1
|
Ma Y, Dong C, Kim JK, Zhu W, Wei L, Wang Y, Kang SM, Wang BZ. Impact of influenza immune imprinting on immune responses to subsequent vaccinations in mice. Vaccine 2025; 46:126670. [PMID: 39731808 DOI: 10.1016/j.vaccine.2024.126670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/23/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
The immune memory imprinted during an individual's initial influenza exposure (influenza imprinting) has long-lasting effects on the host's response to subsequent influenza infections and vaccinations. Here, we investigate how different influenza virus imprinting impacts the immune responses to subunit, inactivated virus, and protein-based nanoparticle vaccines in Balb/c mice. Our results indicated a phylogenetic distance-dependent effect of influenza imprinting on subunit hemagglutinin (HA) or formalin-inactivated (FI) virus vaccine immunizations. Aichi (H3N2, group 2) HA (HA3) or FI-Aichi vaccination in mice imprinted with closely related Phili (H3N2) triggered significant Aichi-specific HAI antibody and balanced HA3-specific Th1/Th2 antibody immune responses, resulting in robust protection against Aichi. In contrast, HA3 vaccination in PR8 (H1N1, group 1) imprinted mice (PR8-2HA3) induced Th2-leaning responses comparable to those observed in mice without prior influenza immune imprinting (PBS-2HA3). However, subsequent heterosubtypic infections and vaccinations eliminated such effects on antibody subtype profiles. Nonetheless, initial virus exposure established a long-lasting capacity to produce HAI antibody responses against the imprinting strains. Moreover, Phili imprinting followed by HA3/NP nanocluster vaccination protected mice from Aichi infections and induced enhanced cross-reactive immunity. Our study highlights the significance of considering an individual's influenza exposure history when designing and evaluating the effectiveness of influenza vaccines.
Collapse
Affiliation(s)
- Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
2
|
Gnazzo V, Saleh H, Castro ÍA, Boon ACM, Pinto AK, Brien JD, López CB. DDO-adjuvanted influenza A virus nucleoprotein mRNA vaccine induces robust humoral and cellular type 1 immune responses and protects mice from challenge. mBio 2024:e0358924. [PMID: 39692514 DOI: 10.1128/mbio.03589-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024] Open
Abstract
A challenge in viral vaccine development is to produce vaccines that generate both neutralizing antibodies to prevent infection and cytotoxic CD8+ T-cells that target conserved viral proteins and can eliminate infected cells to control virus spread. mRNA technology offers an opportunity to design vaccines based on conserved CD8-targeting epitopes, but achieving robust antigen-specific CD8+ T-cells remains a challenge. Here, we tested the viral-derived oligonucleotide DDO268 as an adjuvant in the context of a model influenza A virus (IAV) nucleoprotein (NP) mRNA vaccine in C57BL/6 mice. DDO268 when co-packaged with mRNA in lipid nanoparticles is sensed by RIG I-like receptors and safely induces local type I interferon (IFN) production followed by dendritic cells type 1 activation and migration to the draining lymph nodes. This early response triggered by DDO268 improved the generation of IgG2c antibodies and antigen-specific Th1 CD4+ and CD8+ T-cells (IFNγ+TNFα+IL2+) that provided enhanced protection against lethal IAV challenge. In addition, the inclusion of DDO268 reduced the antigen dose required to achieve protection. These results highlight the potential of DDO268 as an effective mRNA vaccine adjuvant and show that an IAV NP mRNA/DDO268 vaccine is a promising approach for generating protective immunity against conserved internal IAV epitopes.IMPORTANCEVaccines that generate neutralizing antibodies and cytotoxic CD8+ T-cells targeting conserved epitopes are ideal for effective protection against viruses. mRNA vaccines combined with the right adjuvant offer a promising solution to this challenge. We show that the virus-derived oligonucleotide DDO268 enhances antibody and T-cell responses to an influenza A virus (IAV) nucleoprotein mRNA vaccine in mice. DDO268 safely induces local type I interferon production and stimulates dendritic cell activation providing enhanced protection against IAV challenge. In addition, the adjuvant activity of DDO268 allows for the use of lower antigen doses during vaccination.
Collapse
Affiliation(s)
- Victoria Gnazzo
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Hanaa Saleh
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ítalo A Castro
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Adrianus C M Boon
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Amelia K Pinto
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - James D Brien
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Carolina B López
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
3
|
Kong D, He Y, Wang J, Chi L, Ao X, Ye H, Qiu W, Zhu X, Liao M, Fan H. A single immunization with H5N1 virus-like particle vaccine protects chickens against divergent H5N1 influenza viruses and vaccine efficacy is determined by adjuvant and dosage. Emerg Microbes Infect 2024; 13:2287682. [PMID: 37994795 PMCID: PMC10763850 DOI: 10.1080/22221751.2023.2287682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023]
Abstract
The H5N1 subtype highly pathogenic avian influenza virus (HPAIV) reveals high variability and threatens poultry production and public health. To prevent the spread of H5N1 HPAIV, we developed an H5N1 virus-like particle (VLP) vaccine based on the insect cell-baculovirus expression system. Single immunization of the H5N1 VLP vaccines induced high levels of HI antibody titres and provided effective protection against homologous virus challenge comparable to the commercial inactivated vaccine. Meanwhile, we assessed the relative efficacy of different adjuvants by carrying out a head-to-head comparison of the adjuvants ISA 201 and ISA 71 and evaluated whether the two adjuvants could induce broadly protective immunity. The ISA 71 adjuvanted vaccine induced significantly higher levels of Th1 and Th2 immune responses and provided superior cross-protection against antigenically divergent H5N1 virus challenge than the ISA 201 adjuvanted vaccine. Importantly, increasing the vaccine dose could further enhance the cross-protective efficacy of H5N1 VLP vaccine and confer completely sterilizing protection against antigenically divergent H5N1 virus challenge, which was mediated by neutralizing antibodies. Our results suggest that the H5N1 VLP vaccine can provide broad-spectrum protection against divergent H5N1 influenza viruses as determined by adjuvant and vaccine dose.
Collapse
Affiliation(s)
- Dexin Kong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| | - Yanjuan He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| | - Jiaxin Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| | - Lanyan Chi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| | - Xiang Ao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| | - Hejia Ye
- Guangzhou South China Biological Medicine Co., Ltd, Guangzhou, People’s Republic of China
| | - Weihong Qiu
- Guangzhou South China Biological Medicine Co., Ltd, Guangzhou, People’s Republic of China
| | - Xiutong Zhu
- Guangzhou South China Biological Medicine Co., Ltd, Guangzhou, People’s Republic of China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People’s Republic of China
| |
Collapse
|
4
|
Hsiung KC, Chiang HJ, Reinig S, Shih SR. Vaccine Strategies Against RNA Viruses: Current Advances and Future Directions. Vaccines (Basel) 2024; 12:1345. [PMID: 39772007 PMCID: PMC11679499 DOI: 10.3390/vaccines12121345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The development of vaccines against RNA viruses has undergone a rapid evolution in recent years, particularly driven by the COVID-19 pandemic. This review examines the key roles that RNA viruses, with their high mutation rates and zoonotic potential, play in fostering vaccine innovation. We also discuss both traditional and modern vaccine platforms and the impact of new technologies, such as artificial intelligence, on optimizing immunization strategies. This review evaluates various vaccine platforms, ranging from traditional approaches (inactivated and live-attenuated vaccines) to modern technologies (subunit vaccines, viral and bacterial vectors, nucleic acid vaccines such as mRNA and DNA, and phage-like particle vaccines). To illustrate these platforms' practical applications, we present case studies of vaccines developed for RNA viruses such as SARS-CoV-2, influenza, Zika, and dengue. Additionally, we assess the role of artificial intelligence in predicting viral mutations and enhancing vaccine design. The case studies underscore the successful application of RNA-based vaccines, particularly in the fight against COVID-19, which has saved millions of lives. Current clinical trials for influenza, Zika, and dengue vaccines continue to show promise, highlighting the growing efficacy and adaptability of these platforms. Furthermore, artificial intelligence is driving improvements in vaccine candidate optimization and providing predictive models for viral evolution, enhancing our ability to respond to future outbreaks. Advances in vaccine technology, such as the success of mRNA vaccines against SARS-CoV-2, highlight the potential of nucleic acid platforms in combating RNA viruses. Ongoing trials for influenza, Zika, and dengue demonstrate platform adaptability, while artificial intelligence enhances vaccine design by predicting viral mutations. Integrating these innovations with the One Health approach, which unites human, animal, and environmental health, is essential for strengthening global preparedness against future RNA virus threats.
Collapse
Affiliation(s)
- Kuei-Ching Hsiung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food & Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science & Technology, Taoyuan 33303, Taiwan
| |
Collapse
|
5
|
Shuklina M, Stepanova L, Ozhereleva O, Kovaleva A, Vidyaeva I, Korotkov A, Tsybalova L. Inserting CTL Epitopes of the Viral Nucleoprotein to Improve Immunogenicity and Protective Efficacy of Recombinant Protein against Influenza A Virus. BIOLOGY 2024; 13:801. [PMID: 39452110 PMCID: PMC11505154 DOI: 10.3390/biology13100801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Conserved influenza virus proteins, such as the hemagglutinin stem domain (HA2), nucleoprotein (NP), and matrix protein (M), are the main targets in the development of universal influenza vaccines. Previously, we constructed a recombinant vaccine protein Flg-HA2-2-4M2ehs containing the extracellular domain of the M2 protein (M2e) and the aa76-130 sequence of the second HA subunit as target antigens. It demonstrated immunogenicity and broad protection against influenza A viruses after intranasal and parenteral administration. This study shows that CD8+ epitopes of NP, inserted into a flagellin-fused protein carrying M2e and HA2, affect the post-vaccination immune humoral response to virus antigens without reducing protection. No differences were found between the two proteins in their ability to stimulate the formation of follicular Th in the spleen, which may contribute to a long-lasting antigen-specific humoral response. The data obtained on Balb/c mice suggest that the insertion of CTL NP epitopes into the flagellin-fused protein carrying M2e and HA2 reduces the antibody response to M2e and A/H3N2. In C57Bl6 mice, this stimulates the formation of NP-specific CD8+ Tem and virus-specific mono- and multifunctional CD4+ and CD8+ Tem in the spleen and completely protects mice from influenza virus subtypes A/H1N1pdm09 and A/H3N2.
