on the recent advances in recombinant COVID-19 vaccine research and development and associated issues.
Collapse
Affiliation(s)
- Tushar Yadav
- Department of Zoology, Jawaharlal Nehru Smriti Government Post Graduate College , Shujalpur, India
| | - Nishant Srivastava
- Department of Biotechnology, Meerut Institute of Engineering and Technology , Meerut, India
| | - Gourav Mishra
- Department of Biotechnology, Meerut Institute of Engineering and Technology , Meerut, India
| | - Kuldeep Dhama
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar , Bareilly, India
| | - Swatantra Kumar
- Centre for Advanced Research (CFAR), Faculty of Medicine, King George's Medical University (KGMU) , Lucknow, India
| | - Bipin Puri
- Centre for Advanced Research (CFAR), Faculty of Medicine, King George's Medical University (KGMU) , Lucknow, India
| | - Shailendra K Saxena
- Centre for Advanced Research (CFAR), Faculty of Medicine, King George's Medical University (KGMU) , Lucknow, India
| |
Collapse
|
143
|
Nano-based approaches in the development of antiviral agents and vaccines. Life Sci 2020; 265:118761. [PMID: 33189824 PMCID: PMC7658595 DOI: 10.1016/j.lfs.2020.118761] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022]
Abstract
Outbreaks and the rapid transmission of viruses, such as coronaviruses and influenza viruses, are serious threats to human health. A major challenge in combating infectious diseases caused by viruses is the lack of effective methods for prevention and treatment. Nanotechnology has provided a basis for the development of novel antiviral strategies. Owing to their large modifiable surfaces that can be functionalized with multiple molecules to realize sophisticated designs, nanomaterials have been developed as nanodrugs, nanocarriers, and nano-based vaccines to effectively induce sufficient immunologic memory. From this perspective, we introduce various nanomaterials with diverse antiviral mechanisms and summarize how nano-based antiviral agents protect against viral infection at the molecular, cellular, and organismal levels. We summarize the applications of nanomaterials for defense against emerging viruses by trapping and inactivating viruses and inhibiting viral entry and replication. We also discuss recent progress in nano-based vaccines with a focus on the mechanisms by which nanomaterials contribute to immunogenicity. We further describe how nanotechnology may improve vaccine efficacy by delivering large amounts of antigens to target immune cells and enhancing the immune response by mimicking viral structures and activating dendritic cells. Finally, we provide an overview of future prospects for nano-based antiviral agents and vaccines.
Collapse
|
144
|
Antigen Design for Successful Isolation of Highly Challenging Therapeutic Anti-GPCR Antibodies. Int J Mol Sci 2020; 21:ijms21218240. [PMID: 33153215 PMCID: PMC7663707 DOI: 10.3390/ijms21218240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 02/03/2023] Open
Abstract
G-protein-coupled receptors (GPCR) transmit extracellular signals into cells to regulate a variety of cellular functions and are closely related to the homeostasis of the human body and the progression of various types of diseases. Great attention has been paid to GPCRs as excellent drug targets, and there are many commercially available small-molecule chemical drugs against GPCRs. Despite this, the development of therapeutic anti-GPCR antibodies has been delayed and is challenging due to the difficulty in preparing active forms of GPCR antigens, resulting from their low cellular expression and complex structures. Here, we focus on anti-GPCR antibodies that have been approved or are subject to clinical trials and present various technologies to prepare active GPCR antigens that enable the isolation of therapeutic antibodies to proceed toward clinical validation.
Collapse
|
145
|
Zou Y, Li S, Ngai T, Zhang S, Ma G, Wu J. Green preparation of hydrogel particles-in-emulsions for simultaneous enhancement of humoral and cell-mediated immunity. Eng Life Sci 2020; 20:514-524. [PMID: 33204238 PMCID: PMC7645649 DOI: 10.1002/elsc.202000011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/07/2020] [Accepted: 09/09/2020] [Indexed: 01/07/2023] Open
Abstract
Emulsions are one of the most often used vaccine adjuvant formulations. Although they promote high humoral immunity, the induced cellular immunity is often poor, which restrict their application. To enhance the cellular immunity, some researchers have prepared mixed formulations by adding particles into the aqueous phase of emulsions. However, the particle preparation process usually involves the addition and removal of organic reagents, which is environmentally unfriendly and cumbersome. Moreover, the obtained vaccine adjuvant only induces limited cell-mediated immunity and humoral immunity compared with emulsion-adjuvanted vaccines. Herein, we developed a green and simple method for fabricating a novel nanoparticles-in-emulsions (NPE) formulation. Firstly, a temperature-sensitive hydrogel was used to prepare particles by self-solidification without additional crosslinking reagents. Secondly, the white oil was used as organic phase to avoid the particle washing procedures and organic solvent residues. Moreover, the effect of NPE as vaccine adjuvant was evaluated by using two veterinary vaccines as model antigens. NPE showed advantages than the conventional vaccine formulations in inducing both humoral and cellular immunity. This work provides a facile and broadly applicable approach for preparing nanoparticles-in-emulsions formulation, and presents an effective adjuvant for enhancing immunity against infectious diseases.
