1
|
Jha K, Jaishwal P, Yadav TP, Singh SP. Self-assembling of coiled-coil peptides into virus-like particles: Basic principles, properties, design, and applications with special focus on vaccine design and delivery. Biophys Chem 2025; 318:107375. [PMID: 39674128 DOI: 10.1016/j.bpc.2024.107375] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Self-assembling peptide nanoparticles (SAPN) based delivery systems, including virus-like particles (VLP), have shown great potential for becoming prominent in next-generation vaccine and drug development. The VLP can mimic properties of natural viral capsid in terms of size (20-200 nm), geometry (i.e., icosahedral structures), and the ability to generate a robust immune response (with multivalent epitopes) through activation of innate and/or adaptive immune signals. In this regard, coiled-coil (CC) domains are suitable building blocks for designing VLP because of their programmable interaction specificity, affinity, and well-established sequence-to-structure relationships. Generally, two CC domains with different oligomeric states (trimer and pentamer) are fused to form a monomeric protein through a short, flexible spacer sequence. By using combinations of symmetry axes (2-, 3- and 5- folds) that are unique to the geometry of the desired protein cage, it is possible, in principle, to assemble well-defined protein cages like VLP. In this review, we have discussed the crystallographic rules and the basic principles involved in the design of CC-based VLP. It also explored the functions of numerous noncovalent interactions in generating stable VLP structures, which play a crucial role in improving the properties of vaccine immunogenicity, drug delivery, and 3D cell culturing.
Collapse
Affiliation(s)
- Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Thakur Prasad Yadav
- Department of Physics, Faculty of Science, University of Allahabad, Prayagraj 211002, India.
| | | |
Collapse
|
2
|
Park HI, Sung AY. Establishment of Optimal Drug Delivery System and Evaluation of Utilization of Hydrogel Contact Lens According to the Addition Method of Tretinoin and Bovine Serum Albumin. Polymers (Basel) 2025; 17:159. [PMID: 39861232 PMCID: PMC11768321 DOI: 10.3390/polym17020159] [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: 12/17/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
This study aims to build an optimal drug delivery system by manufacturing and evaluating a hydrogel contact lens using Tretinoin (ATRA) and protein nanoparticles to improve the drug delivery system as an ophthalmic medical contact lens. To evaluate the optical and physical properties of the manufactured lens, the spectral transmittance, refractive index, water content, contact angle, AFM, tensile strength, drug delivery, and antibacterial properties were analyzed. The contact lens was manufactured to contain ATRA and bovine serum albumin (BSA) in different ways, and the results confirmed that A, B, and C each had different physical properties. In particular, for Sample A, using the soak and release method and using ATRA solution in the contact lens with BSA added, the wettability was 55.94°, the tensile strength was 0.1491 kgf/mm2, and drug delivery released 130.35 μm over 336 h, which was found to be superior to samples B and C. Therefore, the three hydrogel contact lenses compared in this study according to the addition method of ATRA and BSA can be used in various ways to build an optimal drug delivery system that is very useful as an ophthalmic medical lens.
Collapse
Affiliation(s)
| | - A-Young Sung
- Department of Optometry & Vision Science, Daegu Catholic University, Gyeongsan 38430, Republic of Korea;
| |
Collapse
|
3
|
Zuckermann FA, Grinkova YV, Husmann RJ, Pires-Alves M, Storms S, Chen WY, Sligar SG. An effective vaccine against influenza A virus based on the matrix protein 2 (M2). Vet Microbiol 2024; 298:110245. [PMID: 39293153 PMCID: PMC11758941 DOI: 10.1016/j.vetmic.2024.110245] [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: 03/19/2024] [Revised: 07/14/2024] [Accepted: 08/30/2024] [Indexed: 09/20/2024]
Abstract
The ever-increasing antigenic diversity of the hemagglutinin (HA) of influenza A virus (IAV) poses a significant challenge for effective vaccine development. Notably, the matrix protein 2 (M2) is a highly conserved 97 amino acid long transmembrane tetrameric protein present in the envelope of IAV. More than 99 % of IAV strains circulating in American swine herds share the identical pandemic (pdm) isoform of M2, making it an ideal target antigen for a vaccine that could elicit broadly protective immunity. Here, using soluble nanoscale membrane assemblies termed nanodiscs (NDs), we designed this membrane mimetic nanostructures displaying full-length M2 in its natural transmembrane configuration (M2ND). Intramuscular (IM) immunization of swine with M2ND mixed with conventional emulsion adjuvant elicited M2-specific IgG antibodies in the serum that recognized influenza virions and M2-specific interferon-γ secreting cells present in the blood. Intranasal (IN) immunization with M2ND adjuvanted with a mycobacterial extract elicited M2-specific IgA in mucosal secretions that also recognized IAV. Immunization with an influenza whole inactivated virus (WIV) vaccine supplemented with a concurrent IM injection of M2ND mixed with an emulsion adjuvant increased the level of protective immunity afforded by the former against a challenge with an antigenically distinct H3N2 IAV, as exhibited by an enhanced elimination of virus from the lung. The lone IM administration of the M2ND vaccine mixed with an emulsion adjuvant provided measurable protection as evidenced by a >10-fold reduction or complete elimination of the challenge virus from the lung, but it did not diminish the viral load in nasal secretions nor the extent of pneumonia that ensued after the virus challenge. In contrast, an improved formulation of the M2ND vaccine that incorporated synthetic CpG oligodeoxynucleotides (CpG-ODN) in the nanostructures administered alone, via the IN and IM routes combined, provided a significant level of protective immunity against IAV as evidenced by a decreased viral load in both the upper and lower respiratory tracts and fully eliminated the occurrence of pneumonia in 89 % of the pigs immunized with this biologic. Notably, to be effective, the M2 protein must be displayed in the ND assemblies, as shown by the observation that simply mixing M2 with empty NDs incorporating CpG-ODN (eND-CpG-ODN) did not provide protective immunity. This novel M2-based vaccine offers great promise to help increase the breadth of protection afforded by conventional WIV vaccines against the diversity of IAV in circulation and, plausibly, as a broadly protective stand-alone biologic.
Collapse
Affiliation(s)
- Federico A Zuckermann
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA.
| | - Yelena V Grinkova
- Department of Biochemistry, 505 South Goodwin Avenue, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Robert J Husmann
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Melissa Pires-Alves
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Suzanna Storms
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Wei-Yu Chen
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Stephen G Sligar
- Department of Biochemistry, 505 South Goodwin Avenue, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
4
|
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
|
5
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
| | | | | | - Weiqi Hong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
6
|
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
|
7
|
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: 3] [Impact Index Per Article: 1.0] [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
|
8
|
Mamuti M, Chen W, Jiang X. Nanotechnology‐Assisted Immunoengineering for Cancer Vaccines. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Muhetaerjiang Mamuti
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| |
Collapse
|
9
|
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
|
10
|
Danielsen M, Hempel C, Andresen TL, Urquhart AJ. Biopharmaceutical nanoclusters: Towards the self-delivery of protein and peptide therapeutics. J Control Release 2022; 347:282-307. [PMID: 35513210 DOI: 10.1016/j.jconrel.2022.04.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/27/2022]
Abstract
Protein and peptide biopharmaceuticals have had a major impact on the treatment of a number of diseases. There is a growing interest in overcoming some of the challenges associated with biopharmaceuticals, such as rapid degradation in physiological fluid, using nanocarrier delivery systems. Biopharmaceutical nanoclusters (BNCs) where the therapeutic protein or peptide is clustered together to form the main constituent of the nanocarrier system have the potential to mimic the benefits of more established nanocarriers (e.g., liposomal and polymeric systems) whilst eliminating the issue of low drug loading and potential side effects from additives. These benefits would include enhanced stability, improved absorption, and increased biopharmaceutical activity. However, the successful development of BNCs is challenged by the physicochemical complexity of the protein and peptide constituents as well as the dynamics of clustering. Here, we present and discuss common methodologies for the synthesis of therapeutic protein and peptide nanoclusters, as well as review the current status of this emerging field.
Collapse
Affiliation(s)
- Mia Danielsen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Casper Hempel
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Andrew J Urquhart
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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: 7] [Impact Index Per Article: 1.8] [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
|
14
|
Mytle N, Leyrer S, Inglefield JR, Harris AM, Hickey TE, Minang J, Lu H, Ma Z, Andersen H, Grubaugh ND, Guina T, Skiadopoulos MH, Lacy MJ. Influenza Antigens NP and M2 Confer Cross Protection to BALB/c Mice against Lethal Challenge with H1N1, Pandemic H1N1 or H5N1 Influenza A Viruses. Viruses 2021; 13:1708. [PMID: 34578289 PMCID: PMC8473317 DOI: 10.3390/v13091708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023] Open
Abstract
Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.