Collapse
Affiliation(s)
- Marina Shuklina
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 15/17 Prof. Popova Str., St. Petersburg 197376, Russia
| | - Liudmila Stepanova
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 15/17 Prof. Popova Str., St. Petersburg 197376, Russia
| | | | | | | | | | | |
Collapse
|
6
|
Wilks LR, Joshi G, Rychener N, Gill HS. Generation of Broad Protection against Influenza with Di-Tyrosine-Cross-Linked M2e Nanoclusters. ACS Infect Dis 2024; 10:1552-1560. [PMID: 38623820 DOI: 10.1021/acsinfecdis.3c00429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.
Collapse
Affiliation(s)
- Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Natalie Rychener
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
7
|
Lim CML, Komarasamy TV, Adnan NAAB, Radhakrishnan AK, Balasubramaniam VRMT. Recent Advances, Approaches and Challenges in the Development of Universal Influenza Vaccines. Influenza Other Respir Viruses 2024; 18:e13276. [PMID: 38513364 PMCID: PMC10957243 DOI: 10.1111/irv.13276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 03/23/2024] Open
Abstract
Every year, influenza virus infections cause significant morbidity and mortality worldwide. They pose a substantial burden of disease, in terms of not only health but also the economy. Owing to the ability of influenza viruses to continuously evolve, annual seasonal influenza vaccines are necessary as a prophylaxis. However, current influenza vaccines against seasonal strains have limited effectiveness and require yearly reformulation due to the virus undergoing antigenic drift or shift. Vaccine mismatches are common, conferring suboptimal protection against seasonal outbreaks, and the threat of the next pandemic continues to loom. Therefore, there is a great need to develop a universal influenza vaccine (UIV) capable of providing broad and durable protection against all influenza virus strains. In the quest to develop a UIV that would obviate the need for annual vaccination and formulation, a multitude of strategies is currently underway. Promising approaches include targeting the highly conserved epitopes of haemagglutinin (HA), neuraminidase (NA), M2 extracellular domain (M2e) and internal proteins of the influenza virus. The identification and characterization of broadly neutralizing antibodies (bnAbs) targeting conserved regions of the viral HA protein, in particular, have provided important insight into novel vaccine designs and platforms. This review discusses universal vaccine approaches presently under development, with an emphasis on those targeting the highly conserved stalk of the HA protein, recent technological advancements used and the future prospects of a UIV in terms of its advantages, developmental obstacles and potential shortcomings.
Collapse
Affiliation(s)
- Caryn Myn Li Lim
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Nur Amelia Azreen Binti Adnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Ammu Kutty Radhakrishnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Vinod R. M. T. Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| |
Collapse
|
8
|
Li R, Chang Z, Liu H, Wang Y, Li M, Chen Y, Fan L, Wang S, Sun X, Liu S, Cheng A, Ding P, Zhang G. Double-layered N-S1 protein nanoparticle immunization elicits robust cellular immune and broad antibody responses against SARS-CoV-2. J Nanobiotechnology 2024; 22:44. [PMID: 38291444 PMCID: PMC10825999 DOI: 10.1186/s12951-024-02293-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic is a persistent global threat to public health. As for the emerging variants of SARS-CoV-2, it is necessary to develop vaccines that can induce broader immune responses, particularly vaccines with weak cellular immunity. METHODS In this study, we generated a double-layered N-S1 protein nanoparticle (N-S1 PNp) that was formed by desolvating N protein into a protein nanoparticle as the core and crosslinking S1 protein onto the core surface against SARS-CoV-2. RESULTS Vaccination with N-S1 PNp elicited robust humoral and vigorous cellular immune responses specific to SARS-CoV-2 in mice. Compared to soluble protein groups, the N-S1 PNp induced a higher level of humoral response, as evidenced by the ability of S1-specific antibodies to block hACE2 receptor binding and neutralize pseudovirus. Critically, N-S1 PNp induced Th1-biased, long-lasting, and cross-neutralizing antibodies, which neutralized the variants of SARS-CoV-2 with minimal loss of activity. N-S1 PNp induced strong responses of CD4+ and CD8+ T cells, mDCs, Tfh cells, and GCs B cells in spleens. CONCLUSIONS These results demonstrate that N-S1 PNp vaccination is a practical approach for promoting protection, which has the potential to counteract the waning immune responses against SARS-CoV-2 variants and confer broad efficacy against future new variants. This study provides a new idea for the design of next-generation SARS-CoV-2 vaccines based on the B and T cells response coordination.
Collapse
Affiliation(s)
- Ruiqi Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- School of Advanced Agricultural Sciences , Peking University, Beijing, 100080, China
- Longhu Laboratory, Zhengzhou, 450046, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Zejie Chang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hongliang Liu
- School of Life Sciences , Zhengzhou University, Zhengzhou, 450001, China
| | - Yanan Wang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Minghui Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Yilan Chen
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Lu Fan
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Siqiao Wang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Xueke Sun
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Siyuan Liu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Anchun Cheng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Peiyang Ding
- School of Life Sciences , Zhengzhou University, Zhengzhou, 450001, China.
| | - Gaiping Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- School of Advanced Agricultural Sciences , Peking University, Beijing, 100080, China.
- Longhu Laboratory, Zhengzhou, 450046, China.
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China.
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
- School of Life Sciences , Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
9
|
Song WW, Wan MY, She JY, Zhao SL, Liu DJ, Chang HY, Deng L. Sequential Immunizations with Influenza Neuraminidase Protein Followed by Peptide Nanoclusters Induce Heterologous Protection. Viruses 2024; 16:77. [PMID: 38257777 PMCID: PMC10819419 DOI: 10.3390/v16010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/26/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Enhancing cross-protections against diverse influenza viruses is desired for influenza vaccinations. Neuraminidase (NA)-specific antibody responses have been found to independently correlate with a broader influenza protection spectrum. Here, we report a sequential immunization regimen that includes priming with NA protein followed by boosting with peptide nanoclusters, with which targeted enhancement of antibody responses in BALB/c mice to certain cross-protective B-cell epitopes of NA was achieved. The nanoclusters were fabricated via desolvation with absolute ethanol and were only composed of composite peptides. Unlike KLH conjugates, peptide nanoclusters would not induce influenza-unrelated immunity. We found that the incorporation of a hemagglutinin peptide of H2-d class II restriction into the composite peptides could be beneficial in enhancing the NA peptide-specific antibody response. Of note, boosters with N2 peptide nanoclusters induced stronger serum cross-reactivities to heterologous N2 and even heterosubtypic N7 and N9 than triple immunizations with the prototype recombinant tetrameric (rt) N2. The mouse challenge experiments with HK68 H3N2 also demonstrated the strong effectiveness of the peptide nanocluster boosters in conferring heterologous protection.
Collapse
Affiliation(s)
- Wen-Wen Song
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410082, China; (W.-W.S.); (M.-Y.W.); (J.-Y.S.); (S.-L.Z.); (D.-J.L.)
| | - Mu-Yang Wan
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410082, China; (W.-W.S.); (M.-Y.W.); (J.-Y.S.); (S.-L.Z.); (D.-J.L.)
| | - Jia-Yue She
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410082, China; (W.-W.S.); (M.-Y.W.); (J.-Y.S.); (S.-L.Z.); (D.-J.L.)
| | - Shi-Long Zhao
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410082, China; (W.-W.S.); (M.-Y.W.); (J.-Y.S.); (S.-L.Z.); (D.-J.L.)
| | - De-Jian Liu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410082, China; (W.-W.S.); (M.-Y.W.); (J.-Y.S.); (S.-L.Z.); (D.-J.L.)
| | - Hai-Yan Chang
- College of Life Sciences, Hunan Normal University, Changsha 410082, China
| | - Lei Deng
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410082, China; (W.-W.S.); (M.-Y.W.); (J.-Y.S.); (S.-L.Z.); (D.-J.L.)
- Beijing Weimiao Biotechnology Co., Ltd., Haidian District, Beijing 100093, China
| |
Collapse
|
10
|
Xia Y, Liu K, Wang F, Xu Z, Wang Y, Zong R, Xu Y, Li P, Deng B, Xu M, Chen G. Self-Assembled Virus-Like Particle Vaccines via Fluorophilic Interactions Enable Infection Mimicry and Immune Protection. Adv Healthc Mater 2023; 12:e2301647. [PMID: 37703498 DOI: 10.1002/adhm.202301647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Influenza epidemics persistently threaten global health. Vaccines based on virus-like particles (VLPs), which resemble the native conformation of viruses, have emerged as vaccine candidates. However, the production of VLPs via genetic engineering remains constrained by challenges such as low yields, high costs, and being time consuming. In this study, a novel VLP platform is developed that could mimic infection and confer influenza protection through fluorination-driven self-assembly. The VLPs closely mimick the key steps in viral infection including dendritic cell (DC) attachment and pH-responsive endo-lysosomal escape, which enhances DC maturation and antigen cross-presentation. It is also observed that the VLPs migrate from the injection site to the draining lymph nodes efficiently. Immunization with VLPs triggers both Th1 and Th2 cellular responses, thereby inducing an improved CD8+ T cell response along with strong antigen-specific antibody responses. In several infected mouse models, VLP vaccines ameliorate weight loss, lung virus titers, pulmonary pathologies, and confer full protection against H1N1, H6N2, H9N2, and mixed influenza viruses. Therefore, the results support the potential of VLPs as an effective influenza vaccine with improved immune potency against infection. A methodology to generate VLPs based on fluorophilic interactions, which can be a general approach for development of pathogenic VLPs, is reported.