Collapse
Affiliation(s)
- Yongjuan Zou
- State Key Laboratory of Biochemical Engineering, Institute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Shuai Li
- State Key Laboratory of Biochemical Engineering, Institute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - To Ngai
- Department of ChemistryThe Chinese University of Hong KongShatinNTHong Kong
| | - Songping Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
- PLA Key Laboratory of Biopharmaceutical Production and Formulation EngineeringInstitute of Process Engineering, Chinese Academy of SciencesBeijingP. R. China
- Jiangsu National Synergetic Innovation Center for Advanced MaterialsNanjingP. R. China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
- PLA Key Laboratory of Biopharmaceutical Production and Formulation EngineeringInstitute of Process Engineering, Chinese Academy of SciencesBeijingP. R. China
- Jiangsu National Synergetic Innovation Center for Advanced MaterialsNanjingP. R. China
| |
Collapse
|
146
|
Ye T, Zhong Z, García‐Sastre A, Schotsaert M, De Geest BG. Current Status of COVID-19 (Pre)Clinical Vaccine Development. Angew Chem Int Ed Engl 2020; 59:18885-18897. [PMID: 32663348 PMCID: PMC7405471 DOI: 10.1002/anie.202008319] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/29/2022]
Abstract
The current COVID-19 pandemic has a tremendous impact on daily life world-wide. Despite the ability to dampen the spread of SARS-CoV-2, the causative agent of the diseases, through restrictive interventions, it is believed that only effective vaccines will provide sufficient control over the disease and revert societal live back to normal. At present, a double-digit number of efforts are devoted to the development of a vaccine against COVID-19. Here, we provide an overview of these (pre)clinical efforts and provide background information on the technologies behind these vaccines. In addition, we discuss potential hurdles that need to be addressed prior to mass scale clinical translation of successful vaccine candidates.
Collapse
Affiliation(s)
- Tingting Ye
- Department of PharmaceuticsGhent UniversityBelgium
| | - Zifu Zhong
- Department of PharmaceuticsGhent UniversityBelgium
| | - Adolfo García‐Sastre
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of MedicineDivision of Infectious DiseasesIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- The Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Michael Schotsaert
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | | |
Collapse
|
147
|
Špakova A, Dalgėdienė I, Insodaitė R, Sasnauskienė A, Žvirblienė A, Petraitytė-Burneikienė R. vB_EcoS_NBD2 bacteriophage-originated polytubes as a carrier for the presentation of foreign sequences. Virus Res 2020; 290:198194. [PMID: 33058966 DOI: 10.1016/j.virusres.2020.198194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/15/2023]
Abstract
Virus-based nanoparticles constitute a promising platform for the creation of efficient vaccines and nanomaterials. Previously we demonstrated, that the recombinant tail tube protein gp39 of vB_EcoS_NBD2 bacteriophage self-assembles into extremely long (from 0.1 to >3.95 μm), flexible, and stable polytubes when produced in Saccharomyces cerevisiae. To develop a tubular platform for multivalent display of foreign antigens, yeast-derived recombinant tail tube protein gp39 was chosen as a scaffold. The carboxy-terminal fusions of gp39 with various antigens up to 238 amino acids in length resulted in different synthesis efficiency and self-assembly capacity. Recombinant gp39 fused with green fluorescent protein (eGFP) comprising 238 amino acid residues was capable to self-assemble into short fluorescent polytubes with retained eGFP functional activity. By demonstrating the display of active foreign antigens on the exterior surface of polytubes, these structures may provide a promising tool for diverse applications in nanotechnology.
Collapse
Affiliation(s)
- Aliona Špakova
- Department of Eukaryote Gene Engineering, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Indrė Dalgėdienė
- Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Rasa Insodaitė
- Department of Eukaryote Gene Engineering, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Aušra Sasnauskienė
- Department of Biochemistry and Molecular Biology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Aurelija Žvirblienė
- Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Rasa Petraitytė-Burneikienė
- Department of Eukaryote Gene Engineering, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania.
