1
|
Sudaraka Tennakoon MSBWTMN, Park JY, Lee HM, Ryu JH, Shin HJ. Ferritin nanoparticle complex with porcine epidemic diarrhea virus spike protein induces neutralizing antibody response against PEDV in mouse models. Microb Pathog 2025; 203:107509. [PMID: 40147557 DOI: 10.1016/j.micpath.2025.107509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
As the spike protein is the major antigen that contains various neutralizing epitopes against porcine epidemic diarrhea virus (PEDV), numerous vaccine trials employing the spike protein have been established. In this study, we developed a ferritin-based nanoparticle vaccine for PEDV by combining gene delivery functions of recombinant adenoviruses. To generate nanoparticles, the S1 subunit of the spike protein was genetically linked to the N-terminus of the human ferritin heavy chain (hFTHC), and recombinant adenoviruses were generated to deliver the genetic material. The efficacy of S1 conjugated human ferritin heavy chain (S1-hFTHC) adenoviruses against S1 adenoviruses was evaluated in BALB/c mice immunized intramuscularly without adjuvant. Two weeks after the final boost, we observed a significantly higher IgG response in S1-hFTHC immunized mice compared with the S1 immunized mice, and results from the virus neutralization assay revealed robust virus neutralization activity in the S1-hFTHC immunized group compared to the S1 immunized group. Furthermore, analysis of the serum based on IgG and neutralizing titers 40 days after the last vaccination revealed the significance and longevity of the immune response induced by S1-hFTCH compared to S1 only. This strategy elucidates the efficacy of combined vaccine strategies for developing promising vaccine candidates against PEDV.
Collapse
Affiliation(s)
| | - Jae-Yeon Park
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Hye-Mi Lee
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | | | - Hyun-Jin Shin
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
2
|
Wu J, Liang J, Li S, Lu J, Li Y, Zhang B, Gao M, Zhou J, Zhang Y, Chen J. Cancer vaccine designed from homologous ferritin-based fusion protein with enhanced DC-T cell crosstalk for durable adaptive immunity against tumors. Bioact Mater 2025; 46:516-530. [PMID: 39868073 PMCID: PMC11764028 DOI: 10.1016/j.bioactmat.2024.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/30/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Peptide vaccines based on tumor antigens face the challenges of rapid clearance of peptides, low immunogenicity, and immune suppressive tumor microenvironment. However, the traditional solution mainly uses exogenous substances as adjuvants or carriers to enhance innate immune responses, but excessive inflammation can damage adaptive immunity. In the current study, we propose a straightforward novel nanovaccine strategy by employing homologous human ferritin light chain for minimized innate immunity and dendritic cell (DC) targeting, the cationic KALA peptide for enhanced cellular uptake, and suppressor of cytokine signaling 1 (SOCS1) siRNA for modulating DC activity. Upon fusing with the KALA peptide, this nanovaccine presents as a novel 40-mer cage structure, with highly enriched antigen peptides of proper size (25 nm) for targeted delivery to lymph nodes. The loading of SOCS1 siRNA onto the KALA peptide promoted DC maturation in tumor environment, leading to a 3-fold increase in antigen presentation compared to alum adjuvant. Moreover, it demonstrates remarkable efficacy in suppressing tumor progression and metastasis, together with prolonged survival. In addition, the nanovaccine stimulates up to 40 % memory T cells, thereby achieving sustained protection against tumor re-challenge. This unprecedented nanovaccine platform can ignite fresh interdisciplinary discussions on interactive strategies for future peptide vaccine development.
Collapse
Affiliation(s)
- Jun Wu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
- School of Chemical & Material Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Jing Liang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Sichen Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Jinjin Lu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Yi Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Bin Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Min Gao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Juan Zhou
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Yan Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| |
Collapse
|
3
|
Wang Q, Nie J, Liu Z, Chang Y, Wei Y, Yao X, Sun L, Liu X, Liu Q, Liang X, Zhang X, Zhang Y, Su W, Zhao Q, Shan Y, Wang Y, Cheng X, Shi Y. Induction of enhanced stem-directed neutralizing antibodies by HA2-16 ferritin nanoparticles with H3 influenza virus boost. NANOSCALE ADVANCES 2025; 7:2011-2020. [PMID: 39974341 PMCID: PMC11833233 DOI: 10.1039/d4na00964a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
Current seasonal influenza vaccines offer limited protection against influenza viruses due to genetic drift. The urgent need for a universal influenza vaccine to combat highly mutated strains is evident. This study utilized the conserved HA2 subunit of hemagglutinin (HA) and a short linear epitope of HA2 (HA2-16) from the H3 influenza virus to conjugate with ferritin, resulting in the construction of recombinant immunogens termed HA2-F and HA2-16-F, respectively. In vitro characterization confirmed the self-assembly of prokaryotically expressed HA2-F and HA2-16-F into nanoparticles (NPs). To simulate natural virus infection in the vaccinated population, intranasal infection with the whole H3N2 virus was administered as a final boost. Enhanced binding activity to A/Hong Kong/4801/2014 (H3N2) and A/17/California/2009/38 (H1N1) virus was detected in the HA2-16 group induced by the A/Wisconsin/67/2005 (H3N2) virus boost (Titer >104). Furthermore, higher titers of neutralizing antibodies were elicited by HA2-16-F NP (ID50: 50.4-631.0) compared to those by HA2-F NP (ID50: 20.3-178.2). These results demonstrated that the H3N2 virus boost focused the antibody response on the HA2-16 epitope. Additionally, our immunization strategy was found to reduce serum ferritin reactive antibodies. In summary, HA2-16 not only holds promise as a vaccine candidate but also exhibits significant potential for influenza vaccine production, particularly in enhancing the levels of induced stem-directed antibodies. This study contributes to the development of recombinant immunogens for improved influenza vaccine efficacy.
Collapse
Affiliation(s)
- Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Zejinxuan Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Yaotian Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Yangang Wei
- MTM Biotechnology Ltd Zhongshan Guangdong 528437 China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Lulu Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Xiaoxi Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Qicheng Liu
- High School Attached to Northeast Normal University Changchun Jilin 130012 China
| | - Xinyu Liang
- High School Attached to Northeast Normal University Changchun Jilin 130012 China
| | - Xinran Zhang
- High School Attached to Northeast Normal University Changchun Jilin 130012 China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Weiheng Su
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Qi Zhao
- Faculty of Health Sciences, University of Macau Taipa Macau China
- MoE Frontiers Science Center for Precision Oncology, University of Macau Taipa Macau SAR China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Yingwu Wang
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| | - Xianbin Cheng
- Department of Thyroid Surgery, The Second Hospital of Jilin University Changchun China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University Changchun Jilin 130012 China
| |
Collapse
|
4
|
Ou L, Setegne MT, Elliot J, Shen F, Dassama LMK. Protein-Based Degraders: From Chemical Biology Tools to Neo-Therapeutics. Chem Rev 2025; 125:2120-2183. [PMID: 39818743 PMCID: PMC11870016 DOI: 10.1021/acs.chemrev.4c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
The nascent field of targeted protein degradation (TPD) could revolutionize biomedicine due to the ability of degrader molecules to selectively modulate disease-relevant proteins. A key limitation to the broad application of TPD is its dependence on small-molecule ligands to target proteins of interest. This leaves unstructured proteins or those lacking defined cavities for small-molecule binding out of the scope of many TPD technologies. The use of proteins, peptides, and nucleic acids (otherwise known as "biologics") as the protein-targeting moieties in degraders addresses this limitation. In the following sections, we provide a comprehensive and critical review of studies that have used proteins and peptides to mediate the degradation and hence the functional control of otherwise challenging disease-relevant protein targets. We describe existing platforms for protein/peptide-based ligand identification and the drug delivery systems that might be exploited for the delivery of biologic-based degraders. Throughout the Review, we underscore the successes, challenges, and opportunities of using protein-based degraders as chemical biology tools to spur discoveries, elucidate mechanisms, and act as a new therapeutic modality.
Collapse
Affiliation(s)
- Lisha Ou
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Mekedlawit T. Setegne
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Jeandele Elliot
- Department
of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Fangfang Shen
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Laura M. K. Dassama
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology & Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
5
|
Wang E, Cohen AA, Caldera LF, Keeffe JR, Rorick AV, Adia YM, Gnanapragasam PNP, Bjorkman PJ, Chakraborty AK. Designed mosaic nanoparticles enhance cross-reactive immune responses in mice. Cell 2025; 188:1036-1050.e11. [PMID: 39855201 PMCID: PMC11845252 DOI: 10.1016/j.cell.2024.12.015] [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: 02/27/2024] [Revised: 09/27/2024] [Accepted: 12/11/2024] [Indexed: 01/27/2025]
Abstract
Nanoparticle vaccines displaying combinations of SARS-like betacoronavirus (sarbecovirus) receptor-binding domains (RBDs) could protect against SARS-CoV-2 variants and spillover of zoonotic sarbecoviruses into humans. Using a computational approach, we designed variants of SARS-CoV-2 RBDs and selected 7 natural sarbecovirus RBDs, each predicted to fold properly and abrogate antibody responses to variable epitopes. RBDs were attached to 60-mer nanoparticles to make immunogens displaying two (mosaic-2COMs), five (mosaic-5COM), or seven (mosaic-7COM) different RBDs for comparisons with mosaic-8b, which elicited cross-reactive antibodies and protected animals from sarbecovirus challenges. Naive and COVID-19 pre-vaccinated mice immunized with mosaic-7COM elicited antibodies targeting conserved RBD epitopes, and their sera exhibited higher binding and neutralization titers against sarbecoviruses than mosaic-8b. Mosaic-2COMs and mosaic-5COM elicited higher antibody potencies against some SARS-CoV-2 variants than mosaic-7COM. However, mosaic-7COM elicited more potent responses against zoonotic sarbecoviruses and highly mutated Omicrons, supporting its use to protect against SARS-CoV-2 variants and zoonotic sarbecoviruses.
Collapse
Affiliation(s)
- Eric Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander A Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Luis F Caldera
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Annie V Rorick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yusuf M Adia
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Arup K Chakraborty
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA.