Collapse
Affiliation(s)
- Yinhe Xia
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Kai Liu
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Fei Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Zhou Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Yuesheng Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Rongling Zong
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Yemin Xu
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Ping Li
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Maolei Xu
- The Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China
| | - Gang Chen
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| |
Collapse
|
11
|
Park J, Zhang Z, Belinskaya T, Tsoras AN, Chao CC, Jiang L, Champion JA. Dual-Antigen Subunit Vaccine Nanoparticles for Scrub Typhus. Pathogens 2023; 12:1390. [PMID: 38133275 PMCID: PMC10745692 DOI: 10.3390/pathogens12121390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Orientia tsutsugamushi is the causative pathogen of scrub typhus, an acute febrile disease prevalent in the Asia-Pacific region that is spread to people through chigger bites. Despite the emerging threat, there is no currently available vaccine against O. tsutsugamushi. Here, we developed dual-antigen subunit vaccine nanoparticles using recombinant 47 kD and 56 kD proteins, which are immunogenic outer membrane antigens of O. tsutsugamushi. The biocompatible protein vaccine nanoparticles were formed via desolvation of r56 or r47E antigens with acetone, coating with an additional layer of the 56 kD protein, and stabilization with reducible homobifunctional DTSSP and heterobifunctional SDAD crosslinkers. The dual-antigen subunit vaccine nanoparticles significantly improved antigen-specific antibody responses in vaccinated mice. Most importantly, the dual-antigen nanoparticles coated with an additional layer of the 56 kD protein were markedly more immunogenic than soluble antigens or single-antigen nanoparticles in the context of cellular immune responses. Given the significance of cellular immune responses for protection against O. tsutsugamushi, these results demonstrate the potent immunogenicity of dual-layered antigen nanoparticles and their potential as a promising strategy for developing vaccines against scrub typhus.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA 30332, USA; (J.P.); (A.N.T.)
| | - Zhiwen Zhang
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Bethesda, MD 20817, USA; (Z.Z.); (T.B.)
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Tatyana Belinskaya
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Bethesda, MD 20817, USA; (Z.Z.); (T.B.)
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Alexandra N. Tsoras
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA 30332, USA; (J.P.); (A.N.T.)
| | - Chien-Chung Chao
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Le Jiang
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Bethesda, MD 20817, USA; (Z.Z.); (T.B.)
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Julie A. Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA 30332, USA; (J.P.); (A.N.T.)
| |
Collapse
|
12
|
Gu W, An J, Li Y, Yang Y, Wang S, Shan H, Li S, Li H, Liu G, Li K, Yin Y, Mu J, Chen X. Tuning the Organ Tropism of Polymersome for Spleen-Selective Nanovaccine Delivery to Boost Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301686. [PMID: 37165781 DOI: 10.1002/adma.202301686] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/08/2023] [Indexed: 05/12/2023]
Abstract
The past few decades have witnessed explosive development in drug delivery systems. However, in vivo delivery suffers from non-specific distribution in non-targeted organs or tissues, which may cause undesired side effects and even genotoxicity. Here, a general strategy that enables tuning the tropism of polymersomes for liver- and spleen-selective delivery is reported. By using a library screening approach, spleen-targeted polymersome PH9-Aln-8020 and liver-targeted polymersome PA9-ZP3-5050 are identified accordingly. Meanwhile, the second near-infrared (NIR-II) fluorescence imaging allows for in vivo dynamic evaluation of their spatial and temporal accumulation in specific tissues. O ur findings indicate that both polymer composition and protein corona on the surface are essential to determine the in vivo fate of polymersomes and tendency for specific organs. Importantly, PH9-Aln-8020 is employed as a systemic nanocarrier to co-deliver the antigen and adjuvant, which remarkably boost splenic immune responses in acute myeloid leukemia, melanoma, and melanoma lung metastasis mouse models. This study may open a new frontier for polymersomes in organ-selective delivery and other biomedical applications.
Collapse
Affiliation(s)
- Wenxing Gu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine, College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Jingnan An
- The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Yaxi Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yajie Yang
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Shumin Wang
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Hui Shan
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Shenhua Li
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Hui Li
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine, College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Guoyong Liu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Kai Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yuxin Yin
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Jing Mu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine, College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
13
|
Wang H, Xu J, Xiang L. Microneedle-Mediated Transcutaneous Immunization: Potential in Nucleic Acid Vaccination. Adv Healthc Mater 2023; 12:e2300339. [PMID: 37115817 DOI: 10.1002/adhm.202300339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/07/2023] [Indexed: 04/29/2023]
Abstract
Efforts aimed at exploring economical and efficient vaccination have taken center stage to combat frequent epidemics worldwide. Various vaccines have been developed for infectious diseases, among which nucleic acid vaccines have attracted much attention from researchers due to their design flexibility and wide application. However, the lack of an efficient delivery system considerably limits the clinical translation of nucleic acid vaccines. As mass vaccinations via syringes are limited by low patient compliance and high costs, microneedles (MNs), which can achieve painless, cost-effective, and efficient drug delivery, can provide an ideal vaccination strategy. The MNs can break through the stratum corneum barrier in the skin and deliver vaccines to the immune cell-rich epidermis and dermis. In addition, the feasibility of MN-mediated vaccination is demonstrated in both preclinical and clinical studies and has tremendous potential for the delivery of nucleic acid vaccines. In this work, the current status of research on MN vaccines is reviewed. Moreover, the improvements of MN-mediated nucleic acid vaccination are summarized and the challenges of its clinical translation in the future are discussed.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
14
|
Zhu W, Park J, Pho T, Wei L, Dong C, Kim J, Ma Y, Champion JA, Wang BZ. ISCOMs/MPLA-Adjuvanted SDAD Protein Nanoparticles Induce Improved Mucosal Immune Responses and Cross-Protection in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301801. [PMID: 37162451 PMCID: PMC10524461 DOI: 10.1002/smll.202301801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/20/2023] [Indexed: 05/11/2023]
Abstract
The epidemics caused by the influenza virus are a serious threat to public health and the economy. Adding appropriate adjuvants to improve immunogenicity and finding effective mucosal vaccines to combat respiratory infection at the portal of virus entry are important strategies to boost protection. In this study, a novel type of core/shell protein nanoparticle consisting of influenza nucleoprotein (NP) as the core and NA1-M2e or NA2-M2e fusion proteins as the coating antigens by SDAD hetero-bifunctional crosslinking is exploited. Immune-stimulating complexes (ISCOMs)/monophosphoryl lipid A (MPLA) adjuvants further boost the NP/NA-M2e SDAD protein nanoparticle-induced immune responses when administered intramuscularly. The ISCOMs/MPLA-adjuvanted protein nanoparticles are delivered through the intranasal route to validate the application as mucosal vaccines. ISCOMs/MPLA-adjuvanted nanoparticles induce significantly strengthened antigen-specific antibody responses, cytokine-secreting splenocytes in the systemic compartment, and higher levels of antigen-specific IgA and IgG in the local mucosa. Meanwhile, significantly expanded lung resident memory (RM) T and B cells (TRM /BRM ) and alveolar macrophages population are observed in ISCOMs/MPLA-adjuvanted nanoparticle-immunized mice with a 100% survival rate after homogeneous and heterogeneous H3N2 viral challenges. Taken together, ISCOMs/MPLA-adjuvanted protein nanoparticles could improve strong systemic and mucosal immune responses conferring protection in different immunization routes.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Jaeyoung Park
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Thomas Pho
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
15
|
Heng WT, Lim HX, Tan KO, Poh CL. Validation of Multi-epitope Peptides Encapsulated in PLGA Nanoparticles Against Influenza A Virus. Pharm Res 2023; 40:1999-2025. [PMID: 37344603 DOI: 10.1007/s11095-023-03540-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Influenza is a highly contagious respiratory disease which poses a serious threat to public health globally, causing severe diseases in 3-5 million humans and resulting in 650,000 deaths annually. The current licensed seasonal influenza vaccines lacked cross-reactivity against novel emerging influenza strains as they conferred limited neutralising capabilities. To address the issue, we designed a multi-epitope peptide-based vaccine delivered by the self-adjuvanting PLGA nanoparticles against influenza infections. METHODS A total of six conserved peptides representing B- and T-cell epitopes of Influenza A were identified and they were formulated in either incomplete Freund's adjuvant containing CpG ODN 1826 or being encapsulated in PLGA nanoparticles for the evaluation of immunogenicity in BALB/c mice. RESULTS The self-adjuvanting PLGA nanoparticles encapsulating the six conserved peptides were capable of eliciting the highest levels of IgG and IFN- γ producing cells. In addition, the immunogenicity of the six peptides encapsulated in PLGA nanoparticles showed greater humoral and cellular mediated immune responses elicited by the mixture of six naked peptides formulated in incomplete Freund's adjuvant containing CpG ODN 1826 in the immunized mice. Peptide 3 from the mixture of six peptides was found to exert necrotic effect on CD3+ T-cells and this finding indicated that peptide 3 should be removed from the nanovaccine formulation. CONCLUSION The study demonstrated the self-adjuvanting properties of the PLGA nanoparticles as a delivery system without the need for incorporation of toxic and costly conventional adjuvants in multi-epitope peptide-based vaccines.
Collapse
Affiliation(s)
- Wen Tzuen Heng
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Hui Xuan Lim
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Kuan Onn Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
16
|
Wang Z, Zhang T, Jia F, Ge C, He Y, Tian Y, Wang W, Yang G, Huang H, Wang J, Shi C, Yang W, Cao X, Zeng Y, Wang N, Qian A, Wang C, Jiang Y. Homologous Sequential Immunization Using Salmonella Oral Administration Followed by an Intranasal Boost with Ferritin-Based Nanoparticles Enhanced the Humoral Immune Response against H1N1 Influenza Virus. Microbiol Spectr 2023; 11:e0010223. [PMID: 37154735 PMCID: PMC10269571 DOI: 10.1128/spectrum.00102-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
The influenza virus continues to pose a great threat to public health due to the frequent variations in RNA viruses. Vaccines targeting conserved epitopes, such as the extracellular domain of the transmembrane protein M2 (M2e), a nucleoprotein, and the stem region of hemagglutinin proteins, have been developed, but more efficient strategies, such as nanoparticle-based vaccines, are still urgently needed. However, the labor-intensive in vitro purification of nanoparticles is still necessary, which could hinder the application of nanoparticles in the veterinary field in the future. To overcome this limitation, we used regulated lysis Salmonella as an oral vector with which to deliver three copies of M2e (3M2e-H1N1)-ferritin nanoparticles in situ and evaluated the immune response. Then, sequential immunization using Salmonella-delivered nanoparticles followed by an intranasal boost with purified nanoparticles was performed to further improve the efficiency. Compared with 3M2e monomer administration, Salmonella-delivered in situ nanoparticles significantly increased the cellular immune response. Additionally, the results of sequential immunization showed that the intranasal boost with purified nanoparticles dramatically stimulated the activation of lung CD11b dendritic cells (DCs) and elevated the levels of effector memory T (TEM) cells in both spleen and lung tissues as well as those of CD4 and CD8 tissue-resident memory T (TRM) cells in the lungs. The increased production of mucosal IgG and IgA antibody titers was also observed, resulting in further improvements to protection against a virus challenge, compared with the pure oral immunization group. Salmonella-delivered in situ nanoparticles efficiently increased the cellular immune response, compared with the monomer, and sequential immunization further improved the systemic immune response, as shown by the activation of DCs, the production of TEM cells and TRM cells, and the mucosal immune response, thereby providing us with a novel strategy by which to apply nanoparticle-based vaccines in the future. IMPORTANCE Salmonella-delivered in situ nanoparticle platforms may provide novel nanoparticle vaccines for oral administration, which would be beneficial for veterinary applications. The combination of administering Salmonella-vectored, self-assembled nanoparticles and an intranasal boost with purified nanoparticles significantly increased the production of effector memory T cells and lung resident memory T cells, thereby providing partial protection against an influenza virus challenge. This novel strategy could open a novel avenue for the application of nanoparticle vaccines for veterinary purposes.