| |
Collapse
|
148
|
Hsu CW, Chang MH, Chang HW, Wu TY, Chang YC. Parenterally Administered Porcine Epidemic Diarrhea Virus-Like Particle-Based Vaccine Formulated with CCL25/28 Chemokines Induces Systemic and Mucosal Immune Protectivity in Pigs. Viruses 2020; 12:E1122. [PMID: 33023277 PMCID: PMC7600258 DOI: 10.3390/v12101122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 01/12/2023] Open
Abstract
Generation of a safe, economical, and effective vaccine capable of inducing mucosal immunity is critical for the development of vaccines against enteric viral diseases. In the current study, virus-like particles (VLPs) containing the spike (S), membrane (M), and envelope (E) structural proteins of porcine epidemic diarrhea virus (PEDV) expressed by the novel polycistronic baculovirus expression vector were generated. The immunogenicity and protective efficacy of the PEDV VLPs formulated with or without mucosal adjuvants of CCL25 and CCL28 (CCL25/28) were evaluated in post-weaning pigs. While pigs intramuscularly immunized with VLPs alone were capable of eliciting systemic anti-PEDV S-specific IgG and cellular immunity, co-administration of PEDV VLPs with CCL25/28 could further modulate the immune responses by enhancing systemic anti-PEDV S-specific IgG, mucosal IgA, and cellular immunity. Upon challenge with PEDV, both VLP-immunized groups showed milder clinical signs with reduced fecal viral shedding as compared to the control group. Furthermore, pigs immunized with VLPs adjuvanted with CCL25/28 showed superior immune protection against PEDV. Our results suggest that VLPs formulated with CCL25/28 may serve as a potential PEDV vaccine candidate and the same strategy may serve as a platform for the development of other enteric viral vaccines.
Collapse
Affiliation(s)
- Chin-Wei Hsu
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (C.-W.H.); (H.-W.C.)
| | - Ming-Hao Chang
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan;
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (C.-W.H.); (H.-W.C.)
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 406, Taiwan
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (C.-W.H.); (H.-W.C.)
| |
Collapse
|
149
|
Chen CW, Saubi N, Joseph-Munné J. Design Concepts of Virus-Like Particle-Based HIV-1 Vaccines. Front Immunol 2020; 11:573157. [PMID: 33117367 PMCID: PMC7561392 DOI: 10.3389/fimmu.2020.573157] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/31/2020] [Indexed: 01/04/2023] Open
Abstract
Prophylactic vaccines remain the best approach for controlling the human immunodeficiency virus-1 (HIV-1) transmission. Despite the limited efficacy of the RV144 trial in Thailand, there is still no vaccine candidate that has been proven successful. Consequently, great efforts have been made to improve HIV-1 antigens design and discover delivery platforms for optimal immune elicitation. Owing to immunogenic, structural, and functional diversity, virus-like particles (VLPs) could act as efficient vaccine carriers to display HIV-1 immunogens and provide a variety of HIV-1 vaccine development strategies as well as prime-boost regimes. Here, we describe VLP-based HIV-1 vaccine candidates that have been enrolled in HIV-1 clinical trials and summarize current advances and challenges according to preclinical results obtained from five distinct strategies. This mini-review provides multiple perspectives to help in developing new generations of VLP-based HIV-1 vaccine candidates with better capacity to elicit specific anti-HIV immune responses.
Collapse
Affiliation(s)
- Chun-Wei Chen
- Microbiology Department, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Narcís Saubi
- Microbiology Department, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,EAVI2020 European AIDS Vaccine Initiative H2020 Research Programme, London, United Kingdom
| | - Joan Joseph-Munné
- Microbiology Department, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,EAVI2020 European AIDS Vaccine Initiative H2020 Research Programme, London, United Kingdom.,Microbiology Department, Hospital Universitari de la Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
150
|
Brisse M, Vrba SM, Kirk N, Liang Y, Ly H. Emerging Concepts and Technologies in Vaccine Development. Front Immunol 2020; 11:583077. [PMID: 33101309 PMCID: PMC7554600 DOI: 10.3389/fimmu.2020.583077] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
Despite the success of vaccination to greatly mitigate or eliminate threat of diseases caused by pathogens, there are still known diseases and emerging pathogens for which the development of successful vaccines against them is inherently difficult. In addition, vaccine development for people with compromised immunity and other pre-existing medical conditions has remained a major challenge. Besides the traditional inactivated or live attenuated, virus-vectored and subunit vaccines, emerging non-viral vaccine technologies, such as viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer innovative approaches to address existing challenges of vaccine development. They have also significantly advanced our understanding of vaccine immunology and can guide future vaccine development for many diseases, including rapidly emerging infectious diseases, such as COVID-19, and diseases that have not traditionally been addressed by vaccination, such as cancers and substance abuse. This review provides an integrative discussion of new non-viral vaccine development technologies and their use to address the most fundamental and ongoing challenges of vaccine development.
Collapse
Affiliation(s)
- Morgan Brisse
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Natalie Kirk
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
- Comparative Molecular Biosciences Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| |
Collapse
|