1
|
Wu H, Weng R, Li J, Huang Z, Tie X, Li J, Chen K. Self-Assembling protein nanoparticle platform for multivalent antigen delivery in vaccine development. Int J Pharm 2025; 676:125597. [PMID: 40233885 DOI: 10.1016/j.ijpharm.2025.125597] [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: 01/26/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
Nanoparticle vaccines can efficiently and repeatedly display multivalent antigens, thereby improving the targeted delivery of antigens and inducing more durable immune responses, making them an important representative of novel vaccines. The global COVID-19 pandemic has accelerated the development of nanoparticle vaccines, offering a promising solution for the prevention and control of infectious diseases. Currently, the development of nanoparticle vaccines involves the use of various types of nanoparticles, including liposomes, polymers, inorganic materials, and emulsions. Protein nanoparticles candidate vaccines are attracting increasing attention because of their unique antigen presentation methods and self-assembly characteristics during their development, leading to a broad consensus on their promising future. Naturally self-assembling protein nanoparticles, such as ferritin, enhance antigen presentation, which aids in the activation of both humoral and cellular immune responses. This has led to significant advancements in the study of hepatitis B virus. Meanwhile, some synthetically engineered protein nanoparticles, such as mi3, and I53-50, can induce higher antibody titers through chemical conjugation with the SpyTag-SpyCatcher system, thereby providing better immunoprotection and showing promising prospects in the prevention of H1N1 and H3N2 influenza virus infections. This article reviews the unique advantages of protein nanoparticles as antigen delivery platforms, progress made in immunological design mechanisms, advances in the application of related adjuvants in preclinical and clinical trials, and the performance of commonly used computationally designed protein nanoparticles in preclinical trials, with a particular emphasis on the progress in the application of cationic nanoparticle vaccines. The aim is to provide future researchers with effective adjuvant strategies and high-quality selections for computationally designed protein nanoparticles, thereby promoting the clinical trial process of protein nanoparticles vaccines.
Collapse
Affiliation(s)
- Hao Wu
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Ruiqi Weng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jiaxuan Li
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiwei Huang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Xiaotian Tie
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jianhua Li
- Zhejiang Key Laboratory of Public Health Detection and Pathogenesis Research, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, PR China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China.
| |
Collapse
|
2
|
Yang C, Li E, Guo X, Xie W, Wang Y, Huang X, Chiu S, Wu X. A Self-Assembled Nanovaccine with BA.4/5 Receptor-Binding Domain and CpG Oligodeoxynucleotides Induces Broad-Spectrum Neutralization against SARS-CoV-2 Omicron Subvariants. ACS NANO 2025; 19:16424-16437. [PMID: 40265996 DOI: 10.1021/acsnano.4c17269] [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: 04/24/2025]
Abstract
Over the past 3 years, SARS-CoV-2 Omicron has been circulating globally with the emergence of multiple subvariants, including BA.5, BA.5.2, XBB, XBB.1, EG.5.1, HK.3, BA.2.86, JN.1, and KP.2. To combat these Omicron subvariants, several vaccines based on receptor-binding domain (RBD) dimers have been developed; however, RBD dimer vaccines require frequent updates to cope with the emergence of new variants. In contrast, the development of a safe, effective, and broad-spectrum vaccine against multiple Omicron subvariants, including the latest JN.1 and KP.2, would be a one-size-fits-all solution. Here, we designed BA.4/5 RBD-PC7A conjugate micelles by displaying the BA.4/5 RBD in PC7A micelles. Remarkably, the micelles elicited potent neutralizing antibodies (NAbs) in rabbits, effectively neutralizing BA.5.2, XBB.1.18, and HK.3 infections. Moreover, the micelles alone were able to induce NAbs in mice against the BA.5 variant. When a cytosine-phosphate-guanine (CpG) adjuvant was added and electrostatically adsorbed to the micelles, there was a significant increase in the antibody titers of IgG1, IgG2b, and IgG2c. This enhancement facilitated the broad neutralization of various strains, including BA.5.2, XBB.1.18, HK.3, JN.1, and KP.2. Furthermore, the micelles adsorbed with CpG protected golden hamsters from infection with the BA.5.2 strain. This study presents a potent and broadly neutralizing nanovaccine that includes the BA.4/5 RBD antigen and a CpG adjuvant. It demonstrates efficacy against multiple Omicron subvariants, including BA.5, BA.5.2, XBB.1.18, HK.3, JN.1, and KP.2, highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Chendong Yang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong Key Laboratory of Carbohydrate and Carbohydrate-conjugate Drugs, Shandong University, Qingdao, Shandong 266237, China
| | - Entao Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaoping Guo
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wenyu Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yuanzhan Wang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong Key Laboratory of Carbohydrate and Carbohydrate-conjugate Drugs, Shandong University, Qingdao, Shandong 266237, China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Sandra Chiu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui 230027, China
| | - Xuanjun Wu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong Key Laboratory of Carbohydrate and Carbohydrate-conjugate Drugs, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
3
|
Wang P, Zhang G, Liu Q, Jiang L, Niu X, Fang M, Gao W, He Y, Li Y, Shu J, Zhang S. Immunogenicity assessment of a swine fever-swine circovirus type 2 duplex vaccine candidate created using the Zera nanoparticle delivery method. Vet Immunol Immunopathol 2025; 284:110929. [PMID: 40250144 DOI: 10.1016/j.vetimm.2025.110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/14/2025] [Accepted: 03/30/2025] [Indexed: 04/20/2025]
Abstract
Classical swine fever virus (CSFV) and porcine circovirus (PCV) are the primary pathogens of swine fever and pig circovirus infections, respectively. Co-infections with these diseases result in severe economic losses for the swine sector. To prevent and control the disease, immunization is the primary technique for reducing co-infections and economic losses. In this work, we created a new nanoparticle vaccine using a Zera nanoparticle delivery system that expressed swine fever virus E0 and E2 proteins as well as porcine circovirus Cap proteins. We then inoculated BALB/c mice to test the vaccine's immunogenicity. The findings revealed that this nanoparticle vaccine could stimulate the mouse organism to produce high levels of humoral and cellular immunity, making it a promising candidate for the development of subunit vaccines against swine fever and swine circulatory-associated diseases. This study also provides ideas for other disease vaccines.
Collapse
Affiliation(s)
- Pu Wang
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Gang Zhang
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Qiang Liu
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Lingling Jiang
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - XiaoXia Niu
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Min Fang
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Weifeng Gao
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Yulong He
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University
| | - Yong Li
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China.
| | - Jianhong Shu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University.
| | - Sinong Zhang
- School of Life Sciences, Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China.