Collapse
Affiliation(s)
- Zhannan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tongyu Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Futing Jia
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chongbo Ge
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yingkai He
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yawen Tian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wenfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Guilian Yang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Haibin Huang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jianzhong Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunwei Shi
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wentao Yang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Aidong Qian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yanlong Jiang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
17
|
CpG 1018 Is an Effective Adjuvant for Influenza Nucleoprotein. Vaccines (Basel) 2023; 11:vaccines11030649. [PMID: 36992232 PMCID: PMC10055716 DOI: 10.3390/vaccines11030649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/15/2023] Open
Abstract
Current influenza vaccines mainly induce neutralizing antibodies against the highly variable surface antigen hemagglutinin and require annual manufacturing and immunization. Different from surface antigens, intracellular nucleoprotein (NP) is highly conserved and has been an attractive target to develop universal T cell vaccines against influenza. Yet, influenza NP protein mainly induces humoral immune responses and lacks the ability to induce potent cytotoxic T lymphocyte (CTL) responses, key for the success of universal T cell vaccines. This study compared CpG 1018 and AddaVax to enhance recombinant NP-induced CTL responses and protection in murine models. CpG 1018 was explored to boost intradermal NP immunization, while AddaVax was explored to boost intramuscular NP immunization due to the high risk of AddaVax adjuvant to induce significant local reactions following intradermal delivery. We found CpG 1018 was highly effective to enhance NP-induced humoral and cellular immune responses superior to AddaVax adjuvant. Furthermore, CpG 1018 potentiated Th1-biased antibody responses, while AddaVax enhanced Th1/Th2-balanced antibody responses. CpG 1018 significantly enhanced IFNγ-secreting Th1 cells, while AddaVax adjuvant significantly increased IL4-secreting Th2 cells. Influenza NP immunization in the presence of CpG 1018 induced significant protection against lethal viral challenges, while influenza NP immunization in the presence of AddaVax failed to elicit significant protection. Our data validated CpG 1018 as an effective adjuvant to enhance influenza NP-induced CTL responses and protection.
Collapse
|
18
|
Park J, Champion JA. Effect of Antigen Structure in Subunit Vaccine Nanoparticles on Humoral Immune Responses. ACS Biomater Sci Eng 2023; 9:1296-1306. [PMID: 36848229 PMCID: PMC10015428 DOI: 10.1021/acsbiomaterials.2c01516] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 03/01/2023]
Abstract
Subunit vaccines offer numerous attractive features, including good safety profiles and well-defined components with highly characterized properties because they do not contain whole pathogens. However, vaccine platforms based on one or few selected antigens are often poorly immunogenic. Several advances have been made in improving the effectiveness of subunit vaccines, including nanoparticle formulation and/or co-administration with adjuvants. Desolvation of antigens into nanoparticles is one approach that has been successful in eliciting protective immune responses. Despite this advance, damage to the antigen structure by desolvation can compromise the recognition of conformational antigens by B cells and the subsequent humoral response. Here, we used ovalbumin as a model antigen to demonstrate enhanced efficacy of subunit vaccines by preserving antigen structures in nanoparticles. An altered antigen structure due to desolvation was first validated by GROMACS and circular dichroism. Desolvant-free nanoparticles with a stable ovalbumin structure were successfully synthesized by directly cross-linking ovalbumin or using ammonium sulfate to form nanoclusters. Alternatively, desolvated OVA nanoparticles were coated with a layer of OVA after desolvation. Vaccination with salt-precipitated nanoparticles increased OVA-specific IgG titers 4.2- and 22-fold compared to the desolvated and coated nanoparticles, respectively. In addition, enhanced affinity maturation by both salt precipitated and coated nanoparticles was displayed in contrast to desolvated nanoparticles. These results demonstrate both that salt-precipitated antigen nanoparticles are a potential new vaccine platform with significantly improved humoral immunity and a functional value of preserving antigen structures in vaccine nanoparticle design.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
19
|
Kim CG, Lee JC, Ju DB, Kim SK, Yun CH, Cho CS. Enhancement of Immune Responses Elicited by Nanovaccines through a Cross-Presentation Pathway. Tissue Eng Regen Med 2023; 20:355-370. [PMID: 36884197 PMCID: PMC9994410 DOI: 10.1007/s13770-023-00527-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Numerous studies have aimed to develop novel advanced vaccines, in part because traditional vaccines have been unsuccessful in preventing rapidly emerging and reemerging viral and bacterial infections. There is a need for an advanced vaccine delivery system to ensure the successful induction of humoral and cellular immune responses. In particular, the ability of nanovaccines to modulate intracellular antigen delivery by inducing exogenous antigens (loaded onto major histocompatibility complex class 1 molecules) in CD8+ T cells, the so-called cross-presentation pathway, has attracted a great deal of attention. Protection against viral and intracellular bacterial infections relies on cross-presentation. This review discusses the advantages, requirements, and preparation of nanovaccines, the cross-presentation mechanism, the several parameters affecting cross-presentation by nanovaccines, and future perspectives.
Collapse
Affiliation(s)
- Cheol-Gyun Kim
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeong-Cheol Lee
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Do-Bin Ju
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seo-Kyung Kim
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea.
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-Do, 25354, Republic of Korea.
- Interdisciplinary Programs in Agricultural Genomics, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
20
|
Wang Y, Dong C, Ma Y, Zhu W, Gill HS, Denning TL, Kang SM, Wang BZ. Monophosphoryl lipid A-adjuvanted nucleoprotein-neuraminidase nanoparticles improve immune protection against divergent influenza viruses. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102614. [PMID: 36265560 PMCID: PMC9756393 DOI: 10.1016/j.nano.2022.102614] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Universal influenza vaccines are urgently needed to prevent recurrent influenza epidemics and inevitable pandemics. We generated double-layered protein nanoparticles incorporating two conserved influenza antigens-nucleoprotein and neuraminidase-through a two-step desolvation-crosslinking method. These protein nanoparticles displayed immunostimulatory properties to antigen-presenting cells by promoting inflammatory cytokine (IL-6 and TNF-α) secretion from JAWS II dendric cells. The nanoparticle immunization induced significant antigen-specific humoral and cellular responses, including antigen-binding and neutralizing antibodies, antibody- and cytokine (IFN-γ and IL-4)-secreting cells, and NP147-155 tetramer-specific cytotoxic T lymphocyte (CTL) responses. Co-administration of monophosphoryl lipid A (MPLA, a toll-like receptor 4 agonist) with the protein nanoparticles further improved immune responses and conferred heterologous and heterosubtypic influenza protection. The MPLA-adjuvanted nanoparticles reduced lung inflammation post-infection. The results demonstrated that the combination of MPLA and conserved protein nanoparticles could be developed into an improved universal influenza vaccine strategy.
Collapse
Affiliation(s)
- Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
21
|
Wilks LR, Joshi G, Kang SM, Wang BZ, Gill HS. Peptide Cross-Linking Using Tyrosine Residues Facilitated by an Exogenous Nickel-Histidine Complex: A Facile Approach for Enhancing Vaccine-Specific Immunogenicity. ACS Infect Dis 2022; 8:2389-2395. [PMID: 36346898 DOI: 10.1021/acsinfecdis.2c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An improved method for the generation of peptide vaccines using di-tyrosine cross-linking is described. The conserved ion channel peptide, M2e, of influenza A virus was modified with the addition of small tyrosine-rich regions (GYGY-) at both the N- and C-termini and extensively cross-linked via tyrosine-tyrosine linkages to form peptide nanoclusters. The cross-linking was catalyzed using exogenous nickel(II) ions complexed to an exogenous glycine-glycine-histidine peptide in the presence of an oxidizer. Mice that were intranasally or intramuscularly immunized with the M2e-vaccine nanoclusters induced comparable levels of M2e-specific serum antibodies. Vaccination via the intranasal or intramuscular route protected mice from subsequent lethal challenge with an influenza A virus. In comparison to our previous approach, where a histidine-rich tag was added into the peptide structure, the use of exogenous histidine reduced irrelevant off-target immune response. Additionally, the purity of the resulting nanoclusters is an attractive feature, making this approach appealing for vaccine development.
Collapse
Affiliation(s)
- Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave., Atlanta, Georgia 30302, United States
| | - Bao-Zhong Wang
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave., Atlanta, Georgia 30302, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
22
|
Pho T, Champion JA. Surface Engineering of Protein Nanoparticles Modulates Transport, Adsorption, and Uptake in Mucus. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51697-51710. [PMID: 36354361 DOI: 10.1021/acsami.2c14670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Protein nanoparticles have been demonstrated as effective carriers for protein antigens and therapeutics due to properties endowed by their protein composition. They exhibit high protein to carrier yields, biocompatibility, and heterogeneous surface properties. While protein nanoparticles have been delivered via multiple routes, including intranasal, their interactions with mucosal barriers have not been well studied or modified. Biological barriers associated with intranasal delivery consist of viscoelastic mucus that hinders material transport through surface interactions and the underlying epithelium. Herein, we altered protein nanoparticle surface properties and characterized interactions with nasal mucus and the subsequent effects on diffusion, cellular uptake, and immune cell maturation. Ovalbumin protein nanoparticles were used, serving as a model vaccine nanoparticle. Unmodified ovalbumin protein nanoparticles were compared to cationic ovalbumin particles functionalized with amine groups, neutral particles functionalized with polyethylene glycol, and zwitterionic particles coated layer-by-layer (LBL) with chitosan and oligonucleotides. Transport analysis indicated rapid diffusion of polyethylene glycol and LBL-modified ovalbumin nanoparticles in porcine nasal mucus, while cationic particles were mucoadhesive. Cellular uptake in the presence of mucus by epithelial and dendritic cells was highest for particles containing positive charges, both LBL and amine-functionalized. These particles also exhibited the most diverse adsorbed protein corona from nasal fluids. The corona impacted both dendritic cell uptake and maturation, with polyethylene glycol and LBL modifications improving CD86 expression. Altogether, surface modifications on protein-based nanocarriers are shown to facilitate distinctive physical and cellular behavior associated with mucosal delivery.
Collapse
Affiliation(s)
- Thomas Pho
- School of Chemical and Biomolecular Engineering, BioEngineering Program, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia30332-2000, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, BioEngineering Program, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia30332-2000, United States
| |
Collapse
|
23
|
Oh J, Subbiah J, Kim KH, Park BR, Bhatnagar N, Garcia KR, Liu R, Jung YJ, Shin CH, Seong BL, Kang SM. Impact of hemagglutination activity and M2e immunity on conferring protection against influenza viruses. Virology 2022; 574:37-46. [PMID: 35914365 PMCID: PMC9978532 DOI: 10.1016/j.virol.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/05/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022]
Abstract
To improve cross-protection of influenza vaccination, we tested conjugation of conserved M2e epitopes to the surface of inactivated influenza virus (iPR8-M2e*). Treatment of virus with chemical cross-linker led to diminished hemagglutination activity and failure to induce hemagglutination inhibiting antibodies. Conjugated iPR8-M2e* vaccine was less protective against homologous and heterosubtypic viruses, despite the induction of virus-specific binding IgG antibodies. In alternative approaches to enhance cross-protection, we developed a genetically linked chimeric protein (M2e-B stalk) vaccine with M2e of influenza A and hemagglutinin (HA) stalk of influenza B virus. Vaccination of mice with inactivated influenza A virus supplemented with M2e-B stalk effectively induced hemagglutination inhibiting antibodies, humoral and cellular M2e immune responses, and enhanced heterosubtypic protection. This study demonstrates the importance of HA functional integrity in influenza vaccine efficacy and that supplementation of influenza vaccines with M2e-B stalk protein could be a feasible strategy of improving cross-protection against influenza viruses.
Collapse
Affiliation(s)
- Judy Oh
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Jeeva Subbiah
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Karla Ruiz Garcia
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Rong Liu
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Chong-Hyun Shin
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Baik-Lin Seong
- Department of Microbiology, College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Vaccine Innovative Technology ALliance (VITAL)-Korea, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
24
|
Song Y, Zhu W, Wang Y, Deng L, Ma Y, Dong C, Gonzalez GX, Kim J, Wei L, Kang SM, Wang BZ. Layered protein nanoparticles containing influenza B HA stalk induced sustained cross-protection against viruses spanning both viral lineages. Biomaterials 2022; 287:121664. [PMID: 35810540 PMCID: PMC9822777 DOI: 10.1016/j.biomaterials.2022.121664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 01/11/2023]
Abstract
The influenza epidemics pose a significant threat to public health. Of them, type B influenza coincided with several severe flu outbreaks. The efficacy of the current seasonal flu vaccine is limited due to the antigenicity changes of circulating strains. In this study, we generated structure-stabilized HA stalk antigens from influenza B and fabricated double-layered protein nanoparticles as universal influenza B vaccine candidates. In vitro studies found that the resulting protein nanoparticles were effectively taken up to activate dendritic cells. Nanoparticle immunization induced broadly reactive immune responses conferring robust and sustained cross-immune protection against influenza B virus strains of both lineages. The results reveal the potential of layered protein nanoparticles incorporated with structure-stabilized constant antigens as a universal influenza vaccine with improved immune protective potency and breadth.
Collapse
Affiliation(s)
- Yufeng Song
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Lei Deng
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA; Hunan Provincial Kay Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, 410082, China
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Gilbert X Gonzalez
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
25
|
Zhao Y, Li Z, Voyer J, Li Y, Chen X. Flagellin/Virus-like Particle Hybrid Platform with High Immunogenicity, Safety, and Versatility for Vaccine Development. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21872-21885. [PMID: 35467839 PMCID: PMC9121874 DOI: 10.1021/acsami.2c01028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/13/2022] [Indexed: 05/07/2023]
Abstract
Hepatitis B core (HBc) virus-like particles (VLPs) and flagellin are highly immunogenic and widely explored vaccine delivery platforms. Yet, HBc VLPs mainly allow the insertion of relatively short antigenic epitopes into the immunodominant c/e1 loop without affecting VLP assembly, and flagellin-based vaccines carry the risk of inducing systemic adverse reactions. This study explored a hybrid flagellin/HBc VLP (FH VLP) platform to present heterologous antigens by replacing the surface-exposed D3 domain of flagellin. FH VLPs were prepared by the insertion of flagellin gene into the c/e1 loop of HBc, followed by E. coli expression, purification, and self-assembly into VLPs. Using the ectodomain of influenza matrix protein 2 (M2e) and ovalbumin (OVA) as models, we found that the D3 domain of flagellin could be replaced with four tandem copies of M2e or the cytotoxic T lymphocyte (CTL) epitope of OVA without interfering with the FH VLP assembly, while the insertion of four tandem copies of M2e into the c/e1 loop of HBc disrupted the VLP assembly. FH VLP-based M2e vaccine elicited potent anti-M2e antibody responses and conferred significant protection against multiple influenza A viral strains, while FljB- or HBc-based M2e vaccine failed to elicit significant protection. FH VLP-based OVA peptide vaccine elicited more potent CTL responses and protection against OVA-expressing lymphoma or melanoma challenges than FljB- or HBc-based OVA peptide vaccine. FH VLP-based vaccines showed a good systemic safety, while flagellin-based vaccines significantly increased serum interleukin 6 and tumor necrosis factor α levels and also rectal temperature at increased doses. We further found that the incorporation of a clinical CpG 1018 adjuvant could enhance the efficacy of FH VLP-based vaccines. Our data support FH VLPs to be a highly immunogenic, safe, and versatile platform for vaccine development to elicit potent humoral and cellular immune responses.
Collapse
Affiliation(s)
- Yiwen Zhao
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Zhuofan Li
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Jewel Voyer
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Yibo Li
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Xinyuan Chen
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| |
Collapse
|
26
|
Hassan J, Haigh C, Ahmed T, Uddin MJ, Das DB. Potential of Microneedle Systems for COVID-19 Vaccination: Current Trends and Challenges. Pharmaceutics 2022; 14:1066. [PMID: 35631652 PMCID: PMC9144974 DOI: 10.3390/pharmaceutics14051066] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
To prevent the coronavirus disease 2019 (COVID-19) pandemic and aid restoration to prepandemic normality, global mass vaccination is urgently needed. Inducing herd immunity through mass vaccination has proven to be a highly effective strategy for preventing the spread of many infectious diseases, which protects the most vulnerable population groups that are unable to develop immunity, such as people with immunodeficiencies or weakened immune systems due to underlying medical or debilitating conditions. In achieving global outreach, the maintenance of the vaccine potency, transportation, and needle waste generation become major issues. Moreover, needle phobia and vaccine hesitancy act as hurdles to successful mass vaccination. The use of dissolvable microneedles for COVID-19 vaccination could act as a major paradigm shift in attaining the desired goal to vaccinate billions in the shortest time possible. In addressing these points, we discuss the potential of the use of dissolvable microneedles for COVID-19 vaccination based on the current literature.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Charlotte Haigh
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| | - Tanvir Ahmed
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Md Jasim Uddin
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| |
Collapse
|
27
|
Kong D, Chen T, Hu X, Lin S, Gao Y, Ju C, Liao M, Fan H. Supplementation of H7N9 Virus-Like Particle Vaccine With Recombinant Epitope Antigen Confers Full Protection Against Antigenically Divergent H7N9 Virus in Chickens. Front Immunol 2022; 13:785975. [PMID: 35265069 PMCID: PMC8898936 DOI: 10.3389/fimmu.2022.785975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/27/2022] [Indexed: 01/18/2023] Open
Abstract
The continuous evolution of the H7N9 avian influenza virus suggests a potential outbreak of an H7N9 pandemic. Therefore, to prevent a potential epidemic of the H7N9 influenza virus, it is necessary to develop an effective crossprotective influenza vaccine. In this study, we developed H7N9 virus-like particles (VLPs) containing HA, NA, and M1 proteins derived from H7N9/16876 virus and a helper antigen HMN based on influenza conserved epitopes using a baculovirus expression vector system (BEVS). The results showed that the influenza VLP vaccine induced a strong HI antibody response and provided effective protection comparable with the effects of commercial inactivated H7N9 vaccines against homologous H7N9 virus challenge in chickens. Meanwhile, the H7N9 VLP vaccine induced robust crossreactive HI and neutralizing antibody titers against antigenically divergent H7N9 viruses isolated in wave 5 and conferred on chickens complete clinical protection against heterologous H7N9 virus challenge, significantly inhibiting virus shedding in chickens. Importantly, supplemented vaccination with HMN antigen can enhance Th1 immune responses; virus shedding was completely abolished in the vaccinated chickens. Our study also demonstrated that viral receptor-binding avidity should be taken into consideration in evaluating an H7N9 candidate vaccine. These studies suggested that supplementing influenza VLP vaccine with recombinant epitope antigen will be a promising strategy for the development of broad-spectrum influenza vaccines.
Collapse
Affiliation(s)
- Dexin Kong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Taoran Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaolong Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shaorong Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yinze Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chunmei Ju
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
28
|
Dong C, Wang BZ. Engineered Nanoparticulate Vaccines to Combat Recurring and Pandemic Influenza Threats. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100122. [PMID: 35754779 PMCID: PMC9231845 DOI: 10.1002/anbr.202100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Reoccurring seasonal flu epidemics and occasional pandemics are among the most severe threats to public health. Current seasonal influenza vaccines provide limited protection against drifted circulating strains and no protection against influenza pandemics. Next-generation influenza vaccines, designated as universal influenza vaccines, should be safe, affordable, and elicit long-lasting cross-protective influenza immunity. Nanotechnology plays a critical role in the development of such novel vaccines. Engineered nanoparticles can incorporate multiple advantageous properties into the same nanoparticulate platforms to improve vaccine potency and breadth. These immunological properties include virus-like biomimicry, high antigen-load, controlled antigen release, targeted delivery, and induction of innate signaling pathways. Many nanoparticle influenza vaccines have shown promising results in generating potent and broadly protective immune responses. This review will summarize the necessity and characteristics of next-generation influenza vaccines and the immunological correlates of broad influenza immunity and focus on how cutting-edge nanoparticle technology contributes to such vaccine development. The review will give new insights into the rational design of nanoparticle universal vaccines to combat influenza epidemics and pandemics.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| |
Collapse
|
29
|
Trends in Drug- and Vaccine-based Dissolvable Microneedle Materials and Methods of Fabrication. Eur J Pharm Biopharm 2022; 173:54-72. [DOI: 10.1016/j.ejpb.2022.02.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/24/2022] [Accepted: 02/19/2022] [Indexed: 12/18/2022]
|
30
|
Ma Y, Wang Y, Dong C, Gonzalez GX, Song Y, Zhu W, Kim J, Wei L, Wang BZ. Influenza NP core and HA or M2e shell double-layered protein nanoparticles induce broad protection against divergent influenza A viruses. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102479. [PMID: 34743020 PMCID: PMC8897236 DOI: 10.1016/j.nano.2021.102479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/23/2021] [Accepted: 10/25/2021] [Indexed: 02/03/2023]
Abstract
Influenza viral infection causes acute upper respiratory diseases in humans, posing severe risks to global public health. However, current vaccines provide limited protection against mismatched circulating influenza A viruses. Here, the immune responses induced in mice by novel double-layered protein nanoparticles were investigated. The nanoparticles were composed of influenza nucleoprotein (NP) cores and hemagglutinin (HA) or matrix 2 protein ectodomain (M2e) shells. Vaccination with the nanoparticles significantly enhanced M2e-specific serum antibody titers and concomitant ADCC responses. Robust NP-specific T cell responses and robust HA neutralization were also detected. Moreover, vaccination with a trivalent nanoparticle combination containing two routinely circulated HA, conserved M2e, and NP reduced lung virus titers, pulmonary pathologies, and weight loss after homologous virus challenge. This combination also improved survival rates against heterologous and heterosubtypic influenza virus challenges. Our results demonstrate that the trivalent combination elicited potent and long-lasting immune responses conferring influenza viral cross-protection.
Collapse
Affiliation(s)
- Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Gilbert X Gonzalez
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Yufeng Song
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| |
Collapse
|
31
|
Zheng MZM, Wakim LM. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022; 15:379-388. [PMID: 34671115 PMCID: PMC8526531 DOI: 10.1038/s41385-021-00461-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/04/2023]
Abstract
Owing to their capacity to rapidly spread across the population, airborne pathogens represent a significant risk to global health. Indeed, several of the past major global pandemics have been instigated by respiratory pathogens. A greater understanding of the immune cells tasked with protecting the airways from infection will allow for the development of strategies that curb the spread and impact of these airborne diseases. A specific subset of memory T-cell resident in both the upper and lower respiratory tract, termed tissue-resident memory (Trm), have been shown to play an instrumental role in local immune responses against a wide breadth of both viral and bacterial infections. In this review, we discuss factors that influence respiratory tract Trm development, longevity, and immune surveillance and explore vaccination regimes that harness these cells, such approaches represent exciting new strategies that may be utilized to tackle the next global pandemic.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
32
|
Ruan S, Zhang Y, Feng N. Microneedle-mediated transdermal nanodelivery systems: a review. Biomater Sci 2021; 9:8065-8089. [PMID: 34752590 DOI: 10.1039/d1bm01249e] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The greatest limitation in the development of transdermal drug delivery systems is that only a few drugs can permeate the skin due to the barrier function of the stratum corneum. Active and passive methods are generally available for improving the ability of drug transdermal delivery. However, nanoparticles, as a passive approach, exhibit capacity-constrained permeation enhancement. Thus, microneedle-mediated nanoparticles possess enormous potential and broad prospects. Microneedles promote the penetration of macromolecules by creating microchannels on the skin surface. In this review, the prevailing subknowledge on microneedles (mechanism, classification, and applications of microneedles combined with nanoparticles) is discussed to provide a guideline for readers and a basic reference for further in-depth studies of this novel drug delivery system.
Collapse
Affiliation(s)
- Shuyao Ruan
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
33
|
Li S, Xia D, Prausnitz MR. Efficient Drug Delivery into Skin Using a Biphasic Dissolvable Microneedle Patch with Water-Insoluble Backing. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2103359. [PMID: 34744551 PMCID: PMC8570388 DOI: 10.1002/adfm.202103359] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Indexed: 06/13/2023]
Abstract
Dissolvable microneedle patches (MNPs) enable simplified delivery of therapeutics via the skin. However, most dissolvable MNPs do not deliver their full drug loading to the skin because only some of the drug is localized in the microneedles (MNs), and the rest remains adhered to the patch backing after removal from the skin. In this work, biphasic dissolvable MNPs are developed by mounting water-soluble MNs on a water-insoluble backing layer. These MNPs enable the drug to be contained in the MNs without migrating into the patch backing due to the inability of the drugs to partition into the hydrophobic backing materials during MNP fabrication. In addition, the insoluble backing is poorly wetted upon MN dissolution in the skin, which significantly reduces drug residue on the MNP backing surface after application. These effects enable a drug delivery efficiency of >90% from the MNPs into the skin 5 min after application. This study shows that the biphasic dissolvable MNPs can facilitate efficient drug delivery to the skin, which can improve the accuracy of drug dosing and reduce drug wastage.
Collapse
Affiliation(s)
- Song Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Dengning Xia
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
34
|
Polymeric microneedles for transdermal delivery of nanoparticles: Frontiers of formulation, sterility and stability aspects. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
35
|
Wilks LR, Joshi G, Grisham MR, Gill HS. Tyrosine-Based Cross-Linking of Peptide Antigens to Generate Nanoclusters with Enhanced Immunogenicity: Demonstration Using the Conserved M2e Peptide of Influenza A. ACS Infect Dis 2021; 7:2723-2735. [PMID: 34432416 DOI: 10.1021/acsinfecdis.1c00219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A method of creating nanoclusters (NCs) from soluble peptide molecules is described utilizing an approach based on a tyrosine-tyrosine cross-linking reaction. A reactive tag comprising histidine and tyrosine residues was introduced at the termini of the peptide molecules. The cross-linking reaction led to the creation of dityrosine bonds within the tag, which allowed for the generation of peptide NCs. We show that it is essential for the reactive tag to be present at both the "N" and "C" termini of the peptide for cluster formation to occur. Additionally, the cross-linking reaction was systematically characterized to show the importance of reaction conditions on final cluster diameter, allowing us to generate NCs of various sizes. To demonstrate the immunogenic potential of the peptide clusters, we chose to study the conserved influenza peptide, M2e, as the antigen. M2e NCs were formulated using the cross-linking reaction. We show the ability of the clusters to generate protective immunity in a dose, size, and frequency dependent manner against a lethal influenza A challenge in BALB/c mice. Taken together, the data presented suggest this new cluster formation technique can generate highly immunogenic peptide NCs in a simple and controllable manner.
Collapse
Affiliation(s)
- Logan R. Wilks
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Megan R. Grisham
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
36
|
Umar Y, Al-Batty S, Rahman H, Ashwaq O, Sarief A, Sadique Z, Sreekumar PA, Haque SKM. Polymeric Materials as Potential Inhibitors Against SARS-CoV-2. JOURNAL OF POLYMERS AND THE ENVIRONMENT 2021; 30:1244-1263. [PMID: 34518763 PMCID: PMC8426594 DOI: 10.1007/s10924-021-02272-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/22/2021] [Indexed: 05/02/2023]
Abstract
Recently discovered SARS-CoV-2 caused a pandemic that triggered researchers worldwide to focus their research on all aspects of this new peril to humanity. However, in the absence of specific therapeutic intervention, some preventive strategies and supportive treatment minimize the viral transmission as studied by some factors such as basic reproduction number, case fatality rate, and incubation period in the epidemiology of viral diseases. This review briefly discusses coronaviruses' life cycle of SARS-CoV-2 in a human host cell and preventive strategies at some selected source of infection. The antiviral activities of synthetic and natural polymers such as chitosan, hydrophobically modified chitosan, galactosylated chitosan, amine-based dendrimers, cyclodextrin, carrageenans, polyethyleneimine, nanoparticles are highlighted in this article. Mechanism of virus inhibition, detection and diagnosis are also presented. It also suggests that polymeric materials and nanoparticles can be effective as potential inhibitors and immunization against coronaviruses which would further develop new technologies in the field of polymer and nanoscience.
Collapse
Affiliation(s)
- Yunusa Umar
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Sirhan Al-Batty
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Habibur Rahman
- Department of General Studies, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Omar Ashwaq
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Abdulla Sarief
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Zakariya Sadique
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - P. A. Sreekumar
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - S. K. Manirul Haque
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| |
Collapse
|
37
|
Kang S, Kim Y, Shin Y, Song JJ, Jon S. Antigen-Presenting, Self-Assembled Protein Nanobarrels as an Adjuvant-Free Vaccine Platform against Influenza Virus. ACS NANO 2021; 15:10722-10732. [PMID: 34114799 DOI: 10.1021/acsnano.1c04078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Although naturally occurring, self-assembled protein nanoarchitectures have been utilized as antigen-delivery carriers, and the inability of such carriers to elicit immunogenicity requires additional use of strong adjuvants. Here, we report an immunogenic Brucella outer membrane protein BP26-derived nanoarchitecture displaying the influenza extracellular domain of matrix protein-2 (M2e) as a vaccine platform against influenza virus. Genetic engineering of a monomeric BP26 containing four or eight tandem repeats of M2e resulted in a hollow barrel-shaped nanoarchitecture (BP26-M2e nanobarrel). Immunization with BP26-M2e nanobarrels induced a strong M2e-specific humoral immune response in vivo that was much greater than that of a physical mixture of soluble M2e and BP26, with or without the use of an alum adjuvant. An anti-M2e antibody generated by BP26-M2e nanobarrel-immunized mice specifically bound to influenza virus-infected cells. Furthermore, in viral challenge tests, BP26-M2e nanobarrels effectively protected mice from influenza virus infection-associated death, even without the use of a conventional adjuvant. A mechanism study revealed that both M2e-specific antibody-dependent cellular cytotoxicity and T cell responses are involved in the vaccine efficacy of BP26-M2e nanobarrels. These findings suggest that the BP26-based nanobarrel developed here represents a versatile vaccine platform that can be used against various viral infections.
Collapse
Affiliation(s)
- Sukmo Kang
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Yujin Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Yumi Shin
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Ji-Joon Song
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| |
Collapse
|
38
|
Wang Y, Li S, Dong C, Ma Y, Song Y, Zhu W, Kim J, Deng L, Denning TL, Kang SM, Prausnitz MR, Wang BZ. Skin vaccination with dissolvable microneedle patches incorporating influenza neuraminidase and flagellin protein nanoparticles induces broad immune protection against multiple influenza viruses. ACS APPLIED BIO MATERIALS 2021; 4:4953-4961. [PMID: 34179728 PMCID: PMC8232372 DOI: 10.1021/acsabm.1c00240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We generated self-adjuvanted protein nanoparticles of conserved influenza antigens and immunized mice via skin vaccination with dissolvable microneedle patches (MNPs) to increase the strength and breadth of immune responses. We produced M2e nanoparticles via ethanol desolvation, and double-layered NA1/M2e (shell/core), NA1-FliC/M2e, NA2/M2e, and NA2-FliC/M2e protein nanoparticles by chemically crosslinking influenza NA and flagellin (FliC) onto the surfaces of the M2e nanoparticles. The resulting nanoparticles retained FliC TLR5 innate signaling activity and significantly increased antigen-uptake and dendritic cell maturation in vitro. We incorporated the nanoparticles into MNPs for skin vaccination in mice. The nanoparticle MNPs significantly increased M2e and NA-specific antibody levels, the numbers of germinal center B cells, and IL-4 positive splenocytes. Double-layered nanoparticle MNP skin vaccination protected mice against homologous and heterosubtypic influenza viruses. Our results demonstrated that MNP skin vaccination of NA-FliC/M2e nanoparticles could be developed into a standalone or synergistic component of a universal influenza vaccine strategy.
Collapse
Affiliation(s)
- Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Song Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Yufeng Song
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Lei Deng
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Timothy L. Denning
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Mark R. Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| |
Collapse
|
39
|
Guo X, Sun M, Gao R, Qu A, Chen C, Xu C, Kuang H, Xu L. Ultrasmall Copper (I) Sulfide Nanoparticles Prevent Hepatitis B Virus Infection. Angew Chem Int Ed Engl 2021; 60:13073-13080. [PMID: 33837622 DOI: 10.1002/anie.202103717] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 12/15/2022]
Abstract
Hepatitis B virus (HBV) poses a severe threat to public health and social development. Here, we synthesized 4±0.5 nm copper (I) sulfide (Cu2 S) nanoparticles (NPs) with 46 mdeg chiroptical property at 530 nm to selectively cleavage HBV core antigen (HBcAg) and effectively blocked HBV assembly and prevented HBV infection both in vitro and in vivo under light at 808 nm. Experimental analysis showed that the chiral Cu2 S NPs specific bound with the functional domain from phenylalanine23 (F23 ) to leucine30 (L30 ) from HBcAg primary sequence and the cutting site was between amino acid residues F24 and proline25 (P25 ). Under excitation at 808 nm, the intracellular HBcAg concentration was reduced by 95 %, and in HBV transgenic mice, the levels of HBV surface antigen (HBsAg) and HBV DNA were decreased by 93 % and 86 %, respectively. Together, these results reveal the potential nanomedicine for HBV control and provide fresh tools for viral infection.
Collapse
Affiliation(s)
- Xiao Guo
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Rui Gao
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Chen Chen
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| |
Collapse
|
40
|
Guo X, Sun M, Gao R, Qu A, Chen C, Xu C, Kuang H, Xu L. Ultrasmall Copper (I) Sulfide Nanoparticles Prevent Hepatitis B Virus Infection. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Xiao Guo
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Rui Gao
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Chen Chen
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection State Key Lab of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi Jiangsu 214122 P. R. China
| |
Collapse
|
41
|
Zeigler DF, Gage E, Clegg CH. Epitope-targeting platform for broadly protective influenza vaccines. PLoS One 2021; 16:e0252170. [PMID: 34043704 PMCID: PMC8158873 DOI: 10.1371/journal.pone.0252170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/10/2021] [Indexed: 11/18/2022] Open
Abstract
Seasonal influenza vaccines are often ineffective because they elicit strain-specific antibody responses to mutation-prone sites on the hemagglutinin (HA) head. Vaccines that provide long-lasting immunity to conserved epitopes are needed. Recently, we reported a nanoparticle-based vaccine platform produced by solid-phase peptide synthesis (SPPS) for targeting linear and helical protein-based epitopes. Here, we illustrate its potential for building broadly protective influenza vaccines. Targeting known epitopes in the HA stem, neuraminidase (NA) active site, and M2 ectodomain (M2e) conferred 50-75% survival against 5LD50 influenza B and H1N1 challenge; combining stem and M2e antigens increased survival to 90%. Additionally, protein sequence and structural information were employed in tandem to identify alternative epitopes that stimulate greater protection; we report three novel HA and NA sites that are highly conserved in type B viruses. One new target in the HA stem stimulated 100% survival, highlighting the value of this simple epitope discovery strategy. A candidate influenza B vaccine targeting two adjacent HA stem sites led to >104-fold reduction in pulmonary viral load. These studies describe a compelling platform for building vaccines that target conserved influenza epitopes.
Collapse
Affiliation(s)
- David F. Zeigler
- TRIA Bioscience Corp., Seattle, Washington, United States of America
| | - Emily Gage
- TRIA Bioscience Corp., Seattle, Washington, United States of America
| | | |
Collapse
|
42
|
Li S, Guo X, Gao R, Sun M, Xu L, Xu C, Kuang H. Recent Progress on Biomaterials Fighting against Viruses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005424. [PMID: 33644954 DOI: 10.1002/adma.202005424] [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: 08/10/2020] [Revised: 10/19/2020] [Indexed: 05/24/2023]
Abstract
Viruses not only pose severe threats to public health, but also influence the development of society. Over the past decade, rapid advances have been seen in the application of nanomaterials to virus research. As an interdisciplinary field, nanotechnology offers powerful functions because the structures of nanomaterials are unique, with remarkable physicochemical properties and excellent biocompatibility. Nanomaterials have been developed for virus detection and tracking and for antiviral strategies, to better understand viruses and reduce viral infections, implying a bright future for this field. Herein, the recent advances are systematically summarized regarding the nanomaterials used in viral studies. Representative applications of nanomaterials to viral detection and tracking are described. The antiviral effects achieved with nanomaterials based on different mechanisms are also described, including entry inhibition, inhibition of viral replication, and immunological enhancement. The current challenges and future opportunities in this promising field are also discussed.
Collapse
Affiliation(s)
- Si Li
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Xiao Guo
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Rui Gao
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Maozhong Sun
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Liguang Xu
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Chuanlai Xu
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Hua Kuang
- Key Laboratory of Synthetic and Biological Colloids, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| |
Collapse
|
43
|
O’Shea J, Prausnitz MR, Rouphael N. Dissolvable Microneedle Patches to Enable Increased Access to Vaccines against SARS-CoV-2 and Future Pandemic Outbreaks. Vaccines (Basel) 2021; 9:320. [PMID: 33915696 PMCID: PMC8066809 DOI: 10.3390/vaccines9040320] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 01/02/2023] Open
Abstract
Vaccines are an essential component of pandemic preparedness but can be limited due to challenges in production and logistical implementation. While vaccine candidates were rapidly developed against severe acute respiratory syndrome coronavirus 2 (SARS-COV-2), immunization campaigns remain an obstacle to achieving herd immunity. Dissolvable microneedle patches are advantageous for many possible reasons: improved immunogenicity; dose-sparing effects; expected low manufacturing cost; elimination of sharps; reduction of vaccine wastage; no need for reconstitution; simplified supply chain, with reduction of cold chain supply through increased thermostability; ease of use, reducing the need for healthcare providers; and greater acceptability compared to traditional hypodermic injections. When applied to coronavirus disease 2019 (COVID-19) and future pandemic outbreaks, microneedle patches have great potential to improve vaccination globally and save many lives.
Collapse
Affiliation(s)
- Jesse O’Shea
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, Atlanta, GA 30030, USA;
| | - Mark R. Prausnitz
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, Atlanta, GA 30030, USA;
| |
Collapse
|
44
|
Korkmaz E, Balmert SC, Sumpter TL, Carey CD, Erdos G, Falo LD. Microarray patches enable the development of skin-targeted vaccines against COVID-19. Adv Drug Deliv Rev 2021; 171:164-186. [PMID: 33539853 PMCID: PMC8060128 DOI: 10.1016/j.addr.2021.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is a serious threat to global health and the global economy. The ongoing race to develop a safe and efficacious vaccine to prevent infection by SARS-CoV-2, the causative agent for COVID-19, highlights the importance of vaccination to combat infectious pathogens. The highly accessible cutaneous microenvironment is an ideal target for vaccination since the skin harbors a high density of antigen-presenting cells and immune accessory cells with broad innate immune functions. Microarray patches (MAPs) are an attractive intracutaneous biocargo delivery system that enables safe, reproducible, and controlled administration of vaccine components (antigens, with or without adjuvants) to defined skin microenvironments. This review describes the structure of the SARS-CoV-2 virus and relevant antigenic targets for vaccination, summarizes key concepts of skin immunobiology in the context of prophylactic immunization, and presents an overview of MAP-mediated cutaneous vaccine delivery. Concluding remarks on MAP-based skin immunization are provided to contribute to the rational development of safe and effective MAP-delivered vaccines against emerging infectious diseases, including COVID-19.
Collapse
Affiliation(s)
- Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Stephen C Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
45
|
Lin PH, Liang CY, Yao BY, Chen HW, Pan CF, Wu LL, Lin YH, Hsu YS, Liu YH, Chen PJ, Hu CMJ, Yang HC. Robust induction of T RMs by combinatorial nanoshells confers cross-strain sterilizing immunity against lethal influenza viruses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:299-314. [PMID: 33898629 PMCID: PMC8047433 DOI: 10.1016/j.omtm.2021.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/17/2021] [Indexed: 01/01/2023]
Abstract
Antigen-specific lung-resident memory T cells (TRMs) constitute the first line of defense that mediates rapid protection against respiratory pathogens and inspires novel vaccine designs against infectious pandemic threats, yet effective means of inducing TRMs, particularly via non-viral vectors, remain challenging. Here, we demonstrate safe and potent induction of lung-resident TRMs using a biodegradable polymeric nanoshell that co-encapsulates antigenic peptides and TLR9 agonist CpG-oligodeoxynucleotide (CpG-ODN) in a virus-mimicking structure. Through subcutaneous priming and intranasal boosting, the combinatorial nanoshell vaccine elicits prominent lung-resident CD4+ and CD8+ T cells that surprisingly show better durability than live viral infections. In particular, nanoshells containing CpG-ODN and a pair of conserved class I and II major histocompatibility complex-restricted influenza nucleoprotein-derived antigenic peptides are demonstrated to induce near-sterilizing immunity against lethal infections with influenza A viruses of different strains and subtypes in mice, resulting in rapid elimination of replicating viruses. We further examine the pulmonary transport dynamic and optimal composition of the nanoshell vaccine conducive for robust TRM induction as well as the benefit of subcutaneous priming on TRM replenishment. The study presents a practical vaccination strategy for inducing protective TRM-mediated immunity, offering a compelling platform and critical insights in the ongoing quest toward a broadly protective vaccine against universal influenza as well as other respiratory pathogens.
Collapse
Affiliation(s)
- Pin-Hung Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chieh-Yu Liang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Yu Yao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Fu Pan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Ling Wu
- Institute of Physiology, National Yang-Ming Chiao-Tung University, Taipei City, Taiwan
| | - Yi-Hsuan Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Sung Hsu
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Han Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Yang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
46
|
Ding P, Zhang G, Chen Y, Liu H, Liu Y, Jia R, Wang Y, Li G, Wang A. Reasonable permutation of M2e enhances the effect of universal influenza nanovaccine. Int J Biol Macromol 2021; 173:244-250. [PMID: 33485888 DOI: 10.1016/j.ijbiomac.2021.01.132] [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] [Received: 09/27/2020] [Revised: 10/25/2020] [Accepted: 01/19/2021] [Indexed: 01/19/2023]
Abstract
Influenza A virus (IAV) occasionally cross-species transmission among humans, swine and avian. The ectodomain of matrix protein 2 (M2e) is highly conserved in IAV, and multi-copy M2e from different species are usually displayed on the surface of nanoparticles to improve immunogenicity and constitute universal IAV nanovaccines. In our previous study, three M2e were inserted into the C-terminal of Cap protein of porcine circovirus type 2 (PCV2) to form a universal nanovaccine that provides protection against PCV2 and different subtypes of IAV. However, M2e adopts at least two converted conformations, and the intermolecular linker of M2e enhances the conformational instability, which limits the recognition by B cell receptors and production of high-level antibodies. Here, we report that the permutation of M2e affects effectiveness of nanovaccines. Three M2e derived from humans, swine and avian IAV were inserted into the C-terminal of Cap protein to form nanovaccines. Immunoprotective effects of different M2e arrangements were explored in mice. Results showed that the M2e closest to the surface of nanoparticle induced the most efficient protection against IAV derived from corresponding species. The results will contribute to develop more effective PCV2 and universal IAV bivalent nanovaccines for pigs, as well as species-specific universal IAV vaccines.
Collapse
Affiliation(s)
- Peiyang Ding
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Gaping Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongliang Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Yunchao Liu
- Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Rui Jia
- Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Yanwei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Ge Li
- Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Aiping Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
47
|
van de Wall S, Badovinac VP, Harty JT. Influenza-Specific Lung-Resident Memory CD8 + T Cells. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a037978. [PMID: 33288540 PMCID: PMC7849341 DOI: 10.1101/cshperspect.a037978] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite the availability of seasonal vaccines, influenza A (IAV) prevails as a leading cause of respiratory infection worldwide. Current vaccination efforts aim at increasing protection against heterologous and potentially pandemic IAV strains. Lung-resident CD8+ T cells (Trm) generated upon IAV infection are vital for heterosubtypic immunity to IAV reexposure and provide quick and robust responses upon reactivation. Yet, protection wanes with time as lung Trm cell numbers decline, a contrasting feature with Trm cells at other mucosal sites such as the skin. In this review, we discuss current data on lung Trm compared to Trm cells in other tissues. Furthermore, major knowledge gaps in the generation and maintenance of IAV-specific lung Trm are addressed and mechanisms that may contribute to their decline are discussed. Further understanding in the mechanisms that govern effector function versus immunopathology is paramount for future IAV vaccine design in enhancing durability of lung Trm cells.
Collapse
Affiliation(s)
- Stephanie van de Wall
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Vladimer P. Badovinac
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;,Department of Pathology,, University of Iowa, Iowa City, Iowa 52242, USA;,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | - John T. Harty
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;,Department of Pathology,, University of Iowa, Iowa City, Iowa 52242, USA;,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
48
|
Minakshi P, Ghosh M, Kumar R, Brar B, Lambe UP, Banerjee S, Ranjan K, Kumar B, Goel P, Malik YS, Prasad G. An Insight into Nanomedicinal Approaches to Combat Viral Zoonoses. Curr Top Med Chem 2021; 20:915-962. [PMID: 32209041 DOI: 10.2174/1568026620666200325114400] [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: 12/04/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Emerging viral zoonotic diseases are one of the major obstacles to secure the "One Health" concept under the current scenario. Current prophylactic, diagnostic and therapeutic approaches often associated with certain limitations and thus proved to be insufficient for customizing rapid and efficient combating strategy against the highly transmissible pathogenic infectious agents leading to the disastrous socio-economic outcome. Moreover, most of the viral zoonoses originate from the wildlife and poor knowledge about the global virome database renders it difficult to predict future outbreaks. Thus, alternative management strategy in terms of improved prophylactic vaccines and their delivery systems; rapid and efficient diagnostics and effective targeted therapeutics are the need of the hour. METHODS Structured literature search has been performed with specific keywords in bibliographic databases for the accumulation of information regarding current nanomedicine interventions along with standard books for basic virology inputs. RESULTS Multi-arrayed applications of nanomedicine have proved to be an effective alternative in all the aspects regarding the prevention, diagnosis, and control of zoonotic viral diseases. The current review is focused to outline the applications of nanomaterials as anti-viral vaccines or vaccine/drug delivery systems, diagnostics and directly acting therapeutic agents in combating the important zoonotic viral diseases in the recent scenario along with their potential benefits, challenges and prospects to design successful control strategies. CONCLUSION This review provides significant introspection towards the multi-arrayed applications of nanomedicine to combat several important zoonotic viral diseases.
Collapse
Affiliation(s)
- Prasad Minakshi
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Mayukh Ghosh
- Department of Veterinary Physiology and Biochemistry, RGSC, Banaras Hindu University, Mirzapur (UP) - 231001, India
| | - Rajesh Kumar
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Basanti Brar
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Upendra P Lambe
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Somesh Banerjee
- Department of Veterinary Microbiology, Immunology Section, LUVAS, Hisar-125004, India
| | - Koushlesh Ranjan
- Department of Veterinary Physiology and Biochemistry, Sardar Vallabhbhai Patel University of Agriculture and Technology, Meerut, 250110, India
| | | | - Parveen Goel
- Department of Veterinary Medicine, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India
| | - Yashpal S Malik
- Division of Standardisation, Indian Veterinary Research Institute Izatnagar - Bareilly (UP) - 243122, India
| | - Gaya Prasad
- Sardar Vallabhbhai Patel University of Agriculture and Technology, Meerut, UP, 250110, India
| |
Collapse
|
49
|
Zhu W, Dong C, Wei L, Wang BZ. Promising Adjuvants and Platforms for Influenza Vaccine Development. Pharmaceutics 2021; 13:pharmaceutics13010068. [PMID: 33430259 PMCID: PMC7825707 DOI: 10.3390/pharmaceutics13010068] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Influenza is one of the major threats to public health. Current influenza vaccines cannot provide effective protection against drifted or shifted influenza strains. Researchers have considered two important strategies to develop novel influenza vaccines with improved immunogenicity and broader protective efficacy. One is applying fewer variable viral antigens, such as the haemagglutinin stalk domain. The other is including adjuvants in vaccine formulations. Adjuvants are promising and helpful boosters to promote more rapid and stronger immune responses with a dose-sparing effect. However, few adjuvants are currently licensed for human influenza vaccines, although many potential candidates are in different trials. While many advantages have been observed using adjuvants in influenza vaccine formulations, an improved understanding of the mechanisms underlying viral infection and vaccination-induced immune responses will help to develop new adjuvant candidates. In this review, we summarize the works related to adjuvants in influenza vaccine research that have been used in our studies and other laboratories. The review will provide perspectives for the utilization of adjuvants in developing next-generation and universal influenza vaccines.
Collapse
|
50
|
Protein and Peptide Nanocluster Vaccines. Curr Top Microbiol Immunol 2020. [PMID: 33165870 DOI: 10.1007/82_2020_228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Recombinant protein- and peptide-based vaccines can deliver large amounts of specific antigens for tailored immune responses. One class of these are protein and peptide nanoclusters (PNCs), which are made entirely from the crosslinked antigen. PNCs leverage the inherent immunogenicity of nanoparticulate antigens while minimizing the use of excipients normally used to create them. In this chapter, we discuss PNC fabrication methods, immunostimulatory properties of nanoclusters observed in vitro and in vivo, and protective benefits of PNC vaccines against influenza and cancer mouse models. We conclude with an outlook on future studies of PNCs and PNC design strategies, as well as their use in future vaccine formulations.
Collapse
|