Collapse
Affiliation(s)
- Nutan Mytle
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Biomedical Advanced Research and Development Agency, U.S. Department of Health and Human Services, Washington, DC 20201, USA
| | - Sonja Leyrer
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Jon R. Inglefield
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Andrea M. Harris
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Thomas E. Hickey
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- National Cancer Institute, National Institutes of Health, Frederick, MD 20814, USA
| | - Jacob Minang
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Optimal Health Care, 11377 Robinwood Dr, Hagerstown, MD 21742, USA
| | - Hang Lu
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Zhidong Ma
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Hanné Andersen
- BIOQUAL, Inc., 12301 Parklawn Dr, Rockville, MD 20852, USA;
| | - Nathan D. Grubaugh
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06510, USA
| | - Tina Guina
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- AstraZeneca, Gaithersburg, MD 20878, USA
| | - Mario H. Skiadopoulos
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- U.S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J. Lacy
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| |
Collapse
|
15
|
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
|
16
|
Dong C, Wang Y, Gonzalez GX, Ma Y, Song Y, Wang S, Kang SM, Compans RW, Wang BZ. Intranasal vaccination with influenza HA/GO-PEI nanoparticles provides immune protection against homo- and heterologous strains. Proc Natl Acad Sci U S A 2021; 118:e2024998118. [PMID: 33941704 PMCID: PMC8126854 DOI: 10.1073/pnas.2024998118] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intranasal (i.n.) immunization is a promising vaccination route for infectious respiratory diseases such as influenza. Recombinant protein vaccines can overcome the safety concerns and long production phase of virus-based influenza vaccines. However, soluble protein vaccines are poorly immunogenic if administered by an i.n. route. Here, we report that polyethyleneimine-functionalized graphene oxide nanoparticles (GP nanoparticles) showed high antigen-loading capacities and superior immunoenhancing properties. Via a facile electrostatic adsorption approach, influenza hemagglutinin (HA) was incorporated into GP nanoparticles and maintained structural integrity and antigenicity. The resulting GP nanoparticles enhanced antigen internalization and promoted inflammatory cytokine production and JAWS II dendritic cell maturation. Compared with soluble HA, GP nanoparticle formulations induced significantly enhanced and cross-reactive immune responses at both systemic sites and mucosal surfaces in mice after i.n. immunization. In the absence of any additional adjuvant, the GP nanoparticle significantly boosted antigen-specific humoral and cellular immune responses, comparable to the acknowledged potent mucosal immunomodulator CpG. The robust immune responses conferred immune protection against challenges by homologous and heterologous viruses. Additionally, the solid self-adjuvant effect of GP nanoparticles may mask the role of CpG when coincorporated. In the absence of currently approved mucosal adjuvants, GP nanoparticles can be developed into potent i.n. influenza vaccines, providing broad protection. With versatility and flexibility, the GP nanoplatform can be easily adapted for constructing mucosal vaccines for different respiratory pathogens.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Cell Line
- Cross Reactions/immunology
- Cytokines/immunology
- Cytokines/metabolism
- Female
- Graphite/chemistry
- Graphite/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Immunity, Mucosal/drug effects
- Immunity, Mucosal/immunology
- Influenza A Virus, H3N2 Subtype/drug effects
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/chemistry
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice, Inbred BALB C
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Oligodeoxyribonucleotides/chemistry
- Oligodeoxyribonucleotides/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Polyethyleneimine/chemistry
- Vaccination/methods
- Mice
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302
| | - Ye Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302
| | - Gilbert X Gonzalez
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302
| | - Yao Ma
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302
| | - Yufeng Song
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302
| | - Shelly Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Emory University, Atlanta, GA 30322
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, Emory University, Atlanta, GA 30322
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302;
| |
Collapse
|
17
|
Khramtsov P, Kalashnikova T, Bochkova M, Kropaneva M, Timganova V, Zamorina S, Rayev M. Measuring the concentration of protein nanoparticles synthesized by desolvation method: Comparison of Bradford assay, BCA assay, hydrolysis/UV spectroscopy and gravimetric analysis. Int J Pharm 2021; 599:120422. [PMID: 33647407 DOI: 10.1016/j.ijpharm.2021.120422] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/04/2021] [Accepted: 02/21/2021] [Indexed: 12/12/2022]
Abstract
The desolvation technique is one of the most popular methods for preparing protein nanoparticles for medicine, biotechnology, and food applications. We fabricated 11 batches of BSA nanoparticles and 2 batches of gelatin nanoparticles by desolvation method. BSA nanoparticles from 2 batches were cross-linked by heating at +70 °C for 2 h; other nanoparticles were stabilized by glutaraldehyde. We compared several analytical approaches to measuring their concentration: gravimetric analysis, bicinchoninic acid assay, Bradford assay, and alkaline hydrolysis combined with UV spectroscopy. We revealed that the cross-linking degree and method of cross-linking affect both Bradford and BCA assay. Direct measurement of protein concentration in the suspension of purified nanoparticles by dye-binding assays can lead to significant (up to 50-60%) underestimation of nanoparticle concentration. Quantification of non-desolvated protein (indirect method) is affected by the presence of small nanoparticles in supernatants and can be inaccurate when the yield of desolvation is low. The reaction of cross-linker with protein changes UV absorbance of the latter. Therefore pure protein solution is an inappropriate calibrator when applying UV spectroscopy for the determination of nanoparticle concentration. Our recommendation is to determine the concentration of protein nanoparticles by at least two different methods, including gravimetric analysis.
Collapse
Affiliation(s)
- Pavel Khramtsov
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia.
| | - Tatyana Kalashnikova
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Maria Bochkova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Maria Kropaneva
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Valeria Timganova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Svetlana Zamorina
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Mikhail Rayev
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| |
Collapse
|
18
|
Park BR, Kim KH, Kotomina T, Kim MC, Kwon YM, Jeeva S, Jung YJ, Bhatnagar N, Isakova-Sivak I, Mezhenskaya D, Rudenko L, Wang BZ, Kang SM. Broad cross protection by recombinant live attenuated influenza H3N2 seasonal virus expressing conserved M2 extracellular domain in a chimeric hemagglutinin. Sci Rep 2021; 11:4151. [PMID: 33603072 PMCID: PMC7893060 DOI: 10.1038/s41598-021-83704-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/03/2021] [Indexed: 12/24/2022] Open
Abstract
Hemagglutinin (HA)-based current vaccines provide suboptimum cross protection. Influenza A virus contains an ion channel protein M2 conserved extracellular domain (M2e), a target for developing universal vaccines. Here we generated reassortant influenza virus rgH3N2 4xM2e virus (HA and NA from A/Switzerland/9715293/2013/(H3N2)) expressing chimeric 4xM2e-HA fusion proteins with 4xM2e epitopes inserted into the H3 HA N-terminus. Recombinant rgH3N2 4xM2e virus was found to retain equivalent growth kinetics as rgH3N2 in egg substrates. Intranasal single inoculation of mice with live rgH3N2 4xM2e virus was effective in priming the induction of M2e specific IgG antibody responses in mucosal and systemic sites as well as T cell responses. The rgH3N2 4xM2e primed mice were protected against a broad range of different influenza A virus subtypes including H1N1, H3N2, H5N1, H7N9, and H9N2. The findings support a new approach to improve the efficacy of current vaccine platforms by recombinant influenza virus inducing immunity to HA and cross protective M2e antigens.