| |
Collapse
|
4
|
Wang Z, Li M, Li W, He L, Wang L, Cai K, Zhao X, Chen Y, Li D. Hybrid Outer Membrane Vesicles with Genetically Engineering for Treatment of Implant-Associated Infections and Relapse Prevention Through Host Immunomodulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415379. [PMID: 39950746 PMCID: PMC11984893 DOI: 10.1002/advs.202415379] [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: 11/20/2024] [Revised: 02/01/2025] [Indexed: 04/12/2025]
Abstract
Implant-associated infections (IAIs) are refractory to elimination, and the local immunosuppressive microenvironment (IME) exacerbates therapeutic difficulties, ultimately causing persistence and relapse. Therefore, exploring immunostrengthening treatments holds great promise for reversing IME and thoroughly eradicating chronic or repetitive infections. Bacterial outer membrane vesicles (OMVs) have emerged as potential immunostimulatory candidates; however, they lack active targeting capabilities and cause non-specific inflammatory side effects. In this study, bone marrow-derived mesenchymal stem cells (BMSCs) are genetically engineered to overexpress CXCR4 and isolated cell membranes (mBMSCCXCR4) for hybridization with OMVs derived from Escherichia coli (E. coli) to produce nanovesicles (mBMSCCXCR4@OMV). The resulting mBMSCCXCR4@OMV nanovesicles demonstrate excellent bone marrow targeting capability and are effectively taken up by bone marrow-derived macrophages, triggering the efficient transition to pro-inflammatory M1 status through TLR/NF-κB pathway. This alteration promotes innate bactericidal capacity and antigen presentation. Subsequent activation of T and B cells and inhibition of myeloid-derived suppressor cells (MDSCs) facilitated in vivo adaptive immunity in mouse models. Additionally, mBMSCCXCR4@OMV boosted memory B cell and bacteria-specific antibody responses. Together, these data highlight the potential of mBMSCCXCR4@OMV to eradicate complicated IAIs and provide whole-stage protection against postsurgical relapse, thus marking a significant immunotherapeutic advancement in the post-antibiotic era.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Mingfei Li
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Medical 3D Printing CenterThe First Affiliated Hospital of Zhengzhou UniversityHenan Institute of Advanced Technology of Zhengzhou UniversityZhengzhou450052China
| | - Wenshuai Li
- Medical 3D Printing CenterThe First Affiliated Hospital of Zhengzhou UniversityHenan Institute of Advanced Technology of Zhengzhou UniversityZhengzhou450052China
| | - Liuliang He
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Long Wang
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Kehan Cai
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190China
| | - Yazhou Chen
- Medical 3D Printing CenterThe First Affiliated Hospital of Zhengzhou UniversityHenan Institute of Advanced Technology of Zhengzhou UniversityZhengzhou450052China
| | - Daifeng Li
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| |
Collapse
|
5
|
Alshamrani S, Mashraqi MM, Alzamami A, Alturki NA, Almasoudi HH, Ahmed I, Basharat Z. Leveraging core proteome data of Kingella kingae for multi-epitope vaccine design against TonB dependent receptor (TDR): an in silico approach. J Biomol Struct Dyn 2025:1-18. [PMID: 40105736 DOI: 10.1080/07391102.2025.2480263] [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: 09/19/2023] [Accepted: 04/29/2024] [Indexed: 03/20/2025]
Abstract
Kingella kingae is a Gram-negative bacterium that causes invasive infections in children and older or immunocompromised individuals, making it a significant public health concern. In this study, a pan-proteomic mediated vaccine target mining was attempted to identify potential vaccine targets in K. kingae. Currently, there is no vaccine available against this pathobiont. Therefore, we designed and validated an in silico vaccine construct by targeting the lactoferrin/transferrin-binding TonB-dependent receptor. Antigenic regions of the TonB receptor were mapped, and the predicted epitopes were anticipated to be effective in a broad range of the world population. Using their combinations with linkers and various adjuvants, 12 vaccine constructs were prepared. The best construct (C7) with no allergenicity and high antigenicity was subjected to molecular modeling, docking with important immune receptors of humans, and then molecular dynamics (MD) simulation. After binding validation and stability assessment, it was cloned into a pet-28a + plasmid vector. Immune response was also simulated, and the vaccine was observed to invoke B- and T-cell induction. These findings can help accelerate the development of a new vaccine against K. kingae or other pathogens targeting the homolog of TonB. Nevertheless, we propose additional testing of C7 construct for efficacy and safety in vitro and in vivo.
Collapse
Affiliation(s)
- Saleh Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Mutaib M Mashraqi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah, Saudi Arabia
| | - Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ibrar Ahmed
- Alpha Genomics Private Limited, Islamabad, Pakistan
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, Republic of Korea
| | | |
Collapse
|
6
|
Li Y, Xu Y, Su W, Xu J, Ye Z, Wang Z, Liu Q, Chen F. Exploring the immuno-nano nexus: A paradigm shift in tumor vaccines. Biomed Pharmacother 2025; 184:117897. [PMID: 39921945 DOI: 10.1016/j.biopha.2025.117897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Tumor vaccines have become a crucial strategy in cancer immunotherapy. Challenges of traditional tumor vaccines include inadequate immune activation and low efficacy of antigen delivery. Nanoparticles, with their tunable properties and versatile functionalities, have redefined the landscape of tumor vaccine design. In this review, we outline the multifaceted roles of nanoparticles in tumor vaccines, ranging from their capacity as delivery vehicles to their function as immunomodulatory adjuvants capable of stimulating anti-tumor immunity. We discuss how this innovative approach significantly boosts antigen presentation by leveraging tailored nanoparticles that facilitate efficient uptake by antigen-presenting cells. These nanoparticles have been meticulously designed to overcome biological barriers, ensuring optimal delivery to lymph nodes and effective interaction with the immune system. Overall, this review highlights the transformative power of nanotechnology in redefining the principles of tumor vaccines. The intent is to inform more efficacious and precise cancer immunotherapies. The integration of these advanced nanotechnological strategies should unlock new frontiers in tumor vaccine development, enhancing their potential to elicit robust and durable anti-tumor immunity.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yike Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenwen Su
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jia Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zifei Ye
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhuoyi Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qihui Liu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Fangfang Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
7
|
Romanov A, Knappe GA, Ronsard L, Suh H, Omer M, Chapman AP, Lewis VR, Spivakovsky K, Canales J, Reizis B, Tingle RD, Cottrell CA, Schiffner T, Lingwood D, Bathe M, Irvine DJ. DNA origami vaccines program antigen-focused germinal centers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639354. [PMID: 40060683 PMCID: PMC11888200 DOI: 10.1101/2025.02.21.639354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Recruitment and expansion of rare precursor B cells in germinal centers (GCs) is a central goal of vaccination to generate broadly neutralizing antibodies (bnAbs) against challenging pathogens such as HIV. Multivalent immunogen display is a well-established method to enhance vaccine-induced B cell responses, typically accomplished by using natural or engineered protein scaffolds. However, these scaffolds themselves are targets of antibody responses, with the potential to generate competitor scaffold-specific B cells that could theoretically limit expansion and maturation of "on-target" B cells in the GC response. Here, we rationally designed T-independent, DNA-origami based virus-like particles (VLPs) with optimal antigenic display of the germline targeting HIV Env immunogen, eOD-GT8, and appropriate T cell help to achieve a potent GC response. In preclinical mouse models, these DNA-VLPs expanded significantly higher frequencies of epitope-specific GC B cells compared with a state-of-the-art clinical protein nanoparticle. Optimized DNA-VLPs primed germinal centers focused on the target antigen and rapidly expanded subdominant broadly neutralizing antibody precursor B cells for HIV with a single immunization. Thus, avoiding scaffold-specific responses augments priming of bnAb precursor B cells, and DNA-VLPs are a promising platform for promoting B cell responses towards challenging subdominant epitopes.
Collapse
Affiliation(s)
- Anna Romanov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Grant A Knappe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Larance Ronsard
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Heikyung Suh
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, United States
| | - Marjan Omer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Asheley P Chapman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, United States
| | - Vanessa R Lewis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Katie Spivakovsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Josue Canales
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Ryan D Tingle
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, United States
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, United States
| | - Christopher A Cottrell
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, United States
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, United States
| | - Torben Schiffner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, United States
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, United States
| | - Daniel Lingwood
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Mark Bathe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, MA 02139 United States
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02115, United States
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, United States
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, 92037, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| |
Collapse
|
8
|
Kawaguchi Y, Shimizu T, Takata H, Ando H, Ishida T. An in vitro nanocarrier-based B cell antigen loading system; tumor growth suppression via transfusion of the antigen-loaded B cells in vivo. Int J Pharm 2025; 670:125189. [PMID: 39788395 DOI: 10.1016/j.ijpharm.2025.125189] [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: 09/18/2024] [Revised: 12/04/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
B cell-based vaccines are expected to provide an alternative to DC-based vaccines. However, the efficacy of antigen uptake by B cells in vitro is relatively low, and efficient antigen-loading methods must be established before B cell-based vaccines are viable in clinical settings. We recently developed an in vitro system that efficiently loads antigens into isolated splenic B cells via liposomes decorated with hydroxyl PEG (HO-PEG-Lips). Therefore, the purpose of this study was to expand this system in order to achieve another approach to in vivo tumor growth suppression. By using HO-PEG-Lips as a carrier for model antigen OVA along with an adjuvant, α-galactosylceramide (GC), the amount of antigen loading to the B cells in vitro was increased compared with that of both free OVA and free GC. Transfusion of B cells treated with HO-PEG-Lips that encapsulated OVA and GC suppressed the growth of OVA-expressing murine thymoma (E.G7-OVA) tumors in vivo through strong induction of OVA-specific T cells. Under fluorescence microscopic observation, migration of the transfused B cells in the spleens of recipient mice were confirmed. Our results indicate that our novel antigen-loading system could become a promising approach to facilitate the development of cell-based therapeutic cancer vaccines utilizing B cells as alternative APCs.