| |
Collapse
|
6
|
Jiang Y, Sanyal M, Hussein NA, Baghdasaryan A, Zhang M, Wang F, Ren F, Li J, Zhu G, Meng Y, Adamska JZ, Mellins E, Dai H. A SARS-CoV-2 vaccine on an NIR-II/SWIR emitting nanoparticle platform. SCIENCE ADVANCES 2025; 11:eadp5539. [PMID: 39919189 PMCID: PMC11804919 DOI: 10.1126/sciadv.adp5539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025]
Abstract
The COVID-19 pandemic caused a global health crisis that resulted in millions of deaths. Effective vaccines have played central roles in curtailing the pandemic. Here, we developed a down-converting near-infrared IIb (NIR-IIb; 1500 to 1700 nanometers) luminescent, pure NaErF4@NaYF4 rare-earth nanoparticle (pEr) as vaccine carriers. The pEr nanoparticles were coated with three layers of cross-linked biocompatible polymers (pEr-P3; ~55 nanometers) and conjugated to the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Upon subcutaneous injection of the pEr-P3-RBD nanovaccine in mice, in vivo NIR-IIb imaging revealed active vaccine trafficking and migration to lymph nodes through lymphatic vessels. Two doses of the adjuvant-free vaccine elicited long-lasting (>7 months) high titers of serum viral neutralization antibody and anti-RBD immunoglobulin G, along with robust RBD-specific germinal center B cells and T follicular helper cells. We devised in vivo NIR-II molecular imaging of RBD-specific cells in lymph nodes, opening noninvasive assessments of vaccine-elicited immune responses longitudinally.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Noor A. Hussein
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ani Baghdasaryan
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Mengzhen Zhang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Feifei Wang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Fuqiang Ren
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Jiachen Li
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Guanzhou Zhu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Yifan Meng
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Julia Zofia Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Hongjie Dai
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| |
Collapse
|
7
|
Jung J, Kim TH, Park JY, Kwon S, Sung JS, Kang MJ, Jose J, Lee M, Shin HJ, Pyun JC. SARS-CoV-2 vaccine based on ferritin complexes with screened immunogenic sequences from the Fv-antibody library. J Mater Chem B 2025; 13:1383-1394. [PMID: 39668674 DOI: 10.1039/d4tb01595a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
In this study, the vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was developed using ferritin complexes with the immunogenic sequences screened against the SARS-CoV-2 spike protein (SP) from the Fv-antibody library. The Fv-antibody library was prepared on the outer membrane of E. coli by the expression of the VH region of immunoglobulin G (IgG) with a randomized complementarity-determining region 3 (CDR3). Four Fv-antibodies to the receptor-binding domain (RBD) were screened from the Fv-antibody library, which had a comparable binding constant (KD) between SARS-CoV-2 SP and the angiotensin-converting enzyme 2 (ACE2) receptor. The binding sites of screened Fv-antibodies on the RBD were analyzed using a docking analysis, and these binding sites were used as immunogenic sequences for the vaccine. The four immunogenic sequences were modified and co-expressed as a part of ferritin which was assembled into a ferritin complex. After the vaccination of ferritin complexes to mice, the anti-sera were analyzed to have a high enough titer. Additionally, the immune responses were found to be activated by vaccination, such as the expression of IgG subclasses and the increased level of cytokines. The neutralizing activity of the anti-sera was estimated using a cell-based infection assay based on pseudo-virus expressing the SP of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Tae-Hun Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Jae-Yeon Park
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02456, Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, University of Munster, Münster (48149), Germany
| | - Misu Lee
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Hyun-Jin Shin
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| |
Collapse
|
8
|
Gao X, Zhu X, Liu X, Zhou C, Shang Y, Wu T, Jia H, Zhang Z, Li Y, Xin T. A Ferritin-Based Eg95 Nanoparticle Vaccine Adjuvanted with pCpG Eliciting Robust Immune Responses Against Cystic Echinococcosis in Mice Model. Int J Nanomedicine 2025; 20:309-325. [PMID: 39802377 PMCID: PMC11725278 DOI: 10.2147/ijn.s499938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Cystic echinococcosis (CE), a chronic disabling parasitic zoonosis, poses a great threat to public health and livestock production and causes huge economic losses globally. The commercial Quil-A-adjuvanted Eg95 vaccine was empirically effective for CE control; however, it is expensive and has side effects and insufficient immunity. Purpose This study aimed to employ a novel adjuvant consisting of a delivery system and an immune potentiator and assess its adjuvanticity to Eg95 antigen, thereby developing a safe and cost-effective novel vaccine against the disease. Methods A ferritin-based Eg95 nanoparticle antigen was prepared and then mixed with a plasmid containing the TLR9 agonist CpG to formulate a novel nanovaccine. The safety and efficacy of the vaccine were evaluated in vitro and in vivo. Results The nanovaccine induced potent and enduring Eg95-specific humoral and cellular immune responses, as well as protective immunity-associated Th1 polarization supported by the higher ratios of IgG2a/IgG1 and IFN-γ/IL-4. Meanwhile, this nanovaccines exhibited favorable safety and economic profiles. Conclusion Our data demonstrated that the ferritin-CpG hybrid is a promising combination adjuvant to upgrade the traditional Quil-A and this combination adjuvant-based nanovaccine presents good potential as an alternative to the commercial one for practical CE control.
Collapse
MESH Headings
- Animals
- Ferritins/chemistry
- Nanoparticles/chemistry
- Echinococcosis/prevention & control
- Echinococcosis/immunology
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/administration & dosage
- Mice
- Antigens, Helminth/immunology
- Antigens, Helminth/chemistry
- Disease Models, Animal
- Female
- Mice, Inbred BALB C
- Oligodeoxyribonucleotides/chemistry
- Adjuvants, Vaccine/chemistry
- Adjuvants, Vaccine/administration & dosage
- Immunity, Cellular/drug effects
- Vaccines/immunology
- Vaccines/chemistry
- Immunity, Humoral
- Nanovaccines
Collapse
Affiliation(s)
- Xintao Gao
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xizhou Zhu
- Bioproducts Engineering Center, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xingjian Liu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Chenghao Zhou
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yuting Shang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Tong Wu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Hong Jia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Zhifang Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yinü Li
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Ting Xin
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| |
Collapse
|
9
|
de Almeida CMF, Rios WM, Duarte MPF, Brandão IT, Paiva NF, Vicentini FTMDC. Self-assembled nanovaccine based on apoferritin: Development and vaccine regimen evaluation. Eur J Pharm Biopharm 2025; 206:114589. [PMID: 39613269 DOI: 10.1016/j.ejpb.2024.114589] [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: 07/16/2024] [Revised: 09/18/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Apoferritin-based systems have been explored last decade for their potential as vaccine delivery for viral diseases. The nanosized properties of an apoferritin-based system could increase immunogenicity, contribute to antigen stability, and reduce the vaccines' adverse effects. The mutated extracellular portion of the epidermal growth factor receptor (EGFRvIII peptide, PEPvIII) can be applied as a specific tumoral antigen due to rare expression in normal cells. In this context, the present study proposed the development and the immunogenicity evaluation of an apoferritin-based system (AFt) to deliver a peptide vaccine for an antitumoral purpose. We developed a formulation with different PEPvIII:AFt ratios and during the association efficiency analysis, identified the dependence between the AFt concentration and the PEPvIII association percentage in the formulation. Besides, differences in the immune responses against EGFRvIII were observed depending on the PEPvIII concentration due to the different association efficiencies. Finally, the humoral immune response results showed a high antibody production against AFt, which might affect the immunological tolerance. Collectively, this study establishes the PEPvIII:AFt formulation process and highlights the determinant factors for guaranteeing vaccine safety and efficacy.
Collapse
Affiliation(s)
| | - Wendy Martin Rios
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | | | - Izaíra Tincani Brandão
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | - Natalia Floriano Paiva
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | | |
Collapse
|
10
|
Wang X, Qin Z, Zhang M, Shang B, Li Z, Zhao M, Tang Q, Tang Q, Luo J. Immunogenicity and protection of recombinant self-assembling ferritin-hemagglutinin nanoparticle influenza vaccine in mice. Clin Exp Vaccine Res 2025; 14:23-34. [PMID: 39927225 PMCID: PMC11799580 DOI: 10.7774/cevr.2025.14.e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/21/2024] [Accepted: 12/21/2024] [Indexed: 02/11/2025] Open
Abstract
Purpose Influenza virus remains a serious burden to global public health. Current influenza vaccine fails to provide impeccable protection efficacy to the annual seasonal influenza and cannot offer a timely response to potential pandemic influenza. It is necessary to develop next generation influenza vaccines to solve the current dilemma. Materials and Methods We developed a recombinant, self-assembling ferritin nanoparticle that presents the extracellular domain of the influenza hemagglutinin antigen on its surface, designated as ferritin-HA. After characterizing its structure and properties, we evaluated its capacity to trigger an immune response and offer protection against influenza virus challenge in a mouse model. Results The recombinant ferritin-HA protein expressed in Chinese hamster ovary cells assembles into nanoparticles of a defined size. This nanoparticle vaccine enhances the uptake efficiency of Dendritic cells and promotes their maturation. Immunization with ferritin-HA nanoparticle in mice induced high levels of immunoglobulin G, hemagglutination inhibition antibodies, and microneutralization antibodies, demonstrating their stronger immunogenicity compared to current split virion vaccines. Additionally, ferritin-HA nanoparticle conferred well protection against a lethal challenge with a heterologous H3N2 influenza virus in mice. Conclusion This study indicates that a self-assembling ferritin-HA nanoparticle has great potential for enhancing immune response and protective efficacy in mice, presenting a promising strategy for developing next generation influenza vaccine candidate.
Collapse
Affiliation(s)
- Xu Wang
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Ziyao Qin
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Min Zhang
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Baoyuan Shang
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Zhilei Li
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Meiyi Zhao
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Qing Tang
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Qi Tang
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
| | - Jian Luo
- Department of Virology & Vaccine, Shanghai Institute of Biological Products, Shanghai, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, Beijing, China
| |
Collapse
|
11
|
Catala A, Davenport BJ, Morrison TE, Catalano CE. Second-Generation Phage Lambda Platform Employing SARS-CoV-2 Fusion Proteins as a Vaccine Candidate. Vaccines (Basel) 2024; 12:1201. [PMID: 39591104 PMCID: PMC11598875 DOI: 10.3390/vaccines12111201] [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: 09/18/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
The recent SARS-CoV-2 (COVID-19) pandemic exemplifies how newly emerging and reemerging viruses can quickly overwhelm and cripple global infrastructures. Coupled with synergistic factors such as increasing population densities, the constant and massive mobility of people across geographical areas and substantial changes to ecosystems worldwide, these pathogens pose serious health concerns on a global scale. Vaccines form an indispensable defense, serving to control and mitigate the impact of devastating outbreaks and pandemics. Towards these efforts, we developed a tunable vaccine platform that can be engineered to simultaneously display multiple viral antigens. Here, we describe a second-generation version wherein chimeric proteins derived from SARS-CoV-2 and bacteriophage lambda are engineered and used to decorate phage-like particles with defined surface densities and retention of antigenicity. This streamlines the engineering of particle decoration, thus improving the overall manufacturing potential of the system. In a prime-boost regimen, mice immunized with particles containing as little as 42 copies of the chimeric protein on their surface develop potent neutralizing antibody responses, and immunization protects mice against virulent SARS-CoV-2 challenge. The platform is highly versatile, making it a promising strategy to rapidly develop vaccines against a potentially broad range of infectious diseases.
Collapse
Affiliation(s)
- Alexis Catala
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Bennett J. Davenport
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (B.J.D.); (T.E.M.)
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (B.J.D.); (T.E.M.)
| | - Carlos E. Catalano
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
12
|
Cohen AA, Keeffe JR, Schiepers A, Dross SE, Greaney AJ, Rorick AV, Gao H, Gnanapragasam PNP, Fan C, West AP, Ramsingh AI, Erasmus JH, Pata JD, Muramatsu H, Pardi N, Lin PJC, Baxter S, Cruz R, Quintanar-Audelo M, Robb E, Serrano-Amatriain C, Magneschi L, Fotheringham IG, Fuller DH, Victora GD, Bjorkman PJ. Mosaic sarbecovirus nanoparticles elicit cross-reactive responses in pre-vaccinated animals. Cell 2024; 187:5554-5571.e19. [PMID: 39197450 PMCID: PMC11460329 DOI: 10.1016/j.cell.2024.07.052] [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: 01/17/2024] [Revised: 06/15/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024]
Abstract
Immunization with mosaic-8b (nanoparticles presenting 8 SARS-like betacoronavirus [sarbecovirus] receptor-binding domains [RBDs]) elicits more broadly cross-reactive antibodies than homotypic SARS-CoV-2 RBD-only nanoparticles and protects against sarbecoviruses. To investigate original antigenic sin (OAS) effects on mosaic-8b efficacy, we evaluated the effects of prior COVID-19 vaccinations in non-human primates and mice on anti-sarbecovirus responses elicited by mosaic-8b, admix-8b (8 homotypics), or homotypic SARS-CoV-2 immunizations, finding the greatest cross-reactivity for mosaic-8b. As demonstrated by molecular fate mapping, in which antibodies from specific cohorts of B cells are differentially detected, B cells primed by WA1 spike mRNA-LNP dominated antibody responses after RBD-nanoparticle boosting. While mosaic-8b- and homotypic-nanoparticles boosted cross-reactive antibodies, de novo antibodies were predominantly induced by mosaic-8b, and these were specific for variant RBDs with increased identity to RBDs on mosaic-8b. These results inform OAS mechanisms and support using mosaic-8b to protect COVID-19-vaccinated/infected humans against as-yet-unknown SARS-CoV-2 variants and animal sarbecoviruses with human spillover potential.
Collapse
Affiliation(s)
- Alexander A Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Sandra E Dross
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; National Primate Research Center, Seattle, WA 98121, USA
| | - Allison J Greaney
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Annie V Rorick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Han Gao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Chengcheng Fan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Anthony P West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | - Janice D Pata
- Wadsworth Center, New York State Department of Health and Department of Biomedical Sciences, University at Albany, Albany, NY 12201, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Scott Baxter
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | - Rita Cruz
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | | | - Ellis Robb
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | | | - Leonardo Magneschi
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | - Ian G Fotheringham
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | - Deborah H Fuller
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; National Primate Research Center, Seattle, WA 98121, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
13
|
Ou BS, Baillet J, Filsinger Interrante MV, Adamska JZ, Zhou X, Saouaf OM, Yan J, Klich JH, Jons CK, Meany EL, Valdez AS, Carter L, Pulendran B, King NP, Appel EA. Saponin nanoparticle adjuvants incorporating Toll-like receptor agonists drive distinct immune signatures and potent vaccine responses. SCIENCE ADVANCES 2024; 10:eadn7187. [PMID: 39110802 PMCID: PMC11305391 DOI: 10.1126/sciadv.adn7187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Over the past few decades, the development of potent and safe immune-activating adjuvant technologies has become the heart of intensive research in the constant fight against highly mutative and immune evasive viruses such as influenza, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and human immunodeficiency virus (HIV). Herein, we developed a highly modular saponin-based nanoparticle platform incorporating Toll-like receptor agonists (TLRas) including TLR1/2a, TLR4a, and TLR7/8a adjuvants and their mixtures. These various TLRa-saponin nanoparticle adjuvant constructs induce unique acute cytokine and immune-signaling profiles, leading to specific T helper responses that could be of interest depending on the target disease for prevention. In a murine vaccine study, the adjuvants greatly improved the potency, durability, breadth, and neutralization of both COVID-19 and HIV vaccine candidates, suggesting the potential broad application of these adjuvant constructs to a range of different antigens. Overall, this work demonstrates a modular TLRa-SNP adjuvant platform that could improve the design of vaccines and affect modern vaccine development.
Collapse
Affiliation(s)
- Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Julie Baillet
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Maria V. Filsinger Interrante
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Julia Z. Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Xueting Zhou
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Olivia M. Saouaf
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jerry Yan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - John H. Klich
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Carolyn K. Jons
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Emily L. Meany
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Adian S. Valdez
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Neil P. King
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Pediatrics-Endocrinology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- Woods Institute for the Environment, Stanford University, Stanford CA 94305, USA
| |
Collapse
|
14
|
Sheng Y, Chen Z, Cherrier MV, Martin L, Bui TTT, Li W, Lynham S, Nicolet Y, Ebrahimi KH. A Versatile Virus-Mimetic Engineering Approach for Concurrent Protein Nanocage Surface-Functionalization and Cargo Encapsulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310913. [PMID: 38726952 DOI: 10.1002/smll.202310913] [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: 11/26/2023] [Revised: 04/26/2024] [Indexed: 08/02/2024]
Abstract
Naturally occurring protein nanocages like ferritin are self-assembled from multiple subunits. Because of their unique cage-like structure and biocompatibility, there is a growing interest in their biomedical use. A multipurpose and straightforward engineering approach does not exist for using nanocages to make drug-delivery systems by encapsulating hydrophilic or hydrophobic drugs and developing vaccines by surface functionalization with a protein like an antigen. Here, a versatile engineering approach is described by mimicking the HIV-1 Gap polyprotein precursor. Various PREcursors of nanoCages (PREC) are designed and created by linking two ferritin subunits via a flexible linker peptide containing a protease cleavage site. These precursors can have additional proteins at their N-terminus, and their protease cleavage generates ferritin-like nanocages named protease-induced nanocages (PINCs). It is demonstrated that PINC formation allows concurrent surface decoration with a protein and hydrophilic or hydrophobic drug encapsulation up to fourfold more than the amount achieved using other methods. The PINCs/Drug complex is stable and efficiently kills cancer cells. This work provides insight into the precursors' design rules and the mechanism of PINCs formation. The engineering approach and mechanistic insight described here will facilitate nanocages' applications in drug delivery or as a platform for making multifunctional therapeutics like mosaic vaccines.
Collapse
Affiliation(s)
- Yujie Sheng
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Zilong Chen
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Mickael V Cherrier
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, Grenoble, CS 10090, France
| | - Lydie Martin
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, Grenoble, CS 10090, France
| | - Tam T T Bui
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, SE11UL, UK
| | - Wei Li
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Steven Lynham
- Proteomics Core Facility, James Black Centre, King's College London, London, SE5 9NU, UK
| | - Yvain Nicolet
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, Grenoble, CS 10090, France
| | - Kourosh H Ebrahimi
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| |
Collapse
|
15
|
Xu D, Carter JJ, Li C, Utz A, Weidenbacher PAB, Tang S, Sanyal M, Pulendran B, Barnes CO, Kim PS. Vaccine design via antigen reorientation. Nat Chem Biol 2024; 20:1012-1021. [PMID: 38225471 PMCID: PMC11247139 DOI: 10.1038/s41589-023-01529-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024]
Abstract
A major challenge in creating universal influenza vaccines is to focus immune responses away from the immunodominant, variable head region of hemagglutinin (HA-head) and toward the evolutionarily conserved stem region (HA-stem). Here we introduce an approach to control antigen orientation via site-specific insertion of aspartate residues that facilitates antigen binding to alum. We demonstrate the generalizability of this approach with antigens from Ebola, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses and observe enhanced neutralizing antibody responses in all cases. We then reorient an H2 HA in an 'upside-down' configuration to increase the exposure and immunogenicity of HA-stem. The reoriented H2 HA (reoH2HA) on alum induced stem-directed antibodies that cross-react with both group 1 and group 2 influenza A subtypes. Electron microscopy polyclonal epitope mapping (EMPEM) revealed that reoH2HA (group 1) elicits cross-reactive antibodies targeting group 2 HA-stems. Our results highlight antigen reorientation as a generalizable approach for designing epitope-focused vaccines.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Joshua J Carter
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton A B Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Shaogeng Tang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher O Barnes
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter S Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
16
|
Furukawa H, Nakamura S, Mizuta R, Sakamoto K, Inaba H, Sawada SI, Sasaki Y, Akiyoshi K, Matsuura K. Enveloped Viral Replica Equipped with Spike Protein Derived from SARS-CoV-2. ACS Synth Biol 2024; 13:2029-2037. [PMID: 38885191 DOI: 10.1021/acssynbio.4c00165] [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] [Indexed: 06/20/2024]
Abstract
Synthetic viral nanostructures are useful as materials for analyzing the biological behavior of natural viruses and as vaccine materials. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enveloped virus embedding a spike (S) protein involved in host cell infection. Although nanomaterials modified with an S protein without an envelope membrane have been developed, they are considered unsuitable for stability and functionality. We previously constructed an enveloped viral replica complexed with a cationic lipid bilayer and an anionic artificial viral capsid self-assembled from β-annulus peptides. In this study, we report the first example of an enveloped viral replica equipped with an S protein derived from SARS-CoV-2. Interestingly, even the S protein equipped on the enveloped viral replica bound strongly to the free angiotensin-converting enzyme 2 (ACE2) receptor as well as ACE2 localized on the cell membrane.
Collapse
Affiliation(s)
- Hiroto Furukawa
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Sosuke Nakamura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Ryosuke Mizuta
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kentarou Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Hiroshi Inaba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- Department of Immunology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
| |
Collapse
|
17
|
Wu X, Li W, Rong H, Pan J, Zhang X, Hu Q, Shi ZL, Zhang XE, Cui Z. A Nanoparticle Vaccine Displaying Conserved Epitopes of the Preexisting Neutralizing Antibody Confers Broad Protection against SARS-CoV-2 Variants. ACS NANO 2024; 18:17749-17763. [PMID: 38935412 DOI: 10.1021/acsnano.4c03075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The rapid development of the SARS-CoV-2 vaccine has been used to prevent the spread of coronavirus 2019 (COVID-19). However, the ongoing and future pandemics caused by SARS-CoV-2 variants and mutations underscore the need for effective vaccines that provide broad-spectrum protection. Here, we developed a nanoparticle vaccine with broad protection against divergent SARS-CoV-2 variants. The corresponding conserved epitopes of the preexisting neutralizing (CePn) antibody were presented on a self-assembling Helicobacter pylori ferritin to generate the CePnF nanoparticle. Intranasal immunization of mice with CePnF nanoparticles induced robust humoral, cellular, and mucosal immune responses and a long-lasting immunity. The CePnF-induced antibodies exhibited cross-reactivity and neutralizing activity against different coronaviruses (CoVs). CePnF vaccination significantly inhibited the replication and pathology of SARS-CoV-2 Delta, WIV04, and Omicron strains in hACE2 transgenic mice and, thus, conferred broad protection against these SARS-CoV-2 variants. Our constructed nanovaccine targeting the conserved epitopes of the preexisting neutralizing antibodies can serve as a promising candidate for a universal SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Xuefan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Heng Rong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jingdi Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Zheng-Li Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Xian-En Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
18
|
Liang Y, Xiao W, Peng Y, Zhang S, Dong J, Zhao J, Wang Y, Zhang M, Liu Z, Yu B. Development of nanoparticle vaccines utilizing designed Fc-binding homo-oligomers and RBD-Fc of SARS-CoV-2. Antiviral Res 2024; 227:105917. [PMID: 38782067 DOI: 10.1016/j.antiviral.2024.105917] [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: 02/18/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
The Fc-fused receptor binding domain (RBD-Fc) vaccine for SARS-CoV-2 has garnered significant attention for its capacity to provide effective and specific immune protection. However, its immunogenicity is limited, highlighting the need for improvement in clinical application. Nanoparticle delivery has been shown to be an effective method for enhancing antigen immunogenicity. In this study, we developed bivalent nanoparticle recombinant protein vaccines by assembling the RBD-Fc of SARS-CoV-2 and Fc-binding homo-oligomers o42.1 and i52.3 into octahedral and icosahedral nanoparticles. The formation of RBD-Fc nanoparticles was confirmed through structural characterization and cell binding experiments. Compared to RBD-Fc dimers, the nanoparticle vaccines induced more potent neutralizing antibodies (nAb) and stronger cellular immune responses. Therefore, using bivalent nanoparticle vaccines based on RBD-Fc presents a promising vaccination strategy against SARS-CoV-2 and offers a universal approach for enhancing the immunogenicity of Fc fusion protein vaccines.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19 Vaccines/immunology
- Immunity, Cellular
- Immunogenicity, Vaccine
- Immunoglobulin Fc Fragments/immunology
- Immunoglobulin Fc Fragments/chemistry
- Mice, Inbred BALB C
- Nanovaccines
- Protein Binding
- Protein Multimerization
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Vaccine Development
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/chemistry
Collapse
Affiliation(s)
- Yucai Liang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Weiling Xiao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Yuan Peng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Shengshuo Zhang
- School of Life Science and Technology, Shandong Second Medical University, Weifang, China
| | - Jinhua Dong
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China; International Research Frontiers Initiative, Tokyo Institute of Technology, Yokohama, Japan
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agriculture Sciences in Weifang, Weifang, Shandong, 261325, China
| | - Yuhui Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Mengtao Zhang
- School of Life Science and Technology, Shandong Second Medical University, Weifang, China
| | - Zhijun Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| | - Bowen Yu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
19
|
Xu M, Wei S, Duan L, Ji Y, Han X, Sun Q, Weng L. The recent advancements in protein nanoparticles for immunotherapy. NANOSCALE 2024; 16:11825-11848. [PMID: 38814163 DOI: 10.1039/d4nr00537f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
In recent years, the advancement of nanoparticle-based immunotherapy has introduced an innovative strategy for combatting diseases. Compared with other types of nanoparticles, protein nanoparticles have obtained substantial attention owing to their remarkable biocompatibility, biodegradability, ease of modification, and finely designed spatial structures. Nature provides several protein nanoparticle platforms, including viral capsids, ferritin, and albumin, which hold significant potential for disease treatment. These naturally occurring protein nanoparticles not only serve as effective drug delivery platforms but also augment antigen delivery and targeting capabilities through techniques like genetic modification and covalent conjugation. Motivated by nature's originality and driven by progress in computational methodologies, scientists have crafted numerous protein nanoparticles with intricate assembly structures, showing significant potential in the development of multivalent vaccines. Consequently, both naturally occurring and de novo designed protein nanoparticles are anticipated to enhance the effectiveness of immunotherapy. This review consolidates the advancements in protein nanoparticles for immunotherapy across diseases including cancer and other diseases like influenza, pneumonia, and hepatitis.
Collapse
Affiliation(s)
- Miaomiao Xu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Siyuan Wei
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Lifan Duan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Yifan Ji
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xiaofan Han
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Qipeng Sun
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
20
|
Ou BS, Baillet J, Filsinger Interrante MV, Adamska JZ, Zhou X, Saouaf OM, Yan J, Klich JH, Jons CK, Meany EL, Valdez AS, Carter L, Pulendran B, King NP, Appel EA. Saponin Nanoparticle Adjuvants Incorporating Toll-Like Receptor Agonists Drive Distinct Immune Signatures and Potent Vaccine Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.16.549249. [PMID: 37577608 PMCID: PMC10418080 DOI: 10.1101/2023.07.16.549249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Over the past few decades, the development of potent and safe immune-activating adjuvant technologies has become the heart of intensive research in the constant fight against highly mutative and immune evasive viruses such as influenza, SARS-CoV-2, and HIV. Herein, we developed a highly modular saponin-based nanoparticle platform incorporating toll-like receptor agonists (TLRas) including TLR1/2a, TLR4a, TLR7/8a adjuvants and their mixtures. These various TLRa-SNP adjuvant constructs induce unique acute cytokine and immune-signaling profiles, leading to specific Th-responses that could be of interest depending on the target disease for prevention. In a murine vaccine study, the adjuvants greatly improved the potency, durability, breadth, and neutralization of both COVID-19 and HIV vaccine candidates, suggesting the potential broad application of these adjuvant constructs to a range of different antigens. Overall, this work demonstrates a modular TLRa-SNP adjuvant platform which could improve the design of vaccines for and dramatically impact modern vaccine development. Teaser Saponin-TLRa nanoadjuvants provide distinct immune signatures and drive potent, broad, durable COVID-19 and HIV vaccine responses.
Collapse
|
21
|
Garmeh Motlagh F, Azimzadeh Irani M, Masoomi Nomandan SZ, Assadizadeh M. Computational design and investigation of the monomeric spike SARS-CoV-2-ferritin nanocage vaccine stability and interactions. Front Mol Biosci 2024; 11:1403635. [PMID: 38933369 PMCID: PMC11199398 DOI: 10.3389/fmolb.2024.1403635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Since the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) outbreak, several solutions have been proposed to manage the disease. The most viable option for controlling this virus is to produce effective vaccines. Most of the current SARS-CoV-2 vaccines have focused on the infusion spike protein. Spike exists as a trimer and plays a vital role in infecting host cells by binding to the Angiotensin-Converting Enzyme 2 (ACE2) receptor through its Receptor Binding Domain (RBD). Ferritin protein, a naturally occurring iron-storage protein, has gained attention for vaccine production due to its self-assembling property, non-toxic nature, and biocompatibility. Ferritin nanocages have recently been employed in the development of a SARS-CoV-2 vaccination eliciting not only long-term protective memory cells but also a sustained antibody response. In this study, a combination of in silico investigations including molecular docking, molecular dynamics simulations, and immune simulations were carried out to computationally model the monomeric spike protein on the ferritin nanocage as well as to evaluate its stability and interactions for the first time. The structural dynamics of the modeled complex demonstrated noticeable stability. In particular, the Receptor Binding Domain (RBD) and ferritin within the monomeric spike-ferritin complex illustrated significant stability. The lack of alterations in the secondary structure further supported the overall steadiness of the complex. The decline in the distance between ferritin and spike suggests a strong interaction over time. The cross-correlation matrices revealed that the monomeric spike and ferritin move towards each other supporting the stable interaction between spike and ferritin. Further, the orientation of monomeric spike protein within the ferritin unit facilitated the exposure of critical epitopes, specifically upward active Receptor Binding Domain (RBD), enabling effective interactions with the ACE2 receptor. The immune simulations of the model indicated high-level stimulations of both cellular and humoral immunity in the human body. It was also found that the employed model is effective regardless of the mutated spikes in different variants. These findings shed light on the current status of the SARS-CoV-2-ferritin nanoparticle vaccines and could be used as a framework for other similar vaccine designs.
Collapse
|
22
|
Pandey KK, Sahoo BR, Pattnaik AK. Protein Nanoparticles as Vaccine Platforms for Human and Zoonotic Viruses. Viruses 2024; 16:936. [PMID: 38932228 PMCID: PMC11209504 DOI: 10.3390/v16060936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Vaccines are one of the most effective medical interventions, playing a pivotal role in treating infectious diseases. Although traditional vaccines comprise killed, inactivated, or live-attenuated pathogens that have resulted in protective immune responses, the negative consequences of their administration have been well appreciated. Modern vaccines have evolved to contain purified antigenic subunits, epitopes, or antigen-encoding mRNAs, rendering them relatively safe. However, reduced humoral and cellular responses pose major challenges to these subunit vaccines. Protein nanoparticle (PNP)-based vaccines have garnered substantial interest in recent years for their ability to present a repetitive array of antigens for improving immunogenicity and enhancing protective responses. Discovery and characterisation of naturally occurring PNPs from various living organisms such as bacteria, archaea, viruses, insects, and eukaryotes, as well as computationally designed structures and approaches to link antigens to the PNPs, have paved the way for unprecedented advances in the field of vaccine technology. In this review, we focus on some of the widely used naturally occurring and optimally designed PNPs for their suitability as promising vaccine platforms for displaying native-like antigens from human viral pathogens for protective immune responses. Such platforms hold great promise in combating emerging and re-emerging infectious viral diseases and enhancing vaccine efficacy and safety.
Collapse
Affiliation(s)
- Kush K. Pandey
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.K.P.); (B.R.S.)
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Bikash R. Sahoo
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.K.P.); (B.R.S.)
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Asit K. Pattnaik
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.K.P.); (B.R.S.)
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
23
|
Cohen AA, Keeffe JR, Schiepers A, Dross SE, Greaney AJ, Rorick AV, Gao H, Gnanapragasam PN, Fan C, West AP, Ramsingh AI, Erasmus JH, Pata JD, Muramatsu H, Pardi N, Lin PJ, Baxter S, Cruz R, Quintanar-Audelo M, Robb E, Serrano-Amatriain C, Magneschi L, Fotheringham IG, Fuller DH, Victora GD, Bjorkman PJ. Mosaic sarbecovirus nanoparticles elicit cross-reactive responses in pre-vaccinated animals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.08.576722. [PMID: 38370696 PMCID: PMC10871317 DOI: 10.1101/2024.02.08.576722] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Immunization with mosaic-8b [60-mer nanoparticles presenting 8 SARS-like betacoronavirus (sarbecovirus) receptor-binding domains (RBDs)] elicits more broadly cross-reactive antibodies than homotypic SARS-CoV-2 RBD-only nanoparticles and protects against sarbecoviruses. To investigate original antigenic sin (OAS) effects on mosaic-8b efficacy, we evaluated effects of prior COVID-19 vaccinations in non-human primates and mice on anti-sarbecovirus responses elicited by mosaic-8b, admix-8b (8 homotypics), or homotypic SARS-CoV-2 immunizations, finding greatest cross-reactivity for mosaic-8b. As demonstrated by molecular fate-mapping in which antibodies from specific cohorts of B cells are differentially detected, B cells primed by WA1 spike mRNA-LNP dominated antibody responses after RBD-nanoparticle boosting. While mosaic-8b- and homotypic-nanoparticles boosted cross-reactive antibodies, de novo antibodies were predominantly induced by mosaic-8b, and these were specific for variant RBDs with increased identity to RBDs on mosaic-8b. These results inform OAS mechanisms and support using mosaic-8b to protect COVID-19 vaccinated/infected humans against as-yet-unknown SARS-CoV-2 variants and animal sarbecoviruses with human spillover potential.
Collapse
Affiliation(s)
- Alexander A. Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- These authors contributed equally
| | - Jennifer R. Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- These authors contributed equally
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, 10065, USA
| | - Sandra E. Dross
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
- National Primate Research Center, Seattle, WA 98121, USA
| | - Allison J. Greaney
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Annie V. Rorick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Han Gao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Chengcheng Fan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Anthony P. West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | - Janice D. Pata
- Wadsworth Center, New York State Department of Health and Department of Biomedical Sciences, University at Albany, Albany, NY, 12201, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Scott Baxter
- Ingenza Ltd, Roslin Innovation Centre, Charnock Bradley Building, Roslin, EH25 9RG, UK
| | - Rita Cruz
- Ingenza Ltd, Roslin Innovation Centre, Charnock Bradley Building, Roslin, EH25 9RG, UK
| | - Martina Quintanar-Audelo
- Ingenza Ltd, Roslin Innovation Centre, Charnock Bradley Building, Roslin, EH25 9RG, UK
- Present address: Centre for Inflammation Research and Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Ellis Robb
- Ingenza Ltd, Roslin Innovation Centre, Charnock Bradley Building, Roslin, EH25 9RG, UK
| | | | - Leonardo Magneschi
- Ingenza Ltd, Roslin Innovation Centre, Charnock Bradley Building, Roslin, EH25 9RG, UK
| | - Ian G. Fotheringham
- Ingenza Ltd, Roslin Innovation Centre, Charnock Bradley Building, Roslin, EH25 9RG, UK
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
- National Primate Research Center, Seattle, WA 98121, USA
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, 10065, USA
| | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Lead contact
| |
Collapse
|
24
|
Yang D, Su M, Guo D, Zhao F, Wang M, Liu J, Zhou J, Sun Y, Yang X, Qi S, Li Z, Zhu Q, Xing X, Li C, Cao Y, Feng L, Sun D. Combination of S1-N-Terminal and S1-C-Terminal Domain Antigens Targeting Double Receptor-Binding Domains Bolsters Protective Immunity of a Nanoparticle Vaccine against Porcine Epidemic Diarrhea Virus. ACS NANO 2024; 18:12235-12260. [PMID: 38696217 DOI: 10.1021/acsnano.4c00809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
Variants of coronavirus porcine epidemic diarrhea virus (PEDV) frequently emerge, causing an incomplete match between the vaccine and variant strains, which affects vaccine efficacy. Designing vaccines with rapidly replaceable antigens and high efficacy is a promising strategy for the prevention of infection with PEDV variant strains. In our study, three different types of self-assembled nanoparticles (nps) targeting receptor-binding N-terminal domain (NTD) and C-terminal domain (CTD) of S1 protein, named NTDnps, CTDnps, and NTD/CTDnps, were constructed and evaluated as vaccine candidates against PEDV. NTDnps and CTDnps vaccines mediated significantly higher neutralizing antibody (NAb) titers than NTD and CTD recombinant proteins in mice. The NTD/CTDnps in varying ratios elicited significantly higher NAb titers when compared with NTDnps and CTDnps alone. The NTD/CTDnps (3:1) elicited NAb with titers up to 92.92% of those induced by the commercial vaccine. Piglets immunized with NTD/CTDnps (3:1) achieved a passive immune protection rate of 83.33% of that induced by the commercial vaccine. NTD/CTDnps (3:1) enhanced the capacity of mononuclear macrophages and dendritic cells to take up and present antigens by activating major histocompatibility complex I and II molecules to stimulate humoral and cellular immunity. These data reveal that a combination of S1-NTD and S1-CTD antigens targeting double receptor-binding domains strengthens the protective immunity of nanoparticle vaccines against PEDV. Our findings will provide a promising vaccine candidate against PEDV.
Collapse
Affiliation(s)
- Dan Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Mingjun Su
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Donghua Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Feiyu Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Meijiao Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Jiaying Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Jingxuan Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Ying Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Xu Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Shanshan Qi
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Zhen Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Qinghe Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Xiaoxu Xing
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Chunqiu Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Yang Cao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| | - Li Feng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, P. R. China
| | - Dongbo Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P. R. China
| |
Collapse
|
25
|
Padariya M, Kalathiya U. Single Ferritin Nanocages Expressing SARS-CoV-2 Spike Variants to Receptor and Antibodies. Vaccines (Basel) 2024; 12:446. [PMID: 38793697 PMCID: PMC11125617 DOI: 10.3390/vaccines12050446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
SARS-CoV-2 virus variants of concern (VOCs) have rapidly changed their transmissibility and pathogenicity primarily through mutations in the structural proteins. Herein, we present molecular details with dynamics of the ferritin nanocages stitched with synthetic chimeras displaying the Spike receptor binding domains (RBDs). Our findings demonstrated the potential usage of ferritin-based vaccines that may effectively inhibit viral entry by blocking the Spike-ACE2 network and may induce cross-protective antibody responses. Taking the nanocage constructs into consideration, we evaluated the effects of variants on the docked interface of the SARS-CoV-2 Spike RBD with the ACE2 (angiotensin-converting enzyme 2) host cell receptor and neutralizing antibodies (Abs). Investigating the VOCs revealed that most of the mutations reported a possibly reduced structural stability within the Spike RBD domain. Point mutations have moderate or no effect for VVH-72, CR3022, and S309 Abs when bound with the Spike RBD, whereas a significant effect was observed for B38, CB6, and m396 over the surface of the H-ferritin nanocage. In addition to providing useful therapeutic approaches against COVID-19 (coronavirus disease 2019), these structural details can also be used to fight future coronavirus outbreaks.
Collapse
Affiliation(s)
- Monikaben Padariya
- International Centre for Cancer Vaccine Science, University of Gdansk, ul. Kładki 24, 80-822 Gdansk, Poland
| | - Umesh Kalathiya
- International Centre for Cancer Vaccine Science, University of Gdansk, ul. Kładki 24, 80-822 Gdansk, Poland
| |
Collapse
|
26
|
Xia X, Li H, Zang J, Cheng S, Du M. Advancements of the Molecular Directed Design and Structure-Activity Relationship of Ferritin Nanocage. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7629-7654. [PMID: 38518374 DOI: 10.1021/acs.jafc.3c09903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Ferritin nanocages possess remarkable structural properties and biological functions, making them highly attractive for applications in functional materials and biomedicine. This comprehensive review presents an overview of the molecular characteristics, extraction and identification of ferritin, ferritin receptors, as well as the advancements in the directional design of high-order assemblies of ferritin and the applications based on its unique structural properties. Specifically, this Review focuses on the regulation of ferritin assembly from one to three dimensions, leveraging the symmetry of ferritin and modifications on key interfaces. Furthermore, it discusses targeted delivery of nutrition and drugs through facile loading and functional modification of ferritin. The aim of this Review is to inspire the design of micro/nano functional materials using ferritin and the development of nanodelivery vehicles for nutritional fortification and disease treatment.
Collapse
Affiliation(s)
- Xiaoyu Xia
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Collaborative Innovation Centre of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Han Li
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Collaborative Innovation Centre of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Jiachen Zang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Shuzhen Cheng
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Collaborative Innovation Centre of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Ming Du
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Collaborative Innovation Centre of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
27
|
Zhang W, Wang H, Wu T, Gao X, Shang Y, Zhang Z, Liu X, Li Y. A SARS-CoV-2 Nanobody Displayed on the Surface of Human Ferritin with High Neutralization Activity. Int J Nanomedicine 2024; 19:2429-2440. [PMID: 38476285 PMCID: PMC10929646 DOI: 10.2147/ijn.s450829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose COVID-19 is rampant throughout the world, which has caused great damage to human lives and seriously hindered the development of the global economy. Aiming at the treatment of SARS-CoV-2, in this study, we proposed a novel fenobody strategy based on ferritin (Fe) self-assembly technology. Methods The neutralizing nanobody H11-D4 of SARS-CoV-2 fused to the C-terminus of end-modified human ferritin was expressed in E. coli and silkworm baculovirus expression systems. A large number of nanoparticles were successfully self-assembled in silkworms, while relatively few nanoparticles can be observed in the treated products from E. coli by electron microscopy. Subsequently, the fenobody's expression level and neutralizing activity were then evaluated. Results The results showed that the IC50 of H11-D4 and fenobody Fe-H11-D4 expressed in E. coli were 171.1 nmol L-1 and 20.87 nmol L-1, respectively. However, the IC50 of Fe-HD11-D4 expressed in silkworms was 1.46 nmol L-1 showing better neutralization activity. Conclusion Therefore, fenobodies can be well self-assembled in silkworm baculovirus expression system, and ferritin self-assembly technology can effectively improve nanobody neutralization activity.
Collapse
Affiliation(s)
- Wenrong Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
- College of Life Sciences, Capital Normal University, Beijing, People’s Republic of China
| | - Haining Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Tong Wu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xintao Gao
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yuting Shang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Zhifang Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xingjian Liu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yinü Li
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| |
Collapse
|
28
|
Li M, Sun X, Chen Y, Wang S, Li Q, Wang Y, Wang Y, Li R, Ding P, Zhang G. Enhancing humoral and mucosal immune response of PED vaccine candidate by fusing S1 protein to nanoparticle multimerization. Vet Microbiol 2024; 290:110003. [PMID: 38262114 DOI: 10.1016/j.vetmic.2024.110003] [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: 11/06/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly infectious pathogen with a high mortality rate, which poses a serious threat to newborn piglets. A rapid, safe and effective vaccine is necessary for protecting pigs from PED infection. Nanoparticles have become molecular scaffolds for displaying soluble antigens due to their unique physical and chemical properties. Here, a vaccine candidate was based on the display of PEDV S1 protein on a mi3 nanoparticle platform using SpyTag/SpyCatcher technology. The size, zeta potential and microstructure of the S1-mi3 NPs were investigated, and their effects on the uptake of antigen-presenting cells (APCs) and maturation of dendritic cells (DCs) were analyzed. Mice were immunized via muscular and intranasal administrations, and the levels of humoral, cellular and mucosal immune responses were analyzed. As a result, S1 proteins were surface-displayed on NPs successfully, which self-assembled into nanoparticles composed of 60 subunits and showed superior safety and stability. In addition, mi3 NPs promoted antigen internalization and dendritic cell (DCs) maturation. In the mouse model, S1-mi3 NPs significantly increased the PEDV-specific antibody including serum IgG, secretory IgA (SIgA) and neutralizing antibodies (NAb). Furthermore, S1-mi3 NPs elicited more CD3+CD4+ and CD3+CD8+ T cell and cellular immune-related cytokines (IFN-γ and IL-4) compared to monomeric S1. In particular, it can induce an effective germinal center-specific (GC) B cell response, which is closely related to the production of neutralizing antibodies. Overall, S1-mi3 NPs are a promising subunit vaccine candidate against PEDV, and this self-assembly NPs also provide an attractive platform for improving vaccine efficacy against emerging pathogens.
Collapse
Affiliation(s)
- Minghui Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Xueke Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yilan Chen
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Siqiao Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Qin Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yanan Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yue Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Ruiqi Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Peiyang Ding
- College of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Longhu Laboratory, Zhengzhou, China.
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Longhu Laboratory, Zhengzhou, China; School of Advanced Agricultural Sciences, Peking University, Beijing 100080, China.
| |
Collapse
|
29
|
Wang E, Cohen AA, Caldera LF, Keeffe JR, Rorick AV, Aida YM, Gnanapragasam PN, Bjorkman PJ, Chakraborty AK. Designed mosaic nanoparticles enhance cross-reactive immune responses in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582544. [PMID: 38464322 PMCID: PMC10925254 DOI: 10.1101/2024.02.28.582544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
1Using computational methods, we designed 60-mer nanoparticles displaying SARS-like betacoronavirus (sarbecovirus) receptor-binding domains (RBDs) by (i) creating RBD sequences with 6 mutations in the SARS-COV-2 WA1 RBD that were predicted to retain proper folding and abrogate antibody responses to variable epitopes (mosaic-2COMs; mosaic-5COM), and (ii) selecting 7 natural sarbecovirus RBDs (mosaic-7COM). These antigens were compared with mosaic-8b, which elicits cross-reactive antibodies and protects from sarbecovirus challenges in animals. Immunizations in naïve and COVID-19 pre-vaccinated mice revealed that mosaic-7COM elicited higher binding and neutralization titers than mosaic-8b and related antigens. Deep mutational scanning showed that mosaic-7COM targeted conserved RBD epitopes. Mosaic-2COMs and mosaic-5COM elicited higher titers than homotypic SARS-CoV-2 Beta RBD-nanoparticles and increased potencies against some SARS-CoV-2 variants than mosaic-7COM. However, mosaic-7COM elicited more potent responses against zoonotic sarbecoviruses and highly mutated Omicrons. These results support using mosaic-7COM to protect against highly mutated SARS-CoV-2 variants and zoonotic sarbecoviruses with spillover potential.
Collapse
Affiliation(s)
- Eric Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- These authors contributed equally
| | - Alexander A. Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- These authors contributed equally
| | - Luis F. Caldera
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- These authors contributed equally
| | - Jennifer R. Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Annie V. Rorick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Yusuf M. Aida
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Present address: School of Clinical Medicine, University of Cambridge, Hills Rd, Cambridge, CB2 0SP, UK
| | | | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Arup K. Chakraborty
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139
- Lead contact
| |
Collapse
|
30
|
Xu D, Powell AE, Utz A, Sanyal M, Do J, Patten JJ, Moliva JI, Sullivan NJ, Davey RA, Kim PS. Design of universal Ebola virus vaccine candidates via immunofocusing. Proc Natl Acad Sci U S A 2024; 121:e2316960121. [PMID: 38319964 PMCID: PMC10873634 DOI: 10.1073/pnas.2316960121] [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: 09/29/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024] Open
Abstract
The Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV)-one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here, we describe a universal Ebola virus vaccine approach using a structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Abigail E. Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA94305
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA94305
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - J. J. Patten
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Juan I. Moliva
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Nancy J. Sullivan
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
- Department of Biology, Boston University, Boston, MA02118
| | - Robert A. Davey
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Chan Zuckerberg Biohub, San Francisco, CA94158
| |
Collapse
|
31
|
Nie J, Zhou Y, Ding F, Liu X, Yao X, Xu L, Chang Y, Li Z, Wang Q, Zhan L, Zhu L, Xie K, Li C, Shi Y, Zhao Q, Shan Y. Self-adjuvant multiepitope nanovaccine based on ferritin induced long-lasting and effective mucosal immunity against H3N2 and H1N1 viruses in mice. Int J Biol Macromol 2024; 259:129259. [PMID: 38191112 DOI: 10.1016/j.ijbiomac.2024.129259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
The influenza A virus (IAV) is a ubiquitous and continuously evolving respiratory pathogen. The intranasal vaccination mimicking natural infections is an attractive strategy for controlling IAVs. Multiepitope vaccines accurately targeting multiple conserved domains have the potential to broaden the protective scope of current seasonal influenza vaccines and reduce the risk of generating escape mutants. Here, multiple linear epitopes from the matrix protein 2 ectodomain (M2e) and the hemagglutinin stem domain (HA2) are fused with the Helicobacter pylori ferritin, a self-assembled nanocarrier and mucosal adjuvant, to develop a multiepitope nanovaccine. Through intranasal delivery, the prokaryotically expressed multiepitope nanovaccine elicits long-lasting mucosal immunity, broad humoral immunity, and robust cellular immunity without any adjuvants, and confers complete protection against H3N2 and H1N1 subtypes of IAV in mice. Importantly, this intranasal multiepitope nanovaccine triggers memory B-cell responses, resulting in secretory immunoglobulin A (sIgA) and serum immunoglobulin G (IgG) levels persisting for more than five months post-immunization. Therefore, this intranasal ferritin-based multiepitope nanovaccine represents a promising approach to combating respiratory pathogens.
Collapse
Affiliation(s)
- Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Yongfei Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Fan Ding
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xiaoxi Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Lipeng Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yaotian Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Zeyu Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Li Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Lvzhou Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Kunpeng Xie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Chenxi Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China; Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| |
Collapse
|
32
|
Sevinc Ozdemir N, Belyaev D, Castro MN, Balakin S, Opitz J, Wihadmadyatami H, Anggraeni R, Yucel D, Kenar H, Beshchasna N, Ana ID, Hasirci V. Advances in In Vitro Blood-Air Barrier Models and the Use of Nanoparticles in COVID-19 Research. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:82-96. [PMID: 37597193 DOI: 10.1089/ten.teb.2023.0117] [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] [Indexed: 08/21/2023]
Abstract
Respiratory infections caused by coronaviruses (CoVs) have become a major public health concern in the past two decades as revealed by the emergence of SARS-CoV in 2002, MERS-CoV in 2012, and SARS-CoV-2 in 2019. The most severe clinical phenotypes commonly arise from exacerbation of immune response following the infection of alveolar epithelial cells localized at the pulmonary blood-air barrier. Preclinical rodent models do not adequately represent the essential genetic properties of the barrier, thus necessitating the use of humanized transgenic models. However, existing monolayer cell culture models have so far been unable to mimic the complex lung microenvironment. In this respect, air-liquid interface models, tissue engineered models, and organ-on-a-chip systems, which aim to better imitate the infection site microenvironment and microphysiology, are being developed to replace the commonly used monolayer cell culture models, and their use is becoming more widespread every day. On the contrary, studies on the development of nanoparticles (NPs) that mimic respiratory viruses, and those NPs used in therapy are progressing rapidly. The first part of this review describes in vitro models that mimic the blood-air barrier, the tissue interface that plays a central role in COVID-19 progression. In the second part of the review, NPs mimicking the virus and/or designed to carry therapeutic agents are explained and exemplified.
Collapse
Affiliation(s)
- Neval Sevinc Ozdemir
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- Department of Medical Biotechnology, ACU Graduate School of Health Sciences, Istanbul, Turkey
- ACU Department of Pharmaceutical Basic Sciences, School of Pharmacy, Istanbul, Turkey
| | - Dmitry Belyaev
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Manuel Nieto Castro
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Sascha Balakin
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Joerg Opitz
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Hevi Wihadmadyatami
- Department of Tissue Engineering and Regenerative Medicine, Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN) and Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
| | - Rahmi Anggraeni
- Department of Tissue Engineering and Regenerative Medicine, Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN) and Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
| | - Deniz Yucel
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- ACU Graduate Department of Biomaterials, Istanbul, Turkey
- Department of Histology and Embryology, ACU School of Medicine, Istanbul, Turkey
| | - Halime Kenar
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- ACU Graduate Department of Biomaterials, Istanbul, Turkey
- ACU Faculty of Engineering Sciences, Department of Biomedical Engineering, Istanbul, Turkey
| | - Natalia Beshchasna
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Maria-Reiche Straße 2, Dresden, Germany
| | - Ika Dewi Ana
- Department of Tissue Engineering and Regenerative Medicine, Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN) and Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada (UGM), Bulaksumur, Yogyakarta, Indonesia
| | - Vasif Hasirci
- Acibadem University (ACU) Biomaterials A&R Center, Atasehir, Istanbul, Turkey
- ACU Graduate Department of Biomaterials, Istanbul, Turkey
- ACU Faculty of Engineering Sciences, Department of Biomedical Engineering, Istanbul, Turkey
- BIOMATEN, METU Ctr. of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey
| |
Collapse
|
33
|
Ou BS, Baillet J, Picece VCT, Gale EC, Powell AE, Saouaf OM, Yan J, Nejatfard A, Lopez Hernandez H, Appel EA. Nanoparticle-Conjugated Toll-Like Receptor 9 Agonists Improve the Potency, Durability, and Breadth of COVID-19 Vaccines. ACS NANO 2024; 18:3214-3233. [PMID: 38215338 PMCID: PMC10832347 DOI: 10.1021/acsnano.3c09700] [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: 10/06/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Development of effective vaccines for infectious diseases has been one of the most successful global health interventions in history. Though, while ideal subunit vaccines strongly rely on antigen and adjuvant(s) selection, the mode and time scale of exposure to the immune system has often been overlooked. Unfortunately, poor control over the delivery of many adjuvants, which play a key role in enhancing the quality and potency of immune responses, can limit their efficacy and cause off-target toxicities. There is a critical need for improved adjuvant delivery technologies to enhance their efficacy and boost vaccine performance. Nanoparticles have been shown to be ideal carriers for improving antigen delivery due to their shape and size, which mimic viral structures but have been generally less explored for adjuvant delivery. Here, we describe the design of self-assembled poly(ethylene glycol)-b-poly(lactic acid) nanoparticles decorated with CpG, a potent TLR9 agonist, to increase adjuvanticity in COVID-19 vaccines. By controlling the surface density of CpG, we show that intermediate valency is a key factor for TLR9 activation of immune cells. When delivered with the SARS-CoV-2 spike protein, CpG nanoparticle (CpG-NP) adjuvant greatly improves the magnitude and duration of antibody responses when compared to soluble CpG, and results in overall greater breadth of immunity against variants of concern. Moreover, encapsulation of CpG-NP into injectable polymeric-nanoparticle (PNP) hydrogels enhances the spatiotemporal control over codelivery of CpG-NP adjuvant and spike protein antigen such that a single immunization of hydrogel-based vaccines generates humoral responses comparable to those of a typical prime-boost regimen of soluble vaccines. These delivery technologies can potentially reduce the costs and burden of clinical vaccination, both of which are key elements in fighting a pandemic.
Collapse
Affiliation(s)
- Ben S. Ou
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Julie Baillet
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Vittoria C. T.
M. Picece
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- Department
of Chemistry and Applied Biosciences, ETH
Zurich, Zurich 8093, Switzerland
| | - Emily C. Gale
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Abigail E. Powell
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
| | - Olivia M. Saouaf
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Jerry Yan
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Anahita Nejatfard
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Eric A. Appel
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Pediatrics - Endocrinology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
34
|
Musunuri S, Weidenbacher PAB, Kim PS. Bringing immunofocusing into focus. NPJ Vaccines 2024; 9:11. [PMID: 38195562 PMCID: PMC10776678 DOI: 10.1038/s41541-023-00792-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/07/2023] [Indexed: 01/11/2024] Open
Abstract
Immunofocusing is a strategy to create immunogens that redirect humoral immune responses towards a targeted epitope and away from non-desirable epitopes. Immunofocusing methods often aim to develop "universal" vaccines that provide broad protection against highly variant viruses such as influenza virus, human immunodeficiency virus (HIV-1), and most recently, severe acute respiratory syndrome coronavirus (SARS-CoV-2). We use existing examples to illustrate five main immunofocusing strategies-cross-strain boosting, mosaic display, protein dissection, epitope scaffolding, and epitope masking. We also discuss obstacles for immunofocusing like immune imprinting. A thorough understanding, advancement, and application of the methods we outline here will enable the design of high-resolution vaccines that protect against future viral outbreaks.
Collapse
Affiliation(s)
- Sriharshita Musunuri
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Department of Biochemistry, Stanford University, Stanford, CA, 94305, USA
| | - Payton A B Weidenbacher
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Peter S Kim
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA.
- Department of Biochemistry, Stanford University, Stanford, CA, 94305, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
35
|
Parisi G, Piacentini R, Incocciati A, Bonamore A, Macone A, Rupert J, Zacco E, Miotto M, Milanetti E, Tartaglia GG, Ruocco G, Boffi A, Di Rienzo L. Design of protein-binding peptides with controlled binding affinity: the case of SARS-CoV-2 receptor binding domain and angiotensin-converting enzyme 2 derived peptides. Front Mol Biosci 2024; 10:1332359. [PMID: 38250735 PMCID: PMC10797010 DOI: 10.3389/fmolb.2023.1332359] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
The development of methods able to modulate the binding affinity between proteins and peptides is of paramount biotechnological interest in view of a vast range of applications that imply designed polypeptides capable to impair or favour Protein-Protein Interactions. Here, we applied a peptide design algorithm based on shape complementarity optimization and electrostatic compatibility and provided the first experimental in vitro proof of the efficacy of the design algorithm. Focusing on the interaction between the SARS-CoV-2 Spike Receptor-Binding Domain (RBD) and the human angiotensin-converting enzyme 2 (ACE2) receptor, we extracted a 23-residues long peptide that structurally mimics the major interacting portion of the ACE2 receptor and designed in silico five mutants of such a peptide with a modulated affinity. Remarkably, experimental KD measurements, conducted using biolayer interferometry, matched the in silico predictions. Moreover, we investigated the molecular determinants that govern the variation in binding affinity through molecular dynamics simulation, by identifying the mechanisms driving the different values of binding affinity at a single residue level. Finally, the peptide sequence with the highest affinity, in comparison with the wild type peptide, was expressed as a fusion protein with human H ferritin (HFt) 24-mer. Solution measurements performed on the latter constructs confirmed that peptides still exhibited the expected trend, thereby enhancing their efficacy in RBD binding. Altogether, these results indicate the high potentiality of this general method in developing potent high-affinity vectors for hindering/enhancing protein-protein associations.
Collapse
Affiliation(s)
- Giacomo Parisi
- Department of Basic and Applied Sciences for Engineering (SBAI), Università“Sapienza”, Roma, Italy
| | - Roberta Piacentini
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Università“Sapienza”, Roma, Italy
| | - Alessio Incocciati
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Università“Sapienza”, Roma, Italy
| | - Alessandra Bonamore
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Università“Sapienza”, Roma, Italy
| | - Alberto Macone
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Università“Sapienza”, Roma, Italy
| | - Jakob Rupert
- Department of Biology and Biotechnologies “Charles Darwin”, Università“Sapienza”, Roma, Italy
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Elsa Zacco
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Mattia Miotto
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Roma, Italy
| | - Edoardo Milanetti
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Roma, Italy
- Department of Physics, Università“Sapienza”, Roma, Italy
| | - Gian Gaetano Tartaglia
- Department of Biology and Biotechnologies “Charles Darwin”, Università“Sapienza”, Roma, Italy
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Giancarlo Ruocco
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Roma, Italy
- Department of Physics, Università“Sapienza”, Roma, Italy
| | - Alberto Boffi
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Università“Sapienza”, Roma, Italy
| | - Lorenzo Di Rienzo
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Roma, Italy
| |
Collapse
|
36
|
Matsuura K, Furukawa H. [Creation of enveloped viral replica equipped with SARS-CoV2 spike protein]. Uirusu 2024; 74:159-164. [PMID: 40024799 DOI: 10.2222/jsv.74.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
|
37
|
Li Y, Gao H, Nepovimova E, Wu Q, Adam V, Kuca K. Recombinant ferritins for multimodal nanomedicine. J Enzyme Inhib Med Chem 2023; 38:2219868. [PMID: 37263586 DOI: 10.1080/14756366.2023.2219868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023] Open
Abstract
In all living organisms, ferritins are a group of proteins important for maintaining iron homeostasis. Increasing amount of studies has shown that recombinant ferritins can be widely used in multimodal nanomedicine, especially for anticancer treatment and vaccination. Recombinant particles prepared by fusing viral proteins and ferritin subunits produce a better immune response and higher antibody titres. Moreover, actively-targeted ferritin nanoparticles can recognise receptors and deliver natural or chemical drugs specifically to the tumour tissue. In addition, ferritin-linked or loaded with contrast agents or fluorescent dyes can be used as multimodal particles useful cancer theranostics. In this review, we fully summarised the unitisation of recombinant ferritins in multimodal nanomedicine. The research progress of using recombinant ferritins as nanovaccines, nanozymes, and bioengineered nanocarriers for targeted therapy and bioimaging is emphasised.
Collapse
Affiliation(s)
- Yihao Li
- College of Life Science, Yangtze University, Jingzhou, China
| | - Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Králové, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Králové, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Králové, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
38
|
Ou BS, Saouaf OM, Yan J, Bruun TUJ, Baillet J, Zhou X, King NP, Appel EA. Broad and Durable Humoral Responses Following Single Hydrogel Immunization of SARS-CoV-2 Subunit Vaccine. Adv Healthc Mater 2023; 12:e2301495. [PMID: 37278391 DOI: 10.1002/adhm.202301495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 06/07/2023]
Abstract
Most vaccines require several immunizations to induce robust immunity, and indeed, most SARS-CoV-2 vaccines require an initial two-shot regimen followed by several boosters to maintain efficacy. Such a complex series of immunizations unfortunately increases the cost and complexity of populations-scale vaccination and reduces overall compliance and vaccination rate. In a rapidly evolving pandemic affected by the spread of immune-escaping variants, there is an urgent need to develop vaccines capable of providing robust and durable immunity. In this work, a single immunization SARS-CoV-2 subunit vaccine is developed that can rapidly generate potent, broad, and durable humoral immunity. Injectable polymer-nanoparticle (PNP) hydrogels are leveraged as a depot technology for the sustained delivery of a nanoparticle antigen (RND-NP) displaying multiple copies of the SARS-CoV-2 receptor-binding domain (RBD) and potent adjuvants including CpG and 3M-052. Compared to a clinically relevant prime-boost regimen with soluble vaccines formulated with CpG/alum or 3M-052/alum adjuvants, PNP hydrogel vaccines more rapidly generated higher, broader, and more durable antibody responses. Additionally, these single-immunization hydrogel-based vaccines elicit potent and consistent neutralizing responses. Overall, it is shown that PNP hydrogels elicit improved anti-COVID immune responses with only a single administration, demonstrating their potential as critical technologies to enhance overall pandemic readiness.
Collapse
Affiliation(s)
- Ben S Ou
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Olivia M Saouaf
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Jerry Yan
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Theodora U J Bruun
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Julie Baillet
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
- CNRS, Bordeaux INP, LCPO, University of Bordeaux, Pessac, 33600, France
| | - Xueting Zhou
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, 98109, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pediatrics-Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Woods Institute for the Environment, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
39
|
Kumru OS, Sanyal M, Friedland N, Hickey JM, Joshi R, Weidenbacher P, Do J, Cheng YC, Kim PS, Joshi SB, Volkin DB. Formulation development and comparability studies with an aluminum-salt adjuvanted SARS-CoV-2 spike ferritin nanoparticle vaccine antigen produced from two different cell lines. Vaccine 2023; 41:6502-6513. [PMID: 37620203 PMCID: PMC11181998 DOI: 10.1016/j.vaccine.2023.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
The development of safe and effective second-generation COVID-19 vaccines to improve affordability and storage stability requirements remains a high priority to expand global coverage. In this report, we describe formulation development and comparability studies with a self-assembled SARS-CoV-2 spike ferritin nanoparticle vaccine antigen (called DCFHP), when produced in two different cell lines and formulated with an aluminum-salt adjuvant (Alhydrogel, AH). Varying levels of phosphate buffer altered the extent and strength of antigen-adjuvant interactions, and these formulations were evaluated for their (1) in vivo performance in mice and (2) in vitro stability profiles. Unadjuvanted DCFHP produced minimal immune responses while AH-adjuvanted formulations elicited greatly enhanced pseudovirus neutralization titers independent of ∼100%, ∼40% or ∼10% of the DCFHP antigen adsorbed to AH. These formulations differed, however, in their in vitro stability properties as determined by biophysical studies and a competitive ELISA for measuring ACE2 receptor binding of AH-bound antigen. Interestingly, after one month of 4°C storage, small increases in antigenicity with concomitant decreases in the ability to desorb the antigen from the AH were observed. Finally, we performed a comparability assessment of DCFHP antigen produced in Expi293 and CHO cells, which displayed expected differences in their N-linked oligosaccharide profiles. Despite consisting of different DCFHP glycoforms, these two preparations were highly similar in their key quality attributes including molecular size, structural integrity, conformational stability, binding to ACE2 receptor and mouse immunogenicity profiles. Taken together, these studies support future preclinical and clinical development of an AH-adjuvanted DCFHP vaccine candidate produced in CHO cells.
Collapse
Affiliation(s)
- Ozan S Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Natalia Friedland
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - John M Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Richa Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Payton Weidenbacher
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Ya-Chen Cheng
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Peter S Kim
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
40
|
Blanchet CE, Round A, Mertens HDT, Ayyer K, Graewert M, Awel S, Franke D, Dörner K, Bajt S, Bean R, Custódio TF, de Wijn R, Juncheng E, Henkel A, Gruzinov A, Jeffries CM, Kim Y, Kirkwood H, Kloos M, Knoška J, Koliyadu J, Letrun R, Löw C, Makroczyova J, Mall A, Meijers R, Pena Murillo GE, Oberthür D, Round E, Seuring C, Sikorski M, Vagovic P, Valerio J, Wollweber T, Zhuang Y, Schulz J, Haas H, Chapman HN, Mancuso AP, Svergun D. Form factor determination of biological molecules with X-ray free electron laser small-angle scattering (XFEL-SAS). Commun Biol 2023; 6:1057. [PMID: 37853181 PMCID: PMC10585004 DOI: 10.1038/s42003-023-05416-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
Free-electron lasers (FEL) are revolutionizing X-ray-based structural biology methods. While protein crystallography is already routinely performed at FELs, Small Angle X-ray Scattering (SAXS) studies of biological macromolecules are not as prevalent. SAXS allows the study of the shape and overall structure of proteins and nucleic acids in solution, in a quasi-native environment. In solution, chemical and biophysical parameters that have an influence on the structure and dynamics of molecules can be varied and their effect on conformational changes can be monitored in time-resolved XFEL and SAXS experiments. We report here the collection of scattering form factors of proteins in solution using FEL X-rays. The form factors correspond to the scattering signal of the protein ensemble alone; the scattering contributions from the solvent and the instrument are separately measured and accurately subtracted. The experiment was done using a liquid jet for sample delivery. These results pave the way for time-resolved studies and measurements from dilute samples, capitalizing on the intense and short FEL X-ray pulses.
Collapse
Affiliation(s)
- Clement E Blanchet
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany.
| | - Adam Round
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany.
| | - Haydyn D T Mertens
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
| | - Kartik Ayyer
- The Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
- Max Planck Institute for the Structure and Dynamics of Matter, Luruper Chaussee 149, 22761, Hamburg, Germany
| | - Melissa Graewert
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
| | - Salah Awel
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Daniel Franke
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
- BIOSAXS GmbH, Notkestr. 85, 22607, Hamburg, Germany
| | - Katerina Dörner
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Saša Bajt
- The Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Richard Bean
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Tânia F Custódio
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
| | - Raphael de Wijn
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - E Juncheng
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Alessandra Henkel
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Andrey Gruzinov
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
| | - Cy M Jeffries
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
| | - Yoonhee Kim
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Henry Kirkwood
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Marco Kloos
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Juraj Knoška
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | | | - Romain Letrun
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Christian Löw
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
| | | | - Abhishek Mall
- Max Planck Institute for the Structure and Dynamics of Matter, Luruper Chaussee 149, 22761, Hamburg, Germany
| | - Rob Meijers
- Institute for Protein Innovation (IPI), 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Gisel Esperanza Pena Murillo
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Dominik Oberthür
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Ekaterina Round
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Carolin Seuring
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
- Department of Chemistry, University of Hamburg, Hamburg, Germany
- Leibniz Institute of Virology, Hamburg, Germany
| | - Marcin Sikorski
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Patrik Vagovic
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
| | - Joana Valerio
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | - Tamme Wollweber
- The Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
- Max Planck Institute for the Structure and Dynamics of Matter, Luruper Chaussee 149, 22761, Hamburg, Germany
| | - Yulong Zhuang
- The Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
- Max Planck Institute for the Structure and Dynamics of Matter, Luruper Chaussee 149, 22761, Hamburg, Germany
| | - Joachim Schulz
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
| | | | - Henry N Chapman
- The Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607, Hamburg, Germany
- Department of Physics, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
| | - Adrian P Mancuso
- European XFEL GmbH, Holzkoppel 4, 22869, Schenefeld, Germany
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Dmitri Svergun
- European Molecular Biology Laboratory EMBL, Hamburg Site, c/o DESY Notkestrasse 85, 22603, Hamburg, Germany.
- BIOSAXS GmbH, Notkestr. 85, 22607, Hamburg, Germany.
| |
Collapse
|
41
|
Xu D, Powell AE, Utz A, Sanyal M, Do J, Patten J, Moliva JI, Sullivan NJ, Davey RA, Kim PS. Design of universal Ebola virus vaccine candidates via immunofocusing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.14.562364. [PMID: 37904982 PMCID: PMC10614775 DOI: 10.1101/2023.10.14.562364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV) - one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here we describe a universal Ebola virus vaccine approach using structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Abigail E. Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - J.J. Patten
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Juan I. Moliva
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nancy J. Sullivan
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biology, Boston University, Boston, MA 02118, USA
| | - Robert A. Davey
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
42
|
Hou Y, Chen M, Bian Y, Zheng X, Tong R, Sun X. Advanced subunit vaccine delivery technologies: From vaccine cascade obstacles to design strategies. Acta Pharm Sin B 2023; 13:3321-3338. [PMID: 37655334 PMCID: PMC10465871 DOI: 10.1016/j.apsb.2023.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Designing and manufacturing safe and effective vaccines is a crucial challenge for human health worldwide. Research on adjuvant-based subunit vaccines is increasingly being explored to meet clinical needs. Nevertheless, the adaptive immune responses of subunit vaccines are still unfavorable, which may partially be attributed to the immune cascade obstacles and unsatisfactory vaccine design. An extended understanding of the crosstalk between vaccine delivery strategies and immunological mechanisms could provide scientific insight to optimize antigen delivery and improve vaccination efficacy. In this review, we summarized the advanced subunit vaccine delivery technologies from the perspective of vaccine cascade obstacles after administration. The engineered subunit vaccines with lymph node and specific cell targeting ability, antigen cross-presentation, T cell activation properties, and tailorable antigen release patterns may achieve effective immune protection with high precision, efficiency, and stability. We hope this review can provide rational design principles and inspire the exploitation of future subunit vaccines.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xi Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
43
|
Ko HJ, Kim YJ. Antigen Delivery Systems: Past, Present, and Future. Biomol Ther (Seoul) 2023; 31:370-387. [PMID: 37072288 PMCID: PMC10315343 DOI: 10.4062/biomolther.2023.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 04/20/2023] Open
Abstract
The COVID-19 pandemic has increased demand for safe and effective vaccines. Research to develop vaccines against diseases including Middle East respiratory syndrome, Ebolavirus, human immunodeficiency virus, and various cancers would also contribute to global well-being. For successful vaccine development, the advancement of technologies such as antigen (Ag) screening, Ag delivery systems and adjuvants, and manufacturing processes is essential. Ag delivery systems are required not only to deliver a sufficient amount of Ag for vaccination, but also to enhance immune response. In addition, Ag types and their delivery systems determine the manufacturing processes of the vaccine product. Here, we analyze the characteristics of various Ag delivery systems: plasmids, viral vectors, bacterial vectors, nanoparticles, self-assembled particles, natural and artificial cells, and extracellular vesicles. This review provides insight into the current vaccine landscape and highlights promising avenues of research for the development and improvement of Ag delivery systems.
Collapse
Affiliation(s)
- Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeon-Jeong Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Inje University, Gimhae 50834, Republic of Korea
- Inje Institute of Pharmaceutical Science and Research, Inje University, Gimhae 50834, Republic of Korea
- Smart Marine Therapeutic Center, Inje University, Gimhae 50834, Republic of Korea
| |
Collapse
|
44
|
Shesh BP, Connor JR. A novel view of ferritin in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188917. [PMID: 37209958 PMCID: PMC10330744 DOI: 10.1016/j.bbcan.2023.188917] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/13/2023] [Accepted: 05/13/2023] [Indexed: 05/22/2023]
Abstract
Since its discovery more than 85 years ago, ferritin has principally been known as an iron storage protein. However, new roles, beyond iron storage, are being uncovered. Novel processes involving ferritin such as ferritinophagy and ferroptosis and as a cellular iron delivery protein not only expand our thinking on the range of contributions of this protein but present an opportunity to target these pathways in cancers. The key question we focus on within this review is whether ferritin modulation represents a useful approach for treating cancers. We discussed novel functions and processes of this protein in cancers. We are not limiting this review to cell intrinsic modulation of ferritin in cancers, but also focus on its utility in the trojan horse approach in cancer therapeutics. The novel functions of ferritin as discussed herein realize the multiple roles of ferritin in cell biology that can be probed for therapeutic opportunities and further research.
Collapse
Affiliation(s)
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
45
|
Weidenbacher PAB, Friedland N, Sanyal M, Morris MK, Do J, Hanson C, Kim PS. Decreased efficacy of a COVID-19 vaccine due to mutations present in early SARS-CoV-2 variants of concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546764. [PMID: 37425802 PMCID: PMC10326996 DOI: 10.1101/2023.06.27.546764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
With the SARS-CoV-2 virus still circulating and evolving, there remains an outstanding question if variant-specific vaccines represent the optimal path forward, or if other strategies might be more efficacious towards providing broad protection against emerging variants. Here, we examine the efficacy of strain-specific variants of our previously reported, pan-sarbecovirus vaccine candidate, DCFHP-alum, a ferritin nanoparticle functionalized with an engineered form of the SARS-CoV-2 spike protein. In non-human primates, DCFHP-alum elicits neutralizing antibodies against all known VOCs that have emerged to date and SARS-CoV-1. During development of the DCFHP antigen, we investigated the incorporation of strain-specific mutations from the major VOCs that had emerged to date: D614G, Epsilon, Alpha, Beta, and Gamma. Here, we report the biochemical and immunological characterizations that led us to choose the ancestral Wuhan-1 sequence as the basis for the final DCFHP antigen design. Specifically, we show by size exclusion chromatography and differential scanning fluorimetry that mutations in the VOCs adversely alter the antigen's structure and stability. More importantly, we determined that DCFHP without strain-specific mutations elicits the most robust, cross-reactive response in both pseudovirus and live virus neutralization assays. Our data suggest potential limitations to the variant-chasing approach in the development of protein nanoparticle vaccines, but also have implications for other approaches including mRNA-based vaccines.
Collapse
Affiliation(s)
- Payton A.-B. Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Natalia Friedland
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jonathan Do
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Carl Hanson
- California Department of Public Health, Richmond, CA, USA
| | - Peter S. Kim
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
46
|
Zahid AA, Chakraborty A, Luo W, Coyle A, Paul A. Tailoring the Inherent Properties of Biobased Nanoparticles for Nanomedicine. ACS Biomater Sci Eng 2023. [PMID: 37378614 DOI: 10.1021/acsbiomaterials.3c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Biobased nanoparticles are at the leading edge of the rapidly developing field of nanomedicine and biotherapeutics. Their unique size, shape, and biophysical properties make them attractive tools for biomedical research, including vaccination, targeted drug delivery, and immune therapy. These nanoparticles are engineered to present native cell receptors and proteins on their surfaces, providing a biomimicking camouflage for therapeutic cargo to evade rapid degradation, immune rejection, inflammation, and clearance. Despite showing promising clinical relevance, commercial implementation of these biobased nanoparticles is yet to be fully realized. In this perspective, we discuss advanced biobased nanoparticle designs used in medical applications, such as cell membrane nanoparticles, exosomes, and synthetic lipid-derived nanoparticles, and highlight their benefits and potential challenges. Moreover, we critically assess the future of preparing such particles using artificial intelligence and machine learning. These advanced computational tools will be able to predict the functional composition and behavior of the proteins and cell receptors present on the nanoparticle surfaces. With more advancement in designing new biobased nanoparticles, this field of research could play a key role in dictating the future rational design of drug transporters, thereby ultimately improving overall therapeutic outcomes.
Collapse
Affiliation(s)
- Alap Ali Zahid
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Wei Luo
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Ali Coyle
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- Department of Chemistry, The Centre for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| |
Collapse
|
47
|
Liu X, Song H, Jiang J, Gao X, Yi Y, Shang Y, Li J, Li D, Zeng Z, Li Y, Zhang Z. Baculovirus-expressed self-assembling SARS-CoV-2 nanoparticle vaccines targeting the S protein induce protective immunity in mice. Process Biochem 2023; 129:200-208. [PMID: 37007452 PMCID: PMC10038678 DOI: 10.1016/j.procbio.2023.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Spike (S) protein, a homotrimeric glycoprotein, is the most important antigen target for SARS-CoV-2 vaccines. A complete simulation of the advanced structure of this homotrimer during subunit vaccine development is the most likely method to improve its immunoprotective effects. In this study, preparation strategies for the S protein receptor-binding domain, S1 region, and ectodomain trimer nanoparticles were designed using ferritin nanoparticle self-assembly technology. The Bombyx mori baculovirus expression system was used to prepare three nanoparticle vaccines with high expression levels recorded in silkworms. The results in mice showed that the nanoparticle vaccine prepared using this strategy could induce immune responses when administered via both the subcutaneous administration and oral routes. Given the stability of these ferritin-based nanoparticle vaccines, an easy-to-use and low-cost oral immunization strategy can be employed in vaccine blind areas attributed to shortages of ultralow-temperature equipment and medical resources in underdeveloped areas. Oral vaccines are also promising candidates for limiting the spread of SARS-CoV-2 in domestic and farmed animals, especially in stray and wild animals.
Collapse
Affiliation(s)
- Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Haozhi Song
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianmin Jiang
- Key Laboratory of Vaccine, Prevention and control of Infectious disease of Zhejiang Province, Zhejiang Provincial Center For Disease Control And Prevention, Hangzhou, Zhejiang Province, China
| | - Xintao Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yongzhu Yi
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuting Shang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jialei Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dan Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhen Zeng
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
48
|
Wang C, Liu Q, Huang X, Zhuang J. Ferritin nanocages: a versatile platform for nanozyme design. J Mater Chem B 2023; 11:4153-4170. [PMID: 37158014 DOI: 10.1039/d3tb00192j] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Nanozymes are a class of nanomaterials with enzyme-like activities and have attracted increasing attention due to their potential applications in biomedicine. However, nanozyme design incorporating the desired properties remains challenging. Natural or genetically engineered protein scaffolds, such as ferritin nanocages, have emerged as a promising platform for nanozyme design due to their unique protein structure, natural biomineralization capacity, self-assembly properties, and high biocompatibility. In this review, we highlight the intrinsic properties of ferritin nanocages, especially for nanozyme design. We also discuss the advantages of genetically engineered ferritin in the versatile design of nanozymes over natural ferritin. Additionally, we summarize the bioapplications of ferritin-based nanozymes based on their enzyme-mimicking activities. In this perspective, we mainly provide potential insights into the utilization of ferritin nanocages for nanozyme design.
Collapse
Affiliation(s)
- Chunyu Wang
- School of Medicine, Nankai University, Tianjin 300071, China.
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.
| | - Qiqi Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China.
| | - Jie Zhuang
- School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
49
|
Byrum JR, Waltari E, Janson O, Guo SM, Folkesson J, Chhun BB, Vinden J, Ivanov IE, Forst ML, Li H, Larson AG, Blackmon L, Liu Z, Wu W, Ahyong V, Tato CM, McCutcheon KM, Hoh R, Kelly JD, Martin JN, Peluso MJ, Henrich TJ, Deeks SG, Prakash M, Greenhouse B, Mehta SB, Pak JE. MultiSero: An Open-Source Multiplex-ELISA Platform for Measuring Antibody Responses to Infection. Pathogens 2023; 12:671. [PMID: 37242341 PMCID: PMC10221076 DOI: 10.3390/pathogens12050671] [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: 03/31/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
A multiplexed enzyme-linked immunosorbent assay (ELISA) that simultaneously measures antibody binding to multiple antigens can extend the impact of serosurveillance studies, particularly if the assay approaches the simplicity, robustness, and accuracy of a conventional single-antigen ELISA. Here, we report on the development of multiSero, an open-source multiplex ELISA platform for measuring antibody responses to viral infection. Our assay consists of three parts: (1) an ELISA against an array of proteins in a 96-well format; (2) automated imaging of each well of the ELISA array using an open-source plate reader; and (3) automated measurement of optical densities for each protein within the array using an open-source analysis pipeline. We validated the platform by comparing antibody binding to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) antigens in 217 human sera samples, showing high sensitivity (0.978), specificity (0.977), positive predictive value (0.978), and negative predictive value (0.977) for classifying seropositivity, a high correlation of multiSero determined antibody titers with commercially available SARS-CoV-2 antibody tests, and antigen-specific changes in antibody titer dynamics upon vaccination. The open-source format and accessibility of our multiSero platform can contribute to the adoption of multiplexed ELISA arrays for serosurveillance studies, for SARS-CoV-2 and other pathogens of significance.
Collapse
Affiliation(s)
- Janie R. Byrum
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Eric Waltari
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Owen Janson
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
- EPPIcenter Program, University of California, San Francisco, CA 94143, USA
| | - Syuan-Ming Guo
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Jenny Folkesson
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Bryant B. Chhun
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Joanna Vinden
- Infectious Diseases and Immunity Graduate Program, University of California, Berkeley, CA 94720-3370, USA
| | - Ivan E. Ivanov
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Marcus L. Forst
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Hongquan Li
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Adam G. Larson
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lena Blackmon
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Ziwen Liu
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Wesley Wu
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Vida Ahyong
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Cristina M. Tato
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | | | - Rebecca Hoh
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94158, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94158, USA
| | - Michael J. Peluso
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California, San Francisco, CA 94110, USA
| | - Steven G. Deeks
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
| | - Manu Prakash
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Bryan Greenhouse
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
- EPPIcenter Program, University of California, San Francisco, CA 94143, USA
| | - Shalin B. Mehta
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - John E. Pak
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
50
|
Hoffmann MAG, Yang Z, Huey-Tubman KE, Cohen AA, Gnanapragasam PNP, Nakatomi LM, Storm KN, Moon WJ, Lin PJC, West AP, Bjorkman PJ. ESCRT recruitment to SARS-CoV-2 spike induces virus-like particles that improve mRNA vaccines. Cell 2023; 186:2380-2391.e9. [PMID: 37146611 PMCID: PMC10121106 DOI: 10.1016/j.cell.2023.04.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/28/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023]
Abstract
Prime-boost regimens for COVID-19 vaccines elicit poor antibody responses against Omicron-based variants and employ frequent boosters to maintain antibody levels. We present a natural infection-mimicking technology that combines features of mRNA- and proteinnanoparticle-based vaccines through encoding self-assembling enveloped virus-like particles (eVLPs). eVLP assembly is achieved by inserting an ESCRT- and ALIX-binding region (EABR) into the SARS-CoV-2 spike cytoplasmic tail, which recruits ESCRT proteins to induce eVLP budding from cells. Purified spike-EABR eVLPs presented densely arrayed spikes and elicited potent antibody responses in mice. Two immunizations with mRNA-LNP encoding spike-EABR elicited potent CD8+ T cell responses and superior neutralizing antibody responses against original and variant SARS-CoV-2 compared with conventional spike-encoding mRNA-LNP and purified spike-EABR eVLPs, improving neutralizing titers >10-fold against Omicron-based variants for 3 months post-boost. Thus, EABR technology enhances potency and breadth of vaccine-induced responses through antigen presentation on cell surfaces and eVLPs, enabling longer-lasting protection against SARS-CoV-2 and other viruses.
Collapse
Affiliation(s)
- Magnus A G Hoffmann
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Zhi Yang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Kathryn E Huey-Tubman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Alexander A Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Leesa M Nakatomi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Kaya N Storm
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | - Anthony P West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|