Collapse
Affiliation(s)
- Bo Ryoung Park
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ki-Hye Kim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Min-Chul Kim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
- CARESIDE Co., Ltd., Seongnam, Gyeonggi-do, Republic of Korea
| | - Young-Man Kwon
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Subbiah Jeeva
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Noopur Bhatnagar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Bao-Zhong Wang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
19
|
Cossette B, Kelly SH, Collier JH. Intranasal Subunit Vaccination Strategies Employing Nanomaterials and Biomaterials. ACS Biomater Sci Eng 2020; 7:1765-1779. [DOI: 10.1021/acsbiomaterials.0c01291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Benjamin Cossette
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| | - Sean H. Kelly
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| | - Joel H. Collier
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, North Carolina 27708, United States
| |
Collapse
|
20
|
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
|
21
|
Tang S, Zhu W, Wang BZ. Influenza Vaccines toward Universality through Nanoplatforms and Given by Microneedle Patches. Viruses 2020; 12:E1212. [PMID: 33114336 PMCID: PMC7690886 DOI: 10.3390/v12111212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022] Open
Abstract
Influenza is one of the top threats to public health. The best strategy to prevent influenza is vaccination. Because of the antigenic changes in the major surface antigens of influenza viruses, current seasonal influenza vaccines need to be updated every year to match the circulating strains and are suboptimal for protection. Furthermore, seasonal vaccines do not protect against potential influenza pandemics. A universal influenza vaccine will eliminate the threat of both influenza epidemics and pandemics. Due to the massive challenge in realizing influenza vaccine universality, a single vaccine strategy cannot meet the need. A comprehensive approach that integrates advances in immunogen designs, vaccine and adjuvant nanoplatforms, and vaccine delivery and controlled release has the potential to achieve an effective universal influenza vaccine. This review will summarize the advances in the research and development of an affordable universal influenza vaccine.
Collapse
Affiliation(s)
| | | | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA; (S.T.); (W.Z.)
| |
Collapse
|
22
|
Tsoras AN, Wong KM, Paravastu AK, Champion JA. Rational Design of Antigen Incorporation Into Subunit Vaccine Biomaterials Can Enhance Antigen-Specific Immune Responses. Front Immunol 2020; 11:1547. [PMID: 32849524 PMCID: PMC7396695 DOI: 10.3389/fimmu.2020.01547] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/11/2020] [Indexed: 12/29/2022] Open
Abstract
Peptide subunit vaccines increase safety by reducing the risk of off-target responses and improving the specificity of the induced adaptive immune response. The immunogenicity of most soluble peptides, however, is often insufficient to produce robust and lasting immunity. Many biomaterials and delivery vehicles have been developed for peptide antigens to improve immune response while maintaining specificity. Peptide nanoclusters (PNC) are a subunit peptide vaccine material that has shown potential to increase immunogenicity of peptide antigens. PNC are comprised only of crosslinked peptide antigen and have been synthesized from several peptide antigens as small as 8 amino acids in length. However, as with many peptide vaccine biomaterials, synthesis requires adding residues to the peptide and/or engaging amino acids within the antigen epitope covalently to form a stable material. The impact of antigen modifications made to enable biomaterial incorporation or formation is rarely investigated, since the goal of most studies is to compare the soluble antigen with biomaterial form of antigen. This study investigates PNC as a platform vaccine biomaterial to evaluate how peptide modification and biomaterial formation with different crosslinking chemistries affect epitope-specific immune cell presentation and activation. Several types of PNC were synthesized by desolvation from the model peptide epitope SIINFEKL, which is derived from the immunogenic protein ovalbumin. SIINFEKL was altered to include extra residues on each end, strategically chosen to enable multiple conjugation chemistry options for incorporation into PNC. Several crosslinking methods were used to control which functional groups were used to stabilize the PNC, as well as the reducibility of the crosslinking. These variations were evaluated for immune responses and biodistribution following in vivo immunization. All modified antigen formulations still induced comparable immune responses when incorporated into PNC compared to unmodified soluble antigen alone. However, some crosslinking methods led to a significant increase in desirable immune responses while others did not, suggesting that not all PNC were processed the same. These results help guide future peptide vaccine biomaterial design, including PNC and a wide variety of conjugated and self-assembled peptide antigen materials, to maximize and tune the desired immune response.
Collapse
Affiliation(s)
| | | | | | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Atlanta, GA, United States
| |
Collapse
|
23
|
Zottig X, Côté-Cyr M, Arpin D, Archambault D, Bourgault S. Protein Supramolecular Structures: From Self-Assembly to Nanovaccine Design. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1008. [PMID: 32466176 PMCID: PMC7281494 DOI: 10.3390/nano10051008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
Life-inspired protein supramolecular assemblies have recently attracted considerable attention for the development of next-generation vaccines to fight against infectious diseases, as well as autoimmune diseases and cancer. Protein self-assembly enables atomic scale precision over the final architecture, with a remarkable diversity of structures and functionalities. Self-assembling protein nanovaccines are associated with numerous advantages, including biocompatibility, stability, molecular specificity and multivalency. Owing to their nanoscale size, proteinaceous nature, symmetrical organization and repetitive antigen display, protein assemblies closely mimic most invading pathogens, serving as danger signals for the immune system. Elucidating how the structural and physicochemical properties of the assemblies modulate the potency and the polarization of the immune responses is critical for bottom-up design of vaccines. In this context, this review briefly covers the fundamentals of supramolecular interactions involved in protein self-assembly and presents the strategies to design and functionalize these assemblies. Examples of advanced nanovaccines are presented, and properties of protein supramolecular structures enabling modulation of the immune responses are discussed. Combining the understanding of the self-assembly process at the molecular level with knowledge regarding the activation of the innate and adaptive immune responses will support the design of safe and effective nanovaccines.
Collapse
Affiliation(s)
- Ximena Zottig
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H2L 2C4, Canada; (X.Z.); (M.C.-C.); (D.A.)
- The Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC G1V 0A6, Canada
- The Swine and Poultry Infectious Diseases Research Centre, CRIPA, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H2L 2C4, Canada; (X.Z.); (M.C.-C.); (D.A.)
- The Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC G1V 0A6, Canada
- The Swine and Poultry Infectious Diseases Research Centre, CRIPA, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Dominic Arpin
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H2L 2C4, Canada; (X.Z.); (M.C.-C.); (D.A.)
- The Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC G1V 0A6, Canada
- The Swine and Poultry Infectious Diseases Research Centre, CRIPA, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Denis Archambault
- The Swine and Poultry Infectious Diseases Research Centre, CRIPA, Saint-Hyacinthe, QC J2S 2M2, Canada
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC H2L 2C4, Canada
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC H2L 2C4, Canada; (X.Z.); (M.C.-C.); (D.A.)
- The Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC G1V 0A6, Canada
- The Swine and Poultry Infectious Diseases Research Centre, CRIPA, Saint-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
24
|
Bhardwaj P, Bhatia E, Sharma S, Ahamad N, Banerjee R. Advancements in prophylactic and therapeutic nanovaccines. Acta Biomater 2020; 108:1-21. [PMID: 32268235 PMCID: PMC7163188 DOI: 10.1016/j.actbio.2020.03.020] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Vaccines activate suitable immune responses to fight against diseases but can possess limitations such as compromised efficacy and immunogenic responses, poor stability, and requirement of adherence to multiple doses. ‘Nanovaccines’ have been explored to elicit a strong immune response with the advantages of nano-sized range, high antigen loading, enhanced immunogenicity, controlled antigen presentation, more retention in lymph nodes and promote patient compliance by a lower frequency of dosing. Various types of nanoparticles with diverse pathogenic or foreign antigens can help to overcome immunotolerance and alleviate the need of booster doses as required with conventional vaccines. Nanovaccines have the potential to induce both cell-mediated and antibody-mediated immunity and can render long-lasting immunogenic memory. With such properties, nanovaccines have shown high potential for the prevention of infectious diseases such as acquired immunodeficiency syndrome (AIDS), malaria, tuberculosis, influenza, and cancer. Their therapeutic potential has also been explored in the treatment of cancer. The various kinds of nanomaterials used for vaccine development and their effects on immune system activation have been discussed with special relevance to their implications in various pathological conditions. Statement of Significance Interaction of nanoparticles with the immune system has opened multiple avenues to combat a variety of infectious and non-infectious pathological conditions. Limitations of conventional vaccines have paved the path for nanomedicine associated benefits with a hope of producing effective nanovaccines. This review highlights the role of different types of nanovaccines and the role of nanoparticles in modulating the immune response of vaccines. The applications of nanovaccines in infectious and non-infectious diseases like malaria, tuberculosis, AIDS, influenza, and cancers have been discussed. It will help the readers develop an understanding of mechanisms of immune activation by nanovaccines and design appropriate strategies for novel nanovaccines.
Collapse
|
25
|
Tsoras AN, Champion JA. Protein and Peptide Biomaterials for Engineered Subunit Vaccines and Immunotherapeutic Applications. Annu Rev Chem Biomol Eng 2020; 10:337-359. [PMID: 31173518 DOI: 10.1146/annurev-chembioeng-060718-030347] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although vaccines have been the primary defense against widespread infectious disease for decades, there is a critical need for improvement to combat complex and variable diseases. More control and specificity over the immune response can be achieved by using only subunit components in vaccines. However, these often lack sufficient immunogenicity to fully protect, and conjugation or carrier materials are required. A variety of protein and peptide biomaterials have improved effectiveness and delivery of subunit vaccines for infectious, cancer, and autoimmune diseases. They are biodegradable and have control over both material structure and immune function. Many of these materials are built from naturally occurring self-assembling proteins, which have been engineered for incorporation of vaccine components. In contrast, others are de novo designs of structures with immune function. In this review, protein biomaterial design, engineering, and immune functionality as vaccines or immunotherapies are discussed.
Collapse
Affiliation(s)
- Alexandra N Tsoras
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-2000, USA;
| | - Julie A Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-2000, USA;
| |
Collapse
|
26
|
Wang Y, Deng L, Gonzalez GX, Luthra L, Dong C, Ma Y, Zou J, Kang SM, Wang BZ. Double-Layered M2e-NA Protein Nanoparticle Immunization Induces Broad Cross-Protection against Different Influenza Viruses in Mice. Adv Healthc Mater 2020; 9:e1901176. [PMID: 31840437 PMCID: PMC6980908 DOI: 10.1002/adhm.201901176] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/18/2019] [Indexed: 12/13/2022]
Abstract
The development of a universal influenza vaccine is an ideal strategy to eliminate public health threats from influenza epidemics and pandemics. This ultimate goal is restricted by the low immunogenicity of conserved influenza epitopes. Layered protein nanoparticles composed of well-designed conserved influenza structures have shown improved immunogenicity with new physical and biochemical features. Herein, structure-stabilized influenza matrix protein 2 ectodomain (M2e) and M2e-neuraminidase fusion (M2e-NA) recombinant proteins are generated and M2e protein nanoparticles and double-layered M2e-NA protein nanoparticles are produced by ethanol desolvation and chemical crosslinking. Immunizations with these protein nanoparticles induce immune protection against different viruses of homologous and heterosubtypic NA in mice. Double-layered M2e-NA protein nanoparticles induce higher levels of humoral and cellular responses compared with their comprising protein mixture or M2e nanoparticles. Strong cytotoxic T cell responses are induced in the layered M2e-NA protein nanoparticle groups. Antibody responses contribute to the heterosubtypic NA immune protection. The protective immunity is long lasting. These results demonstrate that double-layered protein nanoparticles containing structure-stabilized M2e and NA can be developed into a universal influenza vaccine or a synergistic component of such vaccines. Layered protein nanoparticles can be a general vaccine platform for different pathogens.
Collapse
Affiliation(s)
- Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | | | - Gilbert X. Gonzalez
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| | - Latika Luthra
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, 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
| | - Jun Zou
- 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
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Road, Atlanta, Georgia 30302, USA
| |
Collapse
|
27
|
Ding P, Jin Q, Zhou W, Chai Y, Liu X, Wang Y, Chen X, Guo J, Deng R, Gao GF, Zhang G. A Universal Influenza Nanovaccine for "Mixing Vessel" Hosts Confers Potential Ability to Block Cross-Species Transmission. Adv Healthc Mater 2019; 8:e1900456. [PMID: 31267679 DOI: 10.1002/adhm.201900456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/01/2019] [Indexed: 12/14/2022]
Abstract
Influenza A virus (IAV), a deadly zoonotic pathogen, poses a tremendous threat and burden to global health systems. Pigs act as "mixing vessel" hosts to support and generate new pandemic viruses. Preventing the spread of IAV in pigs effectively can delay or even block cross-species transmission. Universal vaccines based on the highly conserved ectodomain of influenza matrix protein 2 (M2e) have been widely reported, but have not been applied due to inadequate protection. Porcine circovirus type 2 (PCV2) causes immunosuppression and promotes swine influenza virus (SIV) infection. Here, M2e is inserted into capsid protein of PCV2 without burying the neutralizing epitopes and self-assembles to form a bivalent nanovaccine. Inoculation with the nanovaccine induces robust M2e- and PCV2-specific immune responses. The nanovaccine confers protection against lethal challenges of IAV from different species in mice, and significantly reduces SIV titers in pigs' respiratory tract and blocks SIV transmission. These results indicate that the nanovaccine is an economical and promising PCV2 and universal IAV bivalent vaccine, and it will synergistically and powerfully offer potential ability to block IAV cross-species reassortment and transmission.
Collapse
Affiliation(s)
- Peiyang Ding
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- College of Veterinary MedicineNorthwest A&F University Yangling 712100 China
| | - Qianyue Jin
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- Jiangsu Co‐Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou University Yangzhou 225009 China
| | - Wen Zhou
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- College of Veterinary MedicineNorthwest A&F University Yangling 712100 China
| | - Yongxiao Chai
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- College of Animal Science and Veterinary MedicineHenan Agricultural University Zhengzhou 450002 China
| | - Xiao Liu
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- College of Animal Science and Veterinary MedicineHenan Agricultural University Zhengzhou 450002 China
| | - Yao Wang
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- College of Animal Science and Veterinary MedicineHenan Agricultural University Zhengzhou 450002 China
| | - Xinxin Chen
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
| | - Junqing Guo
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
| | - Ruiguang Deng
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
| | - George F. Gao
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyInstitute of MicrobiologyChinese Academy of Sciences Beijing 100101 China
| | - Gaiping Zhang
- Henan Provincial Key Laboratory of Animal ImmunologyHenan Academy of Agricultural Sciences Zhengzhou 450002 China
- College of Veterinary MedicineNorthwest A&F University Yangling 712100 China
- Jiangsu Co‐Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou University Yangzhou 225009 China
- College of Animal Science and Veterinary MedicineHenan Agricultural University Zhengzhou 450002 China
| |
Collapse
|
28
|
Abstract
Annually recurring seasonal influenza causes massive economic loss and poses severe threats to public health worldwide. The current seasonal influenza vaccines are the most effective means of preventing influenza infections but possess major weaknesses. Seasonal influenza vaccines require annual updating of the vaccine strains. However, it is an unreachable task to accurately predict the future circulating strains. Vaccines with mismatched strains dramatically compromise the vaccine efficacy. In addition, the seasonal influenza vaccines are ineffective against an unpredictable pandemic. A universal influenza vaccine would overcome these weaknesses of the seasonal vaccines and abolish the threat of influenza pandemics. One approach under investigation is to design influenza vaccine immunogens based on conserved, type-specific amino acid sequences and conformational epitopes, rather than strain-specific. Such vaccines can elicit broadly reactive humoral and cellular immunity. Universal influenza vaccine development has intensively employed nanotechnology because the structural and morphological properties of nanoparticles dramatically improve vaccine immunogenicity and the induced immunity duration. Layered protein nanoparticles can decrease off-target immune responses, fine-tune antigen recognition and processing, and facilitate comprehensive immune response induction. Herein, we review the designs of effective nanoparticle universal influenza vaccines, the recent discoveries of specific nanoparticle features that contribute to immunogenicity enhancement, and recent progress in clinical trials.
Collapse
Affiliation(s)
- Lei Deng
- Center for Inflammation, Immunity & Infection, Georgia State University, 145 Piedmont Avenue SE, Atlanta, Georgia 30302-3965, United States
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, 145 Piedmont Avenue SE, Atlanta, Georgia 30302-3965, United States
| |
Collapse
|
29
|
Wang Y, Deng L, Kang SM, Wang BZ. Universal influenza vaccines: from viruses to nanoparticles. Expert Rev Vaccines 2018; 17:967-976. [PMID: 30365905 DOI: 10.1080/14760584.2018.1541408] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The current seasonal influenza vaccine confers only limited protection due to waning antibodies or the antigenic shift and drift of major influenza surface antigens. A universal influenza vaccine which induces broad cross-protection against divergent influenza viruses with a comparable or better efficacy to seasonal influenza vaccines against matched strains will negate the need for an annual update of vaccine strains and protect against possible influenza pandemics. AREAS COVERED In this review, we summarize the recent progress in nanoparticle-based universal influenza vaccine development. We compared the most potent nanoparticle categories, focusing on how they encapsulate conserved influenza epitopes, stimulate the innate and adaptive immune systems, exhibit antigen depot effect, extend the period for antigen-processing and presentation, and exert an intrinsic adjuvant effect on inducing robust immune responses. EXPERT COMMENTARY The development of an effective universal influenza vaccine is an urgent task. Traditional influenza vaccine approaches are not sufficient for preventing recurrent epidemics or occasional pandemics. Nanoparticles are compatible with different immunogens and immune stimulators and can overcome the intrinsically low immunogenicity of conserved influenza virus antigens. We foresee that an affordable universal influenza vaccine will be available within ten years by integrating nanoparticles with other targeted delivery and controlled release technology.
Collapse
Affiliation(s)
- Ye Wang
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| | - Lei Deng
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| | - Sang-Moo Kang
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| | - Bao-Zhong Wang
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| |
Collapse
|
30
|
Tsybalova LM, Stepanova LA, Shuklina MA, Mardanova ES, Kotlyarov RY, Potapchuk MV, Petrov SA, Blokhina EA, Ravin NV. Combination of M2e peptide with stalk HA epitopes of influenza A virus enhances protective properties of recombinant vaccine. PLoS One 2018; 13:e0201429. [PMID: 30138320 PMCID: PMC6107133 DOI: 10.1371/journal.pone.0201429] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
Background Influenza infection could be more effectively controlled if a multi-purpose vaccine with the ability to induce responses against most, or all, influenza A subtypes could be generated. Conserved viral proteins are a promising basis for the creation of a broadly protective vaccine. In the present study, the immunogenicity and protective properties of three recombinant proteins (vaccine candidates), comprising conserved viral proteins fused with bacterial flagellin, were compared. Methods Balb/c mice were immunized intranasally with recombinant proteins comprising either one viral protein (the ectodomain of the M2 protein, ‘M2e’) or two viral proteins (M2e and the hemagglutinin second subunit ‘HA2’ epitope) genetically fused with flagellin. Further, two different consensus variants of HA2 were used. Therefore, three experimental positives were used in addition to the negative control (Flg-his). The mucosal, humoral, and T-cell immune responses to these constructs were evaluated. Result We have demonstrated that insertion of the HA2 consensus polypeptide (aa 76–130), derived from either the first (HA2-1) or second (HA2-2) virus phylogenetic group, into the recombinant Flg4M2e protein significantly enhanced its immunogenicity and protective properties. Intranasal administration of the vaccine candidates (Flg-HA2-2-4M2e or Flg-HA2-1-4M2e) induced considerable mucosal and systemic responses directed at both the M2e-protein and, in general, the influenza A virus. However, the immune response elicited by the Flg-HA2-1-4M2e protein was weaker than the one generated by Flg-HA2-2-4M2e. These recombinant proteins containing both viral peptides provide complete protection from lethal challenge with various influenza viruses: A/H3N2; A/H2N2; and A/H5N1. Conclusion This study demonstrates that the intranasal administration of Flg-HA2-2-4M2e recombinant protein induces a strong immune response which provides broad protection against various influenza viruses. This construct is therefore a strong candidate for development as a universal vaccine.
Collapse
Affiliation(s)
- Liudmila M. Tsybalova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
- * E-mail:
| | - Liudmila A. Stepanova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Marina A. Shuklina
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Eugenia S. Mardanova
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Roman Y. Kotlyarov
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Marina V. Potapchuk
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Sergei A. Petrov
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Elena A. Blokhina
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Nikolai V. Ravin
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
31
|
Chang TZ, Deng L, Wang BZ, Champion JA. H7 Hemagglutinin nanoparticles retain immunogenicity after >3 months of 25°C storage. PLoS One 2018; 13:e0202300. [PMID: 30092060 PMCID: PMC6084952 DOI: 10.1371/journal.pone.0202300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/31/2018] [Indexed: 01/12/2023] Open
Abstract
Vaccine distribution infrastructure remains inadequate in many parts of the world, and it is estimated that up to 40–50% of all vaccine doses are wasted in certain countries. Vaccines that can maintain viability outside of the cold chain would decrease vaccine wastage and increase immunization rates in regions of the world with underdeveloped vaccine distribution infrastructure. We examined the potential of crosslinked protein nanoparticles, made from trimerized influenza hemagglutinin (3HA), to maintain immunogenicity after cold-chain-independent storage. We found that the nanoparticles could be stored for 112 days at room temperature without any loss in hemagglutinating activity or immunogenicity, and that nanoparticles could be stored at 37°C for 2 weeks without any loss in hemagglutinating activity. As vaccine development moves towards the use of recombinant subunit antigens, our results demonstrate the potential of crosslinked antigen nanoparticles as an immunogenic vehicle for bringing effective vaccines to underdeveloped regions outside of the cold chain.
Collapse
Affiliation(s)
- Timothy Z. Chang
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Lei Deng
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States of America
| | - Bao-Zhong Wang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States of America
| | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
32
|
Heterosubtypic influenza protection elicited by double-layered polypeptide nanoparticles in mice. Proc Natl Acad Sci U S A 2018; 115:E7758-E7767. [PMID: 30065113 DOI: 10.1073/pnas.1805713115] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Influenza is a persistent threat to public health. Here we report that double-layered peptide nanoparticles induced robust specific immunity and protected mice against heterosubtypic influenza A virus challenges. We fabricated the nanoparticles by desolvating a composite peptide of tandem copies of nucleoprotein epitopes into nanoparticles as cores and cross-linking another composite peptide of four tandem copies of influenza matrix protein 2 ectodomain epitopes to the core surfaces as a coating. Delivering the nanoparticles via dissolvable microneedle patch-based skin vaccination further enhanced the induced immunity. These peptide-only, layered nanoparticles demonstrated a strong antigen depot effect and migrated into spleens and draining (inguinal) lymph nodes for an extended period compared with soluble antigens. This increased antigen-presentation time correlated with the stronger immune responses in the nanoparticle-immunized group. The protection conferred by nanoparticle immunization was transferable by passive immune serum transfusion and depended partially on a functional IgG receptor FcγRIV. Using a conditional cell depletion, we found that CD8+ T cells were involved in the protection. The immunological potency and stability of the layered peptide nanoparticles indicate applications for other peptide-based vaccines and peptide drug delivery.
Collapse
|
33
|
Si Y, Wen Y, Kelly SH, Chong AS, Collier JH. Intranasal delivery of adjuvant-free peptide nanofibers elicits resident CD8 + T cell responses. J Control Release 2018; 282:120-130. [PMID: 29673645 PMCID: PMC6309200 DOI: 10.1016/j.jconrel.2018.04.031] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 04/03/2018] [Accepted: 04/15/2018] [Indexed: 12/11/2022]
Abstract
Influenza vaccines that can be administered intranasally or by other needle-free delivery routes have potential advantages over injected formulations in terms of patient compliance, cost, and ease of global distribution. Supramolecular peptide nanofibers have been investigated previously as platforms for vaccines and immunotherapies and have been shown to raise immune responses in the absence of exogenous adjuvants and without measurable inflammation. However, at present it has not been tested whether the immunogenicity of these materials extends to the intranasal route. Here we investigated the extent to which self-assembled peptide nanofibers bearing an influenza peptide epitope elicit antigen-specific CD8+ T cell responses when delivered intranasally, and we compared these responses with those elicited by subcutaneous immunization. Peptides containing an epitope from influenza acid polymerase (PA) and the Q11 self-assembly domain formed nanofibers that were avidly taken up by dendritic cells in lung-draining mediastinal lymph nodes after intranasal immunization. Intranasally delivered nanofibers generated greater antigen-specific CD8+ T cell responses in the lung-draining lymph nodes than subcutaneous immunizations while retaining the non-inflammatory character of the materials observed in other delivery sites. The CD8+ T cells elicited systemically were functional as assessed by their ability to produce IFN-γ ex vivo, lyse epitope-pulsed target cells in vivo, and diminish viral loads in infected mice. Compared to subcutaneously delivered nanofibers, intranasally delivered peptide nanofibers significantly increased the number of persisting antigen-specific tissue resident memory CD8+ T cells in the lung, allowing for a more rapid response to infection at 6 weeks post-vaccination. These results indicate that intranasally delivered self-assembled peptide nanofibers are immunogenic when delivering CD8+ epitopes without adjuvant or CD4+ epitopes, are non-inflammatory, and promote more lung-resident memory CD8+ T cells compared to subcutaneous immunization.
Collapse
Affiliation(s)
- Youhui Si
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Yi Wen
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Sean H Kelly
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| | - Joel H Collier
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
34
|
Tang L, Zheng Y, Melo MB, Mabardi L, Castaño AP, Xie YQ, Li N, Kudchodkar SB, Wong HC, Jeng EK, Maus MV, Irvine DJ. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat Biotechnol 2018; 36:707-716. [PMID: 29985479 PMCID: PMC6078803 DOI: 10.1038/nbt.4181] [Citation(s) in RCA: 466] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 06/07/2018] [Indexed: 12/24/2022]
Abstract
Adoptive cell therapy (ACT) with antigen-specific T cells has shown remarkable clinical success, but approaches to safely and effectively augment T cell function, especially in solid tumors, remain of great interest. Here we describe a strategy to “backpack” large quantities of supporting protein drugs on T cells using protein nanogels (NGs) that selectively release these cargos in response to T cell receptor (TCR) activation. We design cell surface-conjugated NGs that respond to an increase in T cell surface reduction potential upon antigen recognition, limiting drug release to sites of antigen encounter such as the tumor microenvironment. Using NGs carrying an IL-15 superagonist complex, we demonstrate that relative to systemic administration of free cytokines, NG delivery selectively expands T cells 16-fold in tumors, and allows at least 8-fold higher doses of cytokine to be administered without toxicity. The improved therapeutic window enables substantially increased tumor clearance by murine T cell and human CAR-T cell therapy in vivo.
Collapse
Affiliation(s)
- Li Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yiran Zheng
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mariane Bandeira Melo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Llian Mabardi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
| | - Ana P Castaño
- Cellular Immunotherapy Program, Massachusetts General Hospital (MGH) Cancer Center, Charlestown, Massachusetts, USA
| | - Yu-Qing Xie
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Na Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Hing C Wong
- Altor BioScience Corporation, Miramar, Florida, USA
| | - Emily K Jeng
- Altor BioScience Corporation, Miramar, Florida, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital (MGH) Cancer Center, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Darrell J Irvine
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
35
|
Dong X, Sun Z, Liang J, Wang H, Zhu D, Leng X, Wang C, Kong D, Lv F. A visible fluorescent nanovaccine based on functional genipin crosslinked ovalbumin protein nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1087-1098. [DOI: 10.1016/j.nano.2018.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/29/2018] [Accepted: 02/10/2018] [Indexed: 01/11/2023]
|
36
|
Sequential immunizations with a panel of HIV-1 Env virus-like particles coach immune system to make broadly neutralizing antibodies. Sci Rep 2018; 8:7807. [PMID: 29773829 PMCID: PMC5958130 DOI: 10.1038/s41598-018-25960-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/26/2018] [Indexed: 12/24/2022] Open
Abstract
Broadly neutralizing antibodies (bnAbs) are correlated with passive HIV/SHIV protection and are desirable components of a HIV protective immunity. In the current study, we have designed a sequential-immunization strategy with a panel of envelope glycoprotein (Env)-enriched virus-like particles (VLPs) from various HIV-1 clades (A-E) to elicit bnAbs with high breadth and potency of neutralization in rabbits. We have compared this regimen with repetitive immunizations of individual Env (subtype B) VLPs or a mixture of various Env VLPs. Our results demonstrate that the sequential immunization group of animals induced significantly higher IgG endpoint titers against respective HIV Env (autologous) antigen than other control groups. Animals vaccinated sequentially showed an increase in the antibody endpoint titers and IgG antibody secreting cells (ASCs) against Con-S Env protein. Sequential immunizations with various Env VLPs promoted antibody avidity indices and enhanced bnAb responses against a panel of HIV pseudotyped virions including some of the tier 3 pseudostrains. Sequential immunizations with various VLPs displaying "native-like" HIV-1 Envs elicited bnAb responses with increased breadth and potency of neutralization.
Collapse
|
37
|
Wang C, Zhu W, Luo Y, Wang BZ. Gold nanoparticles conjugating recombinant influenza hemagglutinin trimers and flagellin enhanced mucosal cellular immunity. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1349-1360. [PMID: 29649593 DOI: 10.1016/j.nano.2018.03.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/09/2018] [Accepted: 03/31/2018] [Indexed: 01/13/2023]
Abstract
The immunogenicity of subunit vaccines can be augmented by formulating them into nanoparticles. We conjugated recombinant trimetric influenza A/Aichi/2/68(H3N2) hemagglutinin (HA) onto functionalized gold nanoparticle (AuNP) surfaces in a repetitive, oriented configuration. To further improve the immunogenicity, we generated Toll-like receptor 5 (TLR5) agonist flagellin (FliC)-coupled AuNPs as particulate adjuvants. Intranasal immunizations with an AuNP-HA and AuNP-FliC particle mixture elicited strong mucosal and systemic immune responses that protected hosts against lethal influenza challenges. Compared with the AuNP-HA alone group, the addition of AuNP-FliC improved mucosal B cell responses as characterized by elevated influenza specific IgA and IgG levels in nasal, tracheal, and lung washes. AuNP-HA/AuNP-FliC also stimulated antigen-specific interferon-γ (IFN-γ)-secreting CD4+ cell proliferation and induced strong effector CD8+ T cell activation. Our results indicate that intranasal co-delivery of antigen and adjuvant-displaying AuNPs enhanced vaccine efficacy by inducing potent cellular immune responses.
Collapse
Affiliation(s)
- Chao Wang
- 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
| | - Yuan Luo
- 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
|
38
|
Qi M, Zhang XE, Sun X, Zhang X, Yao Y, Liu S, Chen Z, Li W, Zhang Z, Chen J, Cui Z. Intranasal Nanovaccine Confers Homo- and Hetero-Subtypic Influenza Protection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703207. [PMID: 29430819 DOI: 10.1002/smll.201703207] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/03/2018] [Indexed: 05/21/2023]
Abstract
Cross-protective and non-invasively administered vaccines are attractive and highly desired for the control of influenza. Self-assembling nanotechnology provides an opportunity for the development of vaccines with superior performance. In this study, an intranasal nanovaccine is developed targeting the conserved ectodomain of influenza matrix protein 2(M2e). 3-sequential repeats of M2e (3M2e) is presented on the self-assembling recombinant human heavy chain ferritin (rHF) cage to form the 3M2e-rHF nanoparticle. Intranasal vaccination with 3M2e-rHF nanoparticles in the absence of an adjuvant induces robust immune responses, including high titers of sera M2e-specific IgG antibodies, T-cell immune responses, and mucosal secretory-IgA antibodies in mice. The 3M2e-rHF nanoparticles also confer complete protection against a lethal infection of homo-subtypic H1N1 and hetero-subtypic H9N2 virus. An analysis of the mechanism of protection underlying the intranasal immunization with the 3M2e-rHF nanoparticle indicates that M2e-specific mucosal secretory-IgA and T-cell immune responses may play critical roles in the prevention of infection. The results suggest that the 3M2e-rHF nanoparticle is a promising, needle-free, intranasally administered, cross-protective influenza vaccine. The use of self-assembling nanovaccines could be an ideal strategy for developing vaccines with characteristics such as high immunogenicity, cross-protection, and convenient administration, as well as being economical and suitable for large-scale production.
Collapse
Affiliation(s)
- Mi Qi
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian-En Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianxun Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yanfeng Yao
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science and Technology, Wuhan, 430208, China
| | - Siling Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ze Chen
- College of Life Sciences, Hunan Normal University, Changsha, 410013, China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhiping Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jianjun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| |
Collapse
|
39
|
Tsoras AN, Champion JA. Cross-Linked Peptide Nanoclusters for Delivery of Oncofetal Antigen as a Cancer Vaccine. Bioconjug Chem 2018; 29:776-785. [DOI: 10.1021/acs.bioconjchem.8b00079] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Alexandra N. Tsoras
- School of Chemical & Biomolecular Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Julie A. Champion
- School of Chemical & Biomolecular Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
40
|
Double-layered protein nanoparticles induce broad protection against divergent influenza A viruses. Nat Commun 2018; 9:359. [PMID: 29367723 PMCID: PMC5783933 DOI: 10.1038/s41467-017-02725-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/20/2017] [Indexed: 01/22/2023] Open
Abstract
Current influenza vaccines provide limited protection against circulating influenza A viruses. A universal influenza vaccine will eliminate the intrinsic limitations of the seasonal flu vaccines. Here we report methodology to generate double-layered protein nanoparticles as a universal influenza vaccine. Layered nanoparticles are fabricated by desolvating tetrameric M2e into protein nanoparticle cores and coating these cores by crosslinking headless HAs. Representative headless HAs of two HA phylogenetic groups are constructed and purified. Vaccinations with the resulting protein nanoparticles in mice induces robust long-lasting immunity, fully protecting the mice against challenges by divergent influenza A viruses of the same group or both groups. The results demonstrate the importance of incorporating both structure-stabilized HA stalk domains and M2e into a universal influenza vaccine to improve its protective potency and breadth. These potent disassemblable protein nanoparticles indicate a wide application in protein drug delivery and controlled release. Relatively well conserved domains of influenza A virus (IAV) proteins are potential candidates for the development of a universal IAV vaccine. Here, Deng et al. combine two such conserved antigens (M2e and HA stalk) in a double-layered protein nanoparticle and show that it protects against divergent IAVs in mice.
Collapse
|
41
|
Wang L, Liu SY, Chen HW, Xu J, Chapon M, Zhang T, Zhou F, Wang YE, Quanquin N, Wang G, Tian X, He Z, Liu L, Yu W, Sanchez DJ, Liang Y, Jiang T, Modlin R, Bloom BR, Li Q, Deng JC, Zhou P, Qin FXF, Cheng G. Generation of a Live Attenuated Influenza Vaccine that Elicits Broad Protection in Mice and Ferrets. Cell Host Microbe 2017; 21:334-343. [PMID: 28279345 DOI: 10.1016/j.chom.2017.02.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/05/2017] [Accepted: 02/06/2017] [Indexed: 11/18/2022]
Abstract
New influenza vaccines that provide effective and broad protection are desperately needed. Live attenuated viruses are attractive vaccine candidates because they can elicit both humoral and cellular immune responses. However, recent formulations of live attenuated influenza vaccines (LAIVs) have not been protective. We combined high-coverage transposon mutagenesis of influenza virus with a rapid high-throughput screening for attenuation to generate W7-791, a live attenuated mutant virus strain. W7-791 produced only a transient asymptomatic infection in adult and neonatal mice even at doses 100-fold higher than the LD50 of the parent strain. A single administration of W7-791 conferred full protection to mice against lethal challenge with H1N1, H3N2, and H5N1 strains, and improved viral clearance in ferrets. Adoptive transfer of T cells from W7-791-immunized mice conferred heterologous protection, indicating a role for T cell-mediated immunity. These studies present an LAIV development strategy to rapidly generate and screen entire libraries of viral clones.
Collapse
Affiliation(s)
- Lulan Wang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Su-Yang Liu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hsiang-Wen Chen
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Juan Xu
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Maxime Chapon
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tao Zhang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Fan Zhou
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200025, China
| | - Yao E Wang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Natalie Quanquin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Guiqin Wang
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200025, China
| | - Xiaoli Tian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming 650106, China
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming 650106, China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming 650106, China
| | - David Jesse Sanchez
- Department of Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Yuying Liang
- 295K Animal Science/Veterinary Medicine, University of Minnesota, 1988 Fitch Avenue, St. Paul, MN 55108, USA
| | - Taijiao Jiang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Robert Modlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Barry R Bloom
- Harvard School of Public Health, Boston, MA 02115, USA
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming 650106, China
| | - Jane C Deng
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paul Zhou
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200025, China
| | - F Xiao-Feng Qin
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China.
| | - Genhong Cheng
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
42
|
Wang C, Zhu W, Wang BZ. Dual-linker gold nanoparticles as adjuvanting carriers for multivalent display of recombinant influenza hemagglutinin trimers and flagellin improve the immunological responses in vivo and in vitro. Int J Nanomedicine 2017; 12:4747-4762. [PMID: 28740382 PMCID: PMC5503497 DOI: 10.2147/ijn.s137222] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Vaccination is the most cost-effective means of infectious disease control. Although current influenza vaccines are effective in battling closely matched strains, such vaccines have major limitations such as the requirement to produce new vaccines every season, an egg-dependent production system, long production periods, uncertainty in matching the vaccine to circulating strains, and the inability to react to new influenza pandemics resulting from genetic drift or shift. To overcome the intrinsic limitations of the conventional influenza vaccine, we have designed dual-linker gold nanoparticles (AuNPs) conjugated with both recombinant trimetric A/Aichi/2/68 (H3N2), hemagglutinin (HA) and TLR5 agonist flagellin (FliC) as a novel vaccine approach. Click chemistry and metal-chelating reactions were used to couple the two proteins. The conjugated proteins were found to possess high coupling specificity, high stability in harsh environments, high conjugation efficiency, and the ability to keep the appropriate protein conformations for immunogenicity and immunostimulation. Both AuNPs-HA/FliC and AuNPs-HA formulations induced higher levels of antibody responses than a mixture of soluble HA and FliC proteins when administered via a single intranasal immunization in mice. To further investigate the adjuvancy of these nanoparticles, in vitro experiments were conducted in both the JAWS II dendritic cell (DC) line and bone marrow-derived DC (BMDC) models. The results showed that dual-conjugated AuNPs were rapidly targeted and taken up by DCs. Consequently, DCs were induced toward maturation, as demonstrated by high levels of cytokine secretions and membrane costimulatory molecule expression. T cell proliferation was observed when splenic T cells were cocultured with AuNPs-HA/FliC-primed BMDCs. These results suggest that dual-conjugated AuNPs are effective at simultaneously displaying antigens and adjuvants in an oriented, multivalent format and can promote a strong immune response by activating DCs and T cells.
Collapse
Affiliation(s)
- Chao Wang
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| |
Collapse
|
43
|
Deng L, Kim JR, Chang TZ, Zhang H, Mohan T, Champion JA, Wang BZ. Protein nanoparticle vaccine based on flagellin carrier fused to influenza conserved epitopes confers full protection against influenza A virus challenge. Virology 2017. [PMID: 28622575 DOI: 10.1016/j.virol.2017.06.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Currently marketed influenza vaccines only confer protection against matching influenza virus strains. The influenza A composition of these vaccines needs to be annually updated. Vaccines that target conserved epitopes of influenza viruses would in principle offer broad cross-protection against influenza A viruses. In our study, we investigated the specific immune responses and protective efficacy of protein nanoparticles based on fusion proteins of flagellin carrier linked to conserved influenza epitopes. We designed fusion proteins by replacing the hyperimmunogenic region of flagellin (FliC) with four tandem copies of the ectodomain of matrix protein 2 (f4M2e), H1 HA2 domain (fHApr8) or H3 HA2 domain (fHAaichi). Protein nanoparticles fabricated from these fusion proteins by using DTSSP crosslinking retained Toll-like receptor 5 agonist activity of FliC. Intranasal immunization with f4M2e, f4M2e/fHApr8 or f4M2e/fHAaichi nanoparticles induced vaccine antigen-specific humoral immune responses. It was also found that the incorporation of the H1 HA2 domain into f4M2e/fHApr8 nanoparticles boosted M2e specific antibody responses. Immunized mice were fully protected against lethal doses of virus challenge.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antibodies, Viral/blood
- Disease Models, Animal
- Drug Carriers/metabolism
- Epitopes/genetics
- Epitopes/immunology
- Flagellin/genetics
- Flagellin/metabolism
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Nanoparticles
- Orthomyxoviridae Infections/prevention & control
- Protein Binding
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Survival Analysis
- Toll-Like Receptor 5/metabolism
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/metabolism
Collapse
Affiliation(s)
- Lei Deng
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Jong R Kim
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Timothy Z Chang
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, Atlanta, GA, USA
| | - Han Zhang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Teena Mohan
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Julie A Champion
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| |
Collapse
|
44
|
Chakraborty I, Pradeep T. Atomically Precise Clusters of Noble Metals: Emerging Link between Atoms and Nanoparticles. Chem Rev 2017; 117:8208-8271. [DOI: 10.1021/acs.chemrev.6b00769] [Citation(s) in RCA: 1305] [Impact Index Per Article: 163.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Indranath Chakraborty
- DST Unit of Nanoscience (DST
UNS) and Thematic Unit of Excellence, Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | - Thalappil Pradeep
- DST Unit of Nanoscience (DST
UNS) and Thematic Unit of Excellence, Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
45
|
Chang TZ, Diambou I, Kim JR, Wang B, Champion JA. Host- and pathogen-derived adjuvant coatings on protein nanoparticle vaccines. Bioeng Transl Med 2017; 2:120-130. [PMID: 28516165 PMCID: PMC5412930 DOI: 10.1002/btm2.10052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/02/2016] [Accepted: 12/03/2016] [Indexed: 02/01/2023] Open
Abstract
Nanoparticulate and molecular adjuvants have shown great efficacy in enhancing immune responses, and the immunogenic vaccines of the future will most likely contain both. To investigate the immunostimulatory effects of molecular adjuvants on nanoparticle vaccines, we have designed ovalbumin (OVA) protein nanoparticles coated with two different adjuvants-flagellin (FliC) and immunoglobulin M (IgM). These proteins, derived from Salmonella and mice, respectively, are representatives of pathogen- and host-derived molecules that can enhance immune responses. FliC-coated OVA nanoparticles, soluble FliC (sFliC) admixed with OVA nanoparticles, IgM-coated nanoparticles, and OVA-coated nanoparticles were assessed for immunogenicity in an in vivo mouse immunization study. IgM coatings on nanoparticles significantly enhanced both antibody and T cell responses, and promoted IgG2a class switching but not affinity maturation. FliC-coated nanoparticles and FliC-admixed with nanoparticles both triggered IgG2a class switching, but only FliC-coated nanoparticles enhanced antibody affinity maturation. Our findings that affinity maturation and class switching can be directed independently of one another suggest that adjuvant coatings on nanoparticles can be tailored to generate specific vaccine effector responses against different classes of pathogens.
Collapse
Affiliation(s)
- Timothy Z. Chang
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA 30332
| | - Ishatou Diambou
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA 30332
| | - Jong Rok Kim
- Institute for Biomedical SciencesGeorgia State UniversityAtlantaGA 30332
| | - Baozhong Wang
- Institute for Biomedical SciencesGeorgia State UniversityAtlantaGA 30332
| | - Julie A. Champion
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA 30332
| |
Collapse
|
46
|
Chang TZ, Stadmiller SS, Staskevicius E, Champion JA. Effects of ovalbumin protein nanoparticle vaccine size and coating on dendritic cell processing. Biomater Sci 2017; 5:223-233. [PMID: 27918020 PMCID: PMC5285395 DOI: 10.1039/c6bm00500d] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nanoparticle vaccine delivery platforms are a promising technology for enhancing vaccine immunogenicity. Protein nanoparticles (PNPs), made entirely from antigen, have been shown to induce protective immune responses against influenza. However, the fundamental mechanisms by which PNPs enhance component protein immunogenicity are not understood. Here, we investigate the role of size and coating of model ovalbumin (OVA) PNPs on particle uptake and trafficking, as well as on inflammation and maturation factor expression in dendritic cells (DCs) in vitro. OVA PNPs enhance antigen uptake in a size-independent manner, and experience attenuated endosomal acidification as compared to soluble OVA. OVA PNPs also trigger Fc receptor upregulation. Expression of cytokines IL-1β and TNF-α were PNP size- and coating-dependent, with small (∼270 nm) nanoparticles triggering greater inflammatory cytokine production than large (∼560 nm) particles. IL-1β expression by DCs in response to PNP stimulation implies activation of the inflammasome, a pathway known to be activated by certain types of nanoparticulate adjuvants. The attenuated acidification and pro-inflammatory profile generated by PNPs in DCs demonstrate that physical biomaterial properties can modulate dendritic cell-mediated antigen processing and adjuvancy. In addition to nanoparticles' enhancement of DC antigen uptake, our work suggests that vaccine nanoparticle size and coating are uptake-independent modulators of immunogenicity.
Collapse
Affiliation(s)
- Timothy Z Chang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Samantha S Stadmiller
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Erika Staskevicius
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| |
Collapse
|
47
|
Wang L, Chang TZ, He Y, Kim JR, Wang S, Mohan T, Berman Z, Tompkins SM, Tripp RA, Compans RW, Champion JA, Wang BZ. Coated protein nanoclusters from influenza H7N9 HA are highly immunogenic and induce robust protective immunity. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2017; 13:253-262. [PMID: 27622321 PMCID: PMC5237404 DOI: 10.1016/j.nano.2016.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 08/05/2016] [Accepted: 09/01/2016] [Indexed: 01/08/2023]
Abstract
Recurring influenza viruses pose an annual threat to public health. A time-saving, cost-effective and egg-independent influenza vaccine approach is important particularly when responding to an emerging pandemic. We fabricated coated, two-layer protein nanoclusters from recombinant trimeric hemagglutinin from an avian-origin H7N9 influenza A virus as an approach for vaccine development in response to an emerging pandemic. Assessment of the virus-specific immune responses and protective efficacy in mice immunized with the nanoclusters demonstrated that the vaccine candidates were highly immunogenic, able to induce protective immunity and long-lasting humoral antibody responses to this virus without the use of adjuvants. Because the advantages of the highly immunogenic coated nanoclusters also include rapid productions in an egg-independent system, this approach has great potential for influenza vaccine production not only in response to an emerging pandemic, but also as a replacement for conventional seasonal influenza vaccines.
Collapse
Affiliation(s)
- Li Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Timothy Z Chang
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, Atlanta, GA, USA.
| | - Yuan He
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Jong R Kim
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Shelly Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Teena Mohan
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Zachary Berman
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Julie A Champion
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, Atlanta, GA, USA.
| | - Bao-Zhong Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| |
Collapse
|
48
|
Zhang J, Fan HY, Zhang Z, Zhang J, Zhang J, Huang JN, Ye Y, Liao M. Recombinant baculovirus vaccine containing multiple M2e and adjuvant LTB induces T cell dependent, cross-clade protection against H5N1 influenza virus in mice. Vaccine 2016; 34:622-629. [DOI: 10.1016/j.vaccine.2015.12.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 10/19/2015] [Accepted: 12/15/2015] [Indexed: 12/01/2022]
|
49
|
Mbewana S, Mortimer E, Pêra FFPG, Hitzeroth II, Rybicki EP. Production of H5N1 Influenza Virus Matrix Protein 2 Ectodomain Protein Bodies in Tobacco Plants and in Insect Cells as a Candidate Universal Influenza Vaccine. Front Bioeng Biotechnol 2015; 3:197. [PMID: 26697423 PMCID: PMC4672040 DOI: 10.3389/fbioe.2015.00197] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/23/2015] [Indexed: 11/13/2022] Open
Abstract
The spread of influenza A viruses is partially controlled and prevented by vaccination. The matrix protein 2 ectodomain (M2e) is the most conserved sequence in influenza A viruses, and is therefore a good potential target for a vaccine to protect against multiple virus subtypes. We explored the feasibility of an M2e-based universal influenza A vaccine candidate based on the highly pathogenic avian influenza A virus, H5N1. A synthetic M2e gene was human- and plant-codon optimized and fused in-frame with a sequence encoding the N-terminal proline-rich domain (Zera(®)) of the γ-zein protein of maize. Zera(®)M2e was expressed transiently in Nicotiana benthamiana and Sf21 baculovirus/insect cell expression systems, and Zera(®)M2e protein bodies (PBs) were successfully produced in both expression systems. The plant-produced Zera(®)M2e PBs were purified and injected into Balb/c mice. Western blot analysis using insect cell-produced Zera(®)M2e PBs and multiple tandem M2e sequences (5xM2e) fused with the avian influenza H5N1 transmembrane and cytosolic tail (5xM2e_tHA) confirmed the presence of M2e-specific antibodies in immunized mice sera. The immunogenicity of the Zera(®)M2e indicates that our plant-produced protein has potential as an inexpensive universal influenza A vaccine.
Collapse
Affiliation(s)
- Sandiswa Mbewana
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town , Rondebosch , South Africa
| | - Elizabeth Mortimer
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town , Rondebosch , South Africa
| | - Francisco F P G Pêra
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town , Rondebosch , South Africa
| | - Inga Isabel Hitzeroth
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town , Rondebosch , South Africa
| | - Edward P Rybicki
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town , Rondebosch , South Africa ; Institute of Infectious Disease and Molecular Medicine, Faculty of Heath Science, University of Cape Town , Cape Town , South Africa
| |
Collapse
|
50
|
Abstract
The development of vaccines that could provide broad protection against antigenically variant influenza viruses has long been the ultimate prize in influenza research. Recent developments have pushed us closer to this goal, and such vaccines may now be within reach. This brief review outlines the current approaches to broadly protective vaccines, and the probable hurdles and roadblocks to achieving this goal.
Collapse
Affiliation(s)
- John Jay Treanor
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Box 689, 601 Elmwood Avenue, Rochester, NY 14642, United States.
| |
Collapse
|