Collapse
Affiliation(s)
- Yoshino Kawaguchi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan.
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505 Tokushima, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505 Tokushima, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505 Tokushima, Japan.
| |
Collapse
|
9
|
Zhong Z, Quiñones-Pérez M, Dai Z, Juarez VM, Bhatia E, Carlson CR, Shah SB, Patel A, Fang Z, Hu T, Allam M, Hicks SL, Gupta M, Gupta SL, Weeks E, Vagelos SD, Molina A, Mulero-Russe A, Mora-Boza A, Joshi DJ, Sekaly RP, Sulchek T, Goudy SL, Wrammert J, Roy K, Boss JM, Coskun AF, Scharer CD, García AJ, Koff JL, Singh A. Human immune organoids to decode B cell response in healthy donors and patients with lymphoma. NATURE MATERIALS 2025; 24:297-311. [PMID: 39506098 PMCID: PMC11866935 DOI: 10.1038/s41563-024-02037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Antibodies are produced when naive B cells differentiate into plasma cells within germinal centres (GCs) of lymphoid tissues. Patients with B cell lymphoma on effective immunotherapies exhibit diminished antibody production, leading to higher infection rates and reduced vaccine efficacy, even after B cell recovery. Current ex vivo models fail to sustain long-term GC reactions and effectively test B cell responses. Here we developed synthetic hydrogels mimicking the lymphoid tissue microenvironment, enabling human GCs from tonsils and peripheral blood mononuclear cell-derived B cells. Immune organoids derived from peripheral blood mononuclear cells maintain GC B cells and plasma cells longer than tonsil-derived ones and exhibit unique B cell programming, including GC compartments, somatic hypermutation, immunoglobulin class switching and B cell clones. Chemical inhibition of transcriptional and epigenetic processes enhances plasma cell formation. While integrating polarized CXCL12 protein in a lymphoid organ-on-chip modulates GC responses in healthy donor B cells, it fails with B cells derived from patients with lymphoma. Our system allows rapid, controlled modelling of immune responses and B cell disorders.
Collapse
Affiliation(s)
- Zhe Zhong
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Manuel Quiñones-Pérez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhonghao Dai
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Valeria M Juarez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Eshant Bhatia
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christopher R Carlson
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shivem B Shah
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Anjali Patel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Thomas Hu
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mayar Allam
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sakeenah L Hicks
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Mansi Gupta
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Sneh Lata Gupta
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ethan Weeks
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Stephanie D Vagelos
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Alejandro Molina
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Adriana Mulero-Russe
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Devyani J Joshi
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Rafick P Sekaly
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Todd Sulchek
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Steven L Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Otolaryngology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Jens Wrammert
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Krishnendu Roy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jean L Koff
- Winship Cancer Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ankur Singh
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
10
|
Lin W, Shen C, Li M, Ma S, Liu C, Huang J, Ren Z, Yang Y, Zhao M, Xie Q, Guo S, Wang W, Wang K, Ma Q, Jiang Y, Zheng J, Liao Y. Programmable Macrophage Vesicle Based Bionic Self-Adjuvanting Vaccine for Immunization against Monkeypox Virus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408608. [PMID: 39513669 PMCID: PMC11714231 DOI: 10.1002/advs.202408608] [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: 07/25/2024] [Revised: 11/01/2024] [Indexed: 11/15/2024]
Abstract
The emergence of monkeypox has become a global health threat after the COVID-19 pandemic. Due to the lack of available specifically treatment against MPV, developing an available vaccine is thus the most prospective and urgent strategy. Herein, a programmable macrophage vesicle based bionic self-adjuvanting vaccine (AM@AEvs-PB) is first developed for defending against monkeypox virus (MPV). Based on MPV-related antigen-stimulated macrophage-derived vesicles, the nanovaccine is constructed by loading the mature virion (MV)-related intracellular protein (A29L/M1R) and simultaneously modifying with the enveloped virion (EV) antigen (B6R), enabling them to effectively promote antigen presentation and enhance adaptive immune through self-adjuvant strategy. Owing to the synergistic properties of bionic vaccine coloaded MV and EV protein in defensing MPV, the activation ratio of antigen-presenting cells is nearly four times than that of single antigen in the same dose, resulting in stronger immunity in host. Notably, intramuscular injection uptake of AM@AEvs-PB demonstrated vigorous immune-protective effects in the mouse challenge attempt, offering a promising strategy for pre-clinical monkeypox vaccine development.
Collapse
Affiliation(s)
- Weiqiang Lin
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
- Institute for Engineering MedicineKunming Medical UniversityKunming650500P. R. China
| | - Chenguang Shen
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Mengjun Li
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Shengchao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Chenxin Liu
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhou510515P. R. China
| | - Jialin Huang
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Zuning Ren
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Yuechao Yang
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Minghai Zhao
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Qiulin Xie
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Shuang Guo
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Wei Wang
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Kaiyuan Wang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingapore119074Singapore
| | - Qiang Ma
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhou510515P. R. China
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Judun Zheng
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Yuhui Liao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
- Institute for Engineering MedicineKunming Medical UniversityKunming650500P. R. China
| |
Collapse
|
11
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [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] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
12
|
Yuan Y, Li H, Song Y, Zhang D, Wang Z, Yi X, Qi B, Zhang X, Jiang P, Yu A. Drug-Free "Triboelectric Immunotherapy" Activating Immunity for Osteomyelitis Treatment and Recurrence Prevention. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408473. [PMID: 39212208 DOI: 10.1002/adma.202408473] [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: 06/14/2024] [Revised: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Treatment of osteomyelitis is clinically challenging with low therapeutic efficacy and high risk of recurrence owing to the immunosuppressive microenvironment. Existing therapies are limited by drug concentration and single regulatory effect on the immune network, and emphasize the role of anti-inflammatory effects in reducing osteoclast rather than the role of proinflammatory effects in accelerating infection clearance, which is not conducive to complete bacteria elimination and recurrence prevention. Herein, a direct-current triboelectric nanogenerator (DC-TENG) is established to perform antibacterial effects and modulate immunological properties of infectious microenvironments of osteomyelitis through electrical stimulation, namely triboelectric immunotherapy. Seeing from the results, the triboelectric immunotherapy successfully activates polarization to proinflammatory (M1) macrophages in vitro, accompanied by satisfying direct antibacterial effects. The antibacterial and osteogenic abilities of triboelectric immunotherapy are verified in rat cranial osteomyelitis models. The effects on the polarization and differentiation of immune-related cells in vivo are investigated by establishing in situ tibial osteomyelitis models and immunosurveillance models in C57 mice respectively, indicating the ability of activating immunity and producing immunological memory for in situ infection and secondary recurrence, thus accelerating healing and preventing relapse. This study provides an efficient, long-acting, multifunctional, and wearable triboelectric immunotherapy strategy for drug-free osteomyelitis treatment systems.
Collapse
Affiliation(s)
- Ying Yuan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Haimei Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, P. R. China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan, 430072, China
| | - Yuchen Song
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Dong Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinzeyu Yi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Baiwen Qi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xianzheng Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Peng Jiang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, P. R. China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan, 430072, China
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
13
|
Chaudhary N, Kasiewicz LN, Newby AN, Arral ML, Yerneni SS, Melamed JR, LoPresti ST, Fein KC, Strelkova Petersen DM, Kumar S, Purwar R, Whitehead KA. Amine headgroups in ionizable lipids drive immune responses to lipid nanoparticles by binding to the receptors TLR4 and CD1d. Nat Biomed Eng 2024; 8:1483-1498. [PMID: 39363106 PMCID: PMC11863198 DOI: 10.1038/s41551-024-01256-w] [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: 11/04/2022] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Lipid nanoparticles (LNPs) are the most clinically advanced delivery vehicle for RNA therapeutics, partly because of established lipid structure-activity relationships focused on formulation potency. Yet such knowledge has not extended to LNP immunogenicity. Here we show that the innate and adaptive immune responses elicited by LNPs are linked to their ionizable lipid chemistry. Specifically, we show that the amine headgroups in ionizable lipids drive LNP immunogenicity by binding to Toll-like receptor 4 and CD1d and by promoting lipid-raft formation. Immunogenic LNPs favour a type-1 T-helper-cell-biased immune response marked by increases in the immunoglobulins IgG2c and IgG1 and in the pro-inflammatory cytokines tumour necrosis factor, interferon γ and the interleukins IL-6 and IL-2. Notably, the inflammatory signals originating from these receptors inhibit the production of anti-poly(ethylene glycol) IgM antibodies, preventing the often-observed loss of efficacy in the LNP-mediated delivery of siRNA and mRNA. Moreover, we identified computational methods for the prediction of the structure-dependent innate and adaptive responses of LNPs. Our findings may help accelerate the discovery of well-tolerated ionizable lipids suitable for repeated dosing.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lisa N Kasiewicz
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alexandra N Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Mariah L Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Jilian R Melamed
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Samuel T LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Katherine C Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Sushant Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rahul Purwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Carmona-Ribeiro AM, Pérez-Betancourt Y. Emerging Cationic Nanovaccines. Pharmaceutics 2024; 16:1362. [PMID: 39598488 PMCID: PMC11597065 DOI: 10.3390/pharmaceutics16111362] [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: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Cationic vaccines of nanometric sizes can directly perform the delivery of antigen(s) and immunomodulator(s) to dendritic cells in the lymph nodes. The positively charged nanovaccines are taken up by antigen-presenting cells (APCs) of the lymphatic system often originating the cellular immunological defense required to fight intracellular microbial infections and the proliferation of cancers. Cationic molecules imparting the positive charges to nanovaccines exhibit a dose-dependent toxicity which needs to be systematically addressed. Against the coronavirus, mRNA cationic nanovaccines evolved rapidly. Nowadays cationic nanovaccines have been formulated against several infections with the advantage of cationic compounds granting protection of nucleic acids in vivo against biodegradation by nucleases. Up to the threshold concentration of cationic molecules for nanovaccine delivery, cationic nanovaccines perform well eliciting the desired Th 1 improved immune response in the absence of cytotoxicity. A second strategy in the literature involves dilution of cationic components in biocompatible polymeric matrixes. Polymeric nanoparticles incorporating cationic molecules at reduced concentrations for the cationic component often result in an absence of toxic effects. The progress in vaccinology against cancer involves in situ designs for cationic nanovaccines. The lysis of transformed cancer cells releases several tumoral antigens, which in the presence of cationic nanoadjuvants can be systemically presented for the prevention of metastatic cancer. In addition, these local cationic nanovaccines allow immunotherapeutic tumor treatment.
Collapse
Affiliation(s)
- Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Yunys Pérez-Betancourt
- Department of Microbiology, University of Chicago, Cummings Life Science Center 920 E 58th St., Chicago, IL 60637, USA;
| |
Collapse
|
15
|
Han J, Sheng T, Zhang Y, Cheng H, Gao J, Yu J, Gu Z. Bioresponsive Immunotherapeutic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2209778. [PMID: 36639983 DOI: 10.1002/adma.202209778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The human immune system is an interaction network of biological processes, and its dysfunction is closely associated with a wide array of diseases, such as cancer, infectious diseases, tissue damage, and autoimmune diseases. Manipulation of the immune response network in a desired and controlled fashion has been regarded as a promising strategy for maximizing immunotherapeutic efficacy and minimizing side effects. Integration of "smart" bioresponsive materials with immunoactive agents including small molecules, biomacromolecules, and cells can achieve on-demand release of agents at targeted sites to reduce overdose-related toxicity and alleviate off-target effects. This review highlights the design principles of bioresponsive immunotherapeutic materials and discusses the critical roles of controlled release of immunoactive agents from bioresponsive materials in recruiting, housing, and manipulating immune cells for evoking desired immune responses. Challenges and future directions from the perspective of clinical translation are also discussed.
Collapse
Affiliation(s)
- Jinpeng Han
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tao Sheng
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Jianqing Gao
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
16
|
Myers ML, Gallagher JR, Woolfork DD, Khorrami ND, Park WB, Maldonado-Puga S, Bohrnsen E, Schwarz BH, Alves DA, Bock KW, Dearborn AD, Harris AK. Structure-guided assembly of an influenza spike nanobicelle vaccine provides pan H1 intranasal protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613335. [PMID: 39372767 PMCID: PMC11451756 DOI: 10.1101/2024.09.16.613335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Development of intranasal vaccines for respiratory viruses has gained popularity. However, currently only a live-attenuated influenza vaccine is FDA-approved for intranasal administration. Here, we focused on influenza virus as it circulates seasonally, has pandemic potential, and has vaccine formulations that present hemagglutinin (HA) in different structural arrangements. These display differences have not been correlated with induction of pan-H1 antibodies or shown to provide intranasal protection. Using electron microscopy, biochemistry and animal studies, we identified HA complexes arranged as lipid discs with multiple trimeric HAs displayed along the perimeter, termed spike nanobicelles (SNB). We utilized a structure-guided approach to synthesize in vitro assembled spiked nanobicelles (IA-SNB) from a classical 1934 H1N1 influenza virus. IA-SNBs elicited pan-H1 antibodies and provided protection against antigenically divergent H1N1 viruses via intranasal immunizations. Viral glycoprotein spikes displayed as SNBs could aid in combating antigenic variation and provide innovative intranasal vaccines to aid universal influenza vaccine development.
Collapse
Affiliation(s)
- Mallory L. Myers
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - John R. Gallagher
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - De’Marcus D. Woolfork
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Noah D. Khorrami
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - William B. Park
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Samantha Maldonado-Puga
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| | - Eric Bohrnsen
- Protein Chemistry Section, Research and Technologies Branch, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, USA 59840
| | - Benjamin H. Schwarz
- Protein Chemistry Section, Research and Technologies Branch, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, USA 59840
| | - Derron A. Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, USA 20892
| | - Kevin W. Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, USA 20892
| | - Altaira D. Dearborn
- Structural Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6531, Bethesda, MD, USA 20892
| | - Audray K. Harris
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, USA 20892
| |
Collapse
|
17
|
Rahman S, Chiou CC, Almutairi MM, Ajmal A, Batool S, Javed B, Tanaka T, Chen CC, Alouffi A, Ali A. Targeting Yezo Virus Structural Proteins for Multi-Epitope Vaccine Design Using Immunoinformatics Approach. Viruses 2024; 16:1408. [PMID: 39339884 PMCID: PMC11437474 DOI: 10.3390/v16091408] [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: 07/27/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
A novel tick-borne orthonairovirus called the Yezo virus (YEZV), primarily transmitted by the Ixodes persulcatus tick, has been recently discovered and poses significant threats to human health. The YEZV is considered endemic in Japan and China. Clinical symptoms associated with this virus include thrombocytopenia, fatigue, headache, leukopenia, fever, depression, and neurological complications ranging from mild febrile illness to severe outcomes like meningitis and encephalitis. At present, there is no treatment or vaccine readily accessible for this pathogenic virus. Therefore, this research employed an immunoinformatics approach to pinpoint potential vaccine targets within the YEZV through an extensive examination of its structural proteins. Three structural proteins were chosen using specific criteria to pinpoint T-cell and B-cell epitopes, which were subsequently validated through interferon-gamma induction. Six overlapping epitopes for cytotoxic T-lymphocytes (CTL), helper T-lymphocytes (HTL), and linear B-lymphocytes (LBL) were selected to construct a multi-epitope vaccine, achieving a 92.29% coverage of the global population. These epitopes were then fused with the 50S ribosomal protein L7/L12 adjuvant to improve protection against international strains. The three-dimensional structure of the designed vaccine construct underwent an extensive evaluation through structural analysis. Following molecular docking studies, the YEZV vaccine construct emerged as a candidate for further investigation, showing the lowest binding energy (-78.7 kcal/mol) along with favorable physiochemical and immunological properties. Immune simulation and molecular dynamics studies demonstrated its stability and potential to induce a strong immune response within the host cells. This comprehensive analysis indicates that the designed vaccine construct could offer protection against the YEZV. It is crucial to conduct additional in vitro and in vivo experiments to verify its safety and effectiveness.
Collapse
Affiliation(s)
- Sudais Rahman
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Chien-Chun Chiou
- Department of Dermatology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Mashal M Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Amar Ajmal
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Sidra Batool
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Bushra Javed
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Tetsuya Tanaka
- Laboratory of Animal Microbiology, Graduate School of Agricultural Science/Faculty of Agriculture, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
18
|
Hu Y, Yang A, Li H, Zhao R, Bao C, Yu Y, Wang Y, Wang Z, Zhuo L, Han Q, Zhang Z, Zhang J, Zhao H. Lymph node-targeted STING agonist nanovaccine against chronic HBV infection. Cell Mol Life Sci 2024; 81:372. [PMID: 39196331 PMCID: PMC11358573 DOI: 10.1007/s00018-024-05404-y] [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: 02/20/2024] [Revised: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
Chronic hepatitis B virus (HBV) infection is a global health problem that substantially increases the risk of developing liver disease. The development of a novel strategy to induce anti-HB seroconversion and achieve a long-lasting immune response against chronic HBV infection remains challenging. Here, we found that chronic HBV infection affected the signaling pathway involved in STING-mediated induction of host immune responses in dendritic cells (DCs) and then generated a lymph node-targeted nanovaccine that co-delivered hepatitis B surface antigen (HBsAg) and cyclic diguanylate monophosphate (c-di-GMP) (named the PP-SG nanovaccine). The feasibility and efficiency of the PP-SG nanovaccine for CHB treatment were evaluated in HBV-carrier mice. Serum samples were analyzed for HBsAg, anti-HBs, HBV DNA, and alanine aminotransferase levels, and liver samples were evaluated for HBV DNA and RNA and HBcAg, accompanied by an analysis of HBV-specific cellular and humoral immune responses during PP-SG nanovaccine treatment. The PP-SG nanovaccine increased antigen phagocytosis and DC maturation, efficiently and safely eliminated HBV, achieved a long-lasting immune response against HBV reinjection, and disrupted chronic HBV infection-induced immune tolerance, as characterized by the generation and multifunctionality of HBV-specific CD8+ T and CD4+ T cells and the downregulation of immune checkpoint molecules. HBV-carrier mice immunized with the PP-SG nanovaccine achieved partial anti-HBs seroconversion. The PP-SG nanovaccine can induce sufficient and persistent viral suppression and achieve anti-HBs seroconversion, rendering it a promising vaccine candidate for clinical chronic hepatitis B therapy.
Collapse
Affiliation(s)
- Yifei Hu
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ailu Yang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong, China
| | - Cuiping Bao
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yating Yu
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yucan Wang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zixuan Wang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Li Zhuo
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiyue Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
19
|
Allam M, Hu T, Fang Z, Pi M, Singh A, Coskun AF. Spatial immunophenotyping using multiplexed imaging of immune follicles in secondary lymphoid tissues. PNAS NEXUS 2024; 3:pgae285. [PMID: 39108301 PMCID: PMC11299982 DOI: 10.1093/pnasnexus/pgae285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Secondary lymphoid organs (SLOs), including tonsils (TS), lymph nodes (LN), and Peyer's Patches, exhibit complementary immune functions. However, little is known about the spatial organization of immune cells and extracellular matrix (ECM) in the SLOs. Traditional imaging is limited to a few markers, confining our understanding of the differences between the SLOs. Herein, imaging mass cytometry addressed this gap by simultaneously profiling 25-plex proteins in SLO tissues at subcellular resolution. The antibody panel targeted immune, stromal, chemokine, epigenetic, and functional markers. For robust cell identification, a computational workflow SpatialVizPheno was developed to spatially phenotype 999,970 cells using two approaches, including manual gating and semi-supervised gating, iterative clustering, and annotation. LN exhibited the highest density of B cells while the intestinal tissues contained the highest proportion of regulatory and follicular helper T cells. SpatialVizPheno identified the most prevalent interaction between follicular dendritic cells and stromal cells (SCs), plasmablasts/plasma cells, and the SCs across the lymphoid tissues. Collagen-enriched regions were associated with the spatial orientation of B cell follicles in both TS and LN tissues, but not in intestinal lymphoid tissues. Such spatial differences of immunophenotypes and ECM in different SLO tissues can be used to quantify the relationship between cellular organization and ultimate immune responses.
Collapse
Affiliation(s)
- Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Michelle Pi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ankur Singh
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
20
|
Guo Z, Zhu AT, Wei X, Jiang Y, Yu Y, Noh I, Gao W, Fang RH, Zhang L. A genetically engineered neuronal membrane-based nanotoxoid elicits protective immunity against neurotoxins. Bioact Mater 2024; 38:321-330. [PMID: 38764446 PMCID: PMC11101676 DOI: 10.1016/j.bioactmat.2024.05.006] [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: 01/07/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Given their dangerous effects on the nervous system, neurotoxins represent a significant threat to public health. Various therapeutic approaches, including chelating agents, receptor decoys, and toxin-neutralizing antibodies, have been explored. While prophylactic vaccines are desirable, it is oftentimes difficult to effectively balance their safety and efficacy given the highly dangerous nature of neurotoxins. To address this, we report here on a nanovaccine against neurotoxins that leverages the detoxifying properties of cell membrane-coated nanoparticles. A genetically modified cell line with constitutive overexpression of the α7 nicotinic acetylcholine receptor is developed as a membrane source to generate biomimetic nanoparticles that can effectively and irreversibly bind to α-bungarotoxin, a model neurotoxin. This abrogates the biological activity of the toxin, enabling the resulting nanotoxoid to be safely delivered into the body and processed by the immune system. When co-administered with an immunological adjuvant, a strong humoral response against α-bungarotoxin is generated that protects vaccinated mice against a lethal dose of the toxin. Overall, this work highlights the potential of using genetic modification strategies to develop nanotoxoid formulations against various biological threats.
Collapse
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Audrey T. Zhu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xiaoli Wei
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yiyan Yu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ilkoo Noh
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
21
|
Fan CY, Wang SW, Chung C, Chen JY, Chang CY, Chen YC, Hsu TL, Cheng TJR, Wong CH. Synthesis of a dendritic cell-targeted self-assembled polymeric nanoparticle for selective delivery of mRNA vaccines to elicit enhanced immune responses. Chem Sci 2024; 15:11626-11632. [PMID: 39055027 PMCID: PMC11268467 DOI: 10.1039/d3sc06575h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/23/2024] [Indexed: 07/27/2024] Open
Abstract
Recent development of SARS-CoV-2 spike mRNA vaccines to control the pandemic is a breakthrough in the field of vaccine development. mRNA vaccines are generally formulated with lipid nanoparticles (LNPs) which are composed of several lipids with specific ratios; however, they generally lack selective delivery. To develop a selective delivery method for mRNA vaccine formulation, we reported here the synthesis of polymeric nanoparticles (PNPs) composed of a guanidine copolymer containing zwitterionic groups and a dendritic cell (DC)-targeted aryl-trimannoside ligand for encapsulation and selective delivery of an mRNA to dendritic cells. A DC-targeted SARS-CoV-2 spike mRNA-PNP vaccine was shown to elicit a stronger protective immune response in mice compared to the traditional mRNA-LNP vaccine and those without the selective delivery design. It is anticipated that this technology is generally applicable to other mRNA vaccines for DC-targeted delivery with enhanced immune response.
Collapse
Affiliation(s)
- Chen-Yo Fan
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Szu-Wen Wang
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Cinya Chung
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Jia-Yan Chen
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Chia-Yen Chang
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Yu-Chen Chen
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Tsui-Ling Hsu
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | | | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
- Department of Chemistry, The Scripps Research Institute La Jolla California 92037 USA
| |
Collapse
|
22
|
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
|
23
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
24
|
Wang X, Cottrell CA, Hu X, Ray R, Bottermann M, Villavicencio PM, Yan Y, Xie Z, Warner JE, Ellis-Pugh JR, Kalyuzhniy O, Liguori A, Willis JR, Menis S, Rämisch S, Eskandarzadeh S, Kubitz M, Tingle R, Phelps N, Groschel B, Himansu S, Carfi A, Kirsch KH, Weldon SR, Nair U, Schief WR, Batista FD. mRNA-LNP prime boost evolves precursors toward VRC01-like broadly neutralizing antibodies in preclinical humanized mouse models. Sci Immunol 2024; 9:eadn0622. [PMID: 38753808 PMCID: PMC11488661 DOI: 10.1126/sciimmunol.adn0622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Germline-targeting (GT) protein immunogens to induce VRC01-class broadly neutralizing antibodies (bnAbs) to the CD4-binding site of the HIV envelope (Env) have shown promise in clinical trials. Here, we preclinically validated a lipid nanoparticle-encapsulated nucleoside mRNA (mRNA-LNP) encoding eOD-GT8 60mer as a soluble self-assembling nanoparticle in mouse models. In a model with three humanized B cell lineages bearing distinct VRC01-precursor B cell receptors (BCRs) with similar affinities for eOD-GT8, all lineages could be simultaneously primed and undergo diversification and affinity maturation without exclusionary competition. Boosts drove precursor B cell participation in germinal centers; the accumulation of somatic hypermutations, including in key VRC01-class positions; and affinity maturation to boost and native-like antigens in two of the three precursor lineages. We have preclinically validated a prime-boost regimen of soluble self-assembling nanoparticles encoded by mRNA-LNP, demonstrating that multiple lineages can be primed, boosted, and diversified along the bnAb pathway.
Collapse
Affiliation(s)
- Xuesong Wang
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Christopher A. Cottrell
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Xiaozhen Hu
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
- Moderna Inc.; Cambridge, 02139, USA
| | - Rashmi Ray
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Maria Bottermann
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | | | - Yu Yan
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Zhenfei Xie
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - John E. Warner
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | | | - Oleksandr Kalyuzhniy
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Alessia Liguori
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Jordan R. Willis
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Sergey Menis
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Sebastian Rämisch
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Saman Eskandarzadeh
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Michael Kubitz
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Ryan Tingle
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Nicole Phelps
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Bettina Groschel
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | | | | | - Kathrin H. Kirsch
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Stephanie R. Weldon
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Usha Nair
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - William R. Schief
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
- Moderna Inc.; Cambridge, 02139, USA
| | - Facundo D. Batista
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
- Department of Biology, Massachusetts Institute of Technology; Cambridge, 02139, USA
| |
Collapse
|
25
|
Li C, Clauson R, Bugada LF, Ke F, He B, Yu Z, Chen H, Jacobovitz B, Hu H, Chuikov P, Hill BD, Rizvi SM, Song Y, Sun K, Axenov P, Huynh D, Wang X, Garmire L, Lei YL, Grigorova I, Wen F, Cascalho M, Gao W, Sun D. Antigen-Clustered Nanovaccine Achieves Long-Term Tumor Remission by Promoting B/CD 4 T Cell Crosstalk. ACS NANO 2024; 18:9584-9604. [PMID: 38513119 PMCID: PMC11130742 DOI: 10.1021/acsnano.3c13038] [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: 03/23/2024]
Abstract
Current cancer vaccines using T cell epitopes activate antitumor T cell immunity through dendritic cell/macrophage-mediated antigen presentation, but they lack the ability to promote B/CD4 T cell crosstalk, limiting their anticancer efficacy. We developed antigen-clustered nanovaccine (ACNVax) to achieve long-term tumor remission by promoting B/CD4 T cell crosstalk. The topographic features of ACNVax were achieved using an iron nanoparticle core attached with an optimal number of gold nanoparticles, where the clusters of HER2 B/CD4 T cell epitopes were conjugated on the gold surface with an optimal intercluster distance of 5-10 nm. ACNVax effectively trafficked to lymph nodes and cross-linked with BCR, which are essential for stimulating B cell antigen presentation-mediated B/CD4 T cell crosstalk in vitro and in vivo. ACNVax, combined with anti-PD-1, achieved long-term tumor remission (>200 days) with 80% complete response in mice with HER2+ breast cancer. ACNVax not only remodeled the tumor immune microenvironment but also induced a long-term immune memory, as evidenced by complete rejection of tumor rechallenge and a high level of antigen-specific memory B, CD4, and CD8 cells in mice (>200 days). This study provides a cancer vaccine design strategy, using B/CD4 T cell epitopes in an antigen clustered topography, to achieve long-term durable anticancer efficacy through promoting B/CD4 T cell crosstalk.
Collapse
Affiliation(s)
- Chengyi Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ryan Clauson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Luke F Bugada
- Department of Chemical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fang Ke
- Department of Microbiology and Immunology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bing He
- Department of Computational Medicine & Bioinformatics, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhixin Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hongwei Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Binyamin Jacobovitz
- Microscopy Core, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hongxiang Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Polina Chuikov
- Department of Microbiology and Immunology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brett Dallas Hill
- Department of Chemical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Syed M Rizvi
- Department of Chemical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yudong Song
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kai Sun
- Department of Materials Science and Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Pasieka Axenov
- Department of Microbiology and Immunology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Daniel Huynh
- Department of Microbiology and Immunology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xinyi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lana Garmire
- Department of Computational Medicine & Bioinformatics, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Leo Lei
- Departments of Head and Neck Surgery, Cancer Biology, and Translational Molecular Pathology, the University of Texas M.D. Anderson Cancer Center, Houston, Texas 77054, United States
| | - Irina Grigorova
- Department of Microbiology and Immunology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Marilia Cascalho
- Department of Microbiology and Immunology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Wei Gao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
26
|
Wang J, Zhu H, Gan J, Liang G, Li L, Zhao Y. Engineered mRNA Delivery Systems for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308029. [PMID: 37805865 DOI: 10.1002/adma.202308029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/05/2023] [Indexed: 10/09/2023]
Abstract
Messenger RNA (mRNA)-based therapeutic strategies have shown remarkable promise in preventing and treating a staggering range of diseases. Optimizing the structure and delivery system of engineered mRNA has greatly improved its stability, immunogenicity, and protein expression levels, which has led to a wider range of uses for mRNA therapeutics. Herein, a thorough analysis of the optimization strategies used in the structure of mRNA is first provided and delivery systems are described in great detail. Furthermore, the latest advancements in biomedical engineering for mRNA technology, including its applications in combatting infectious diseases, treating cancer, providing protein replacement therapy, conducting gene editing, and more, are summarized. Lastly, a perspective on forthcoming challenges and prospects concerning the advancement of mRNA therapeutics is offered. Despite these challenges, mRNA-based therapeutics remain promising, with the potential to revolutionize disease treatment and contribute to significant advancements in the biomedical field.
Collapse
Affiliation(s)
- Ji Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Haofang Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingjing Gan
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Gaofeng Liang
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, 450009, China
| |
Collapse
|
27
|
Sun L, Zhang J, Shi L, Peng Y, Feng X, Huang F, Yang F, Li J, Wang S, Niu J, Liu J, Li Y, Li S, Chen Z. Development and Immunological Evaluation of a Multiantigen Thermostable Nanovaccine Adjuvanted with T-Cell-Activating Scaffold for African Swine Fever. ACS APPLIED BIO MATERIALS 2024; 7:1547-1557. [PMID: 38346262 DOI: 10.1021/acsabm.3c01035] [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: 03/19/2024]
Abstract
African swine fever is an acute and highly contagious infectious disease with a mortality rate of up to 100%. The lack of commercial vaccines and drugs is a serious economic threat to the global pig industry. Cell-mediated immunity plays an essential role in protection against viral infection. We previously reported the rational design of a T-cell-activating thermostable scaffold (RPT) for antigen delivery and improved cellular immunity. We conjugated antigens P30, P54, P72, CD2 V, and CP312R to RPT, using a SpyCatcher/SpyTag covalent attachment strategy to construct nanovaccines (multiantigens-RPT). Multiantigens-RPT exhibited significantly higher thermal, storage, and freeze-thaw stability. The specific antibodies IgG and IgG2a of the multiantigen-RPT-immunized were higher than the antigens cocktail-immunized by approximately 10-100 times. ELISpot demonstrated that more IFN-γ-secreting cells were produced by the multiantigen-RPT-immunized than by the antigens cocktail-immunized. Delivery of the multiantigen nanovaccine by a T-cell-activating scaffold induced strong humoral and cellular immune responses in mice and pigs and is a potentially useful candidate vaccine for the African swine fever virus.
Collapse
Affiliation(s)
- Lidan Sun
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
- Beijing Tonghe Litai Biotechnology Co., Ltd. Beijing 100080, China
| | - Jinsong Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lijun Shi
- Beijing Tonghe Litai Biotechnology Co., Ltd. Beijing 100080, China
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yuanli Peng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiangning Feng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Feixia Yang
- Beijing Tonghe Litai Biotechnology Co., Ltd. Beijing 100080, China
| | - Jinyu Li
- Beijing Tonghe Litai Biotechnology Co., Ltd. Beijing 100080, China
| | - Shuo Wang
- Beijing Tonghe Litai Biotechnology Co., Ltd. Beijing 100080, China
| | - Jingqi Niu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
| | - Jinling Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
| | - Yingjun Li
- Beijing Tonghe Litai Biotechnology Co., Ltd. Beijing 100080, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao 028000, China
| |
Collapse
|
28
|
Huang Z, Zhuang X, Liu L, Zhao J, Ma S, Si X, Zhu Z, Wu F, Jin N, Tian M, Song W, Chen X. Modularized viromimetic polymer nanoparticle vaccines (VPNVaxs) to elicit durable and effective humoral immune responses. Natl Sci Rev 2024; 11:nwad310. [PMID: 38312378 PMCID: PMC10833449 DOI: 10.1093/nsr/nwad310] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/27/2023] [Accepted: 11/23/2023] [Indexed: 02/06/2024] Open
Abstract
Virus-like particle (VLP) vaccines had shown great potential during the COVID-19 pandemic, and was thought to be the next generation of antiviral vaccine technology due to viromimetic structures. However, the time-consuming and complicated processes in establishing a current recombinant-protein-based VLP vaccine has limited its quick launch to the out-bursting pandemic. To simplify and optimize VLP vaccine design, we herein report a kind of viromimetic polymer nanoparticle vaccine (VPNVax), with subunit receptor-binding domain (RBD) proteins conjugated to the surface of polyethylene glycol-b-polylactic acid (PEG-b-PLA) nanoparticles for vaccination against SARS-CoV-2. The preparation of VPNVax based on synthetic polymer particle and chemical post-conjugation makes it possible to rapidly replace the antigens and construct matched vaccines at the emergence of different viruses. Using this modular preparation system, we identified that VPNVax with surface protein coverage of 20%-25% had the best immunostimulatory activity, which could keep high levels of specific antibody titers over 5 months and induce virus neutralizing activity when combined with an aluminum adjuvant. Moreover, the polymer nano-vectors could be armed with more immune-adjuvant functions by loading immunostimulant agents or chemical chirality design. This VPNVax platform provides a novel kind of rapidly producing and efficient vaccine against different variants of SARS-CoV-2 as well as other viral pandemics.
Collapse
Affiliation(s)
- Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xinyu Zhuang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Liping Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Zhenyi Zhu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Fan Wu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Ningyi Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Mingyao Tian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| |
Collapse
|
29
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
30
|
Wamhoff EC, Ronsard L, Feldman J, Knappe GA, Hauser BM, Romanov A, Case JB, Sanapala S, Lam EC, Denis KJS, Boucau J, Barczak AK, Balazs AB, Diamond MS, Schmidt AG, Lingwood D, Bathe M. Enhancing antibody responses by multivalent antigen display on thymus-independent DNA origami scaffolds. Nat Commun 2024; 15:795. [PMID: 38291019 PMCID: PMC10828404 DOI: 10.1038/s41467-024-44869-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Protein-based virus-like particles (P-VLPs) are commonly used to spatially organize antigens and enhance humoral immunity through multivalent antigen display. However, P-VLPs are thymus-dependent antigens that are themselves immunogenic and can induce B cell responses that may neutralize the platform. Here, we investigate thymus-independent DNA origami as an alternative material for multivalent antigen display using the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, the primary target of neutralizing antibody responses. Sequential immunization of mice with DNA-based VLPs (DNA-VLPs) elicits protective neutralizing antibodies to SARS-CoV-2 in a manner that depends on the valency of the antigen displayed and on T cell help. Importantly, the immune sera do not contain boosted, class-switched antibodies against the DNA scaffold, in contrast to P-VLPs that elicit strong B cell memory against both the target antigen and the scaffold. Thus, DNA-VLPs enhance target antigen immunogenicity without generating scaffold-directed immunity and thereby offer an important alternative material for particulate vaccine design.
Collapse
Affiliation(s)
- Eike-Christian Wamhoff
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Larance Ronsard
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Jared Feldman
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Grant A Knappe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Blake M Hauser
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Anna Romanov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shilpa Sanapala
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Evan C Lam
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Kerri J St Denis
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Julie Boucau
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Amy K Barczak
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Alejandro B Balazs
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aaron G Schmidt
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Daniel Lingwood
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA.
| | - Mark Bathe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA.
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Mir S, Mir M. The mRNA vaccine, a swift warhead against a moving infectious disease target. Expert Rev Vaccines 2024; 23:336-348. [PMID: 38369742 DOI: 10.1080/14760584.2024.2320327] [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: 09/11/2023] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
INTRODUCTION The rapid development of mRNA vaccines against SARS-CoV-2 has revolutionized vaccinology, offering hope for swift responses to emerging infectious diseases. Initially met with skepticism, mRNA vaccines have proven effective and safe, reducing vaccine hesitancy amid the evolving COVID-19 pandemic. The COVID-19 pandemic has demonstrated that the time required to modify mRNA vaccines to counter new mutant strains is significantly shorter than the time it takes for pathogens to mutate and generate new variants that can thrive in vaccinated populations. This highlights the notion that mRNA vaccine technology appears to be outpacing viruses in the ongoing evolutionary race. AREAS COVERED This review article offers valuable insights into several crucial aspects of mRNA vaccine development and deployment, including the fundamentals of mRNA vaccine design and synthesis, the utilization of delivery systems, considerations regarding vaccine safety, the longevity of the immune response, strategies for modifying the original mRNA vaccine to address emerging mutant strains, as well as addressing vaccine hesitancy and potential approaches to mitigate reluctance. EXPERT OPINION Challenges such as stability, storage, manufacturing complexities, production capacity, allergic reactions, long-term effects, accessibility, and misinformation must be addressed. Despite these hurdles, mRNA vaccine technology holds promise for revolutionizing future vaccination strategies.
Collapse
Affiliation(s)
- Sheema Mir
- College of Veterinary Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Mohammad Mir
- College of Veterinary Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
32
|
Chen K, Jiang M, Liu J, Huang D, Yang YR. DNA nanostructures as biomolecular scaffolds for antigen display. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1921. [PMID: 37562787 DOI: 10.1002/wnan.1921] [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: 04/30/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023]
Abstract
Nanoparticle-based vaccines offer a multivalent approach for antigen display, efficiently activating T and B cells in the lymph nodes. Among various nanoparticle design strategies, DNA nanotechnology offers an innovative alternative platform, featuring high modularity, spatial addressing, nanoscale regulation, high functional group density, and lower self-antigenicity. This review delves into the potential of DNA nanostructures as biomolecular scaffolds for antigen display, addressing: (1) immunological mechanisms behind nanovaccines and commonly used nanoparticles in their design, (2) techniques for characterizing protein NP-antigen complexes, (3) advancements in DNA nanotechnology and DNA-protein assembly approach, (4) strategies for precise antigen presentation on DNA scaffolds, and (5) current applications and future possibilities of DNA scaffolds in antigen display. This analysis aims to highlight the transformative potential of DNA nanoscaffolds in immunology and vaccinology. This article is categorized under: Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Kun Chen
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Ming Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jin Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Deli Huang
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuhe R Yang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Gao X, Wang X, Li S, Saif Ur Rahman M, Xu S, Liu Y. Nanovaccines for Advancing Long-Lasting Immunity against Infectious Diseases. ACS NANO 2023; 17:24514-24538. [PMID: 38055649 DOI: 10.1021/acsnano.3c07741] [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: 12/08/2023]
Abstract
Infectious diseases, particularly life-threatening pathogens such as small pox and influenza, have substantial implications on public health and global economies. Vaccination is a key approach to combat existing and emerging pathogens. Immunological memory is an essential characteristic used to evaluate vaccine efficacy and durability and the basis for the long-term effects of vaccines in protecting against future infections; however, optimizing the potency, improving the quality, and enhancing the durability of immune responses remains challenging and a focus for research involving investigation of nanovaccine technologies. In this review, we describe how nanovaccines can address the challenges for conventional vaccines in stimulating adaptive immune memory responses to protect against reinfection. We discuss protein and nonprotein nanoparticles as useful antigen platforms, including those with highly ordered and repetitive antigen array presentation to enhance immunogenicity through cross-linking with multiple B cell receptors, and with a focus on antigen properties. In addition, we describe how nanoadjuvants can improve immune responses by providing enhanced access to lymph nodes, lymphnode targeting, germinal center retention, and long-lasting immune response generation. Nanotechnology has the advantage to facilitate vaccine induction of long-lasting immunity against infectious diseases, now and in the future.
Collapse
Affiliation(s)
- Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | | | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
34
|
Sun L, Niu J, Zhang J, Peng Y, Feng X, Huang F, Liu J, Li S, Chen Z. Thermostable T Cell Multiepitope Nanoparticle Antigens Inducing Potent Immune Responses against the Swine Fever Virus. ACS Infect Dis 2023; 9:2358-2368. [PMID: 37861250 DOI: 10.1021/acsinfecdis.3c00506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
African swine fever (ASF) is caused by the African swine fever virus (ASFV) and is a highly contagious, acute, febrile disease that has high morbidity and mortality rates in domestic and wild swine. However, a safe and effective vaccine against ASF remains unavailable as single antigens fail to provide sufficient protection. Therefore, a combination of multiple antigens with an efficient delivery system might be an alternative strategy. Herein, a de novo-designed antigen with multiple T-cell epitopes (TEPs) of ASFV was conjugated for surface display on self-assembled nanoparticles (NPs) of Aquifex aeolicus lumazine synthase (AaLS) and Quasibacillus thermotolerans encapsulin (QT) through the SpyCatcher/SpyTag system to construct nanovaccines (TEP-Spy-NPs). TEP-Spy-NPs exhibited significantly more thermal, storage, and freeze-thaw stability in comparison to TEP monomers. TEP-Spy-NPs were highly immunogenic and induced strong polyclonal antibody responses in mice and pigs. The specific antibody titers against the TEP of the TEP-Spy-AaLS and TEP-Spy-QT groups were significantly higher than those of the TEP monomer immune group after the second booster immunization. The antibody titer against TEP of the TEP-Spy-QT group was approximately twice that of the TEP-Spy-AaLS group in mice. ELISpot analysis demonstrated that more IFN-γ- and IL-2-secreting splenic lymphocytes were produced by TEP-Spy-AaLS- and TEP-Spy-QT-immunized mice than by TEP monomer-immunized mice. TEP-Spy-NPs elicited stronger cellular immunity and in vivo immunity in immunized pigs than did TEP monomers. Thus, the TEP nanovaccine successfully induced strong humoral and cellular immune responses in mice and pigs, and TEP-Spy-NPs have the potential as candidate vaccines for ASFV.
Collapse
Affiliation(s)
- Lidan Sun
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Jingqi Niu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
| | - Jinsong Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanli Peng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiangning Feng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Jinling Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning 110866, China
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao 028000, China
| |
Collapse
|
35
|
Sun C, Kang YF, Fang XY, Liu YN, Bu GL, Wang AJ, Li Y, Zhu QY, Zhang H, Xie C, Kong XW, Peng YJ, Lin WJ, Zhou L, Chen XC, Lu ZZ, Xu HQ, Hong DC, Zhang X, Zhong L, Feng GK, Zeng YX, Xu M, Zhong Q, Liu Z, Zeng MS. A gB nanoparticle vaccine elicits a protective neutralizing antibody response against EBV. Cell Host Microbe 2023; 31:1882-1897.e10. [PMID: 37848029 DOI: 10.1016/j.chom.2023.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 08/17/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Epstein-Barr virus (EBV) is a global public health concern, as it is known to cause multiple diseases while also being etiologically associated with a wide range of epithelial and lymphoid malignancies. Currently, there is no available prophylactic vaccine against EBV. gB is the EBV fusion protein that mediates viral membrane fusion and participates in host recognition, making it critical for EBV infection in both B cells and epithelial cells. Here, we present a gB nanoparticle, gB-I53-50 NP, that displays multiple copies of gB. Compared with the gB trimer, gB-I53-50 NP shows improved structural integrity and stability, as well as enhanced immunogenicity in mice and non-human primate (NHP) preclinical models. Immunization and passive transfer demonstrate a robust and durable protective antibody response that protects humanized mice against lethal EBV challenge. This vaccine candidate demonstrates significant potential in preventing EBV infection, providing a possible platform for developing prophylactic vaccines for EBV.
Collapse
Affiliation(s)
- Cong Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yin-Feng Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Xin-Yan Fang
- Cryo-Electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yi-Na Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Guo-Long Bu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Ao-Jie Wang
- Cryo-Electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Qian-Ying Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Hua Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China; MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Chu Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Xiang-Wei Kong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yong-Jian Peng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Wen-Jie Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Ling Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Xin-Chun Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Zheng-Zhou Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Hui-Qin Xu
- Cryo-Electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Dong-Chun Hong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Xiao Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Ling Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Guo-Kai Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China.
| | - Zheng Liu
- Cryo-Electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China.
| |
Collapse
|
36
|
Priyanka, Abusalah MAH, Chopra H, Sharma A, Mustafa SA, Choudhary OP, Sharma M, Dhawan M, Khosla R, Loshali A, Sundriyal A, Saini J. Nanovaccines: A game changing approach in the fight against infectious diseases. Biomed Pharmacother 2023; 167:115597. [PMID: 37783148 DOI: 10.1016/j.biopha.2023.115597] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
The field of nanotechnology has revolutionised global attempts to prevent, treat, and eradicate infectious diseases in the foreseen future. Nanovaccines have proven to be a valuable pawn in this novel technology. Nanovaccines are made up of nanoparticles that are associated with or prepared with components that can stimulate the host's immune system. In addition to their delivery capabilities, the nanocarriers have been demonstrated to possess intrinsic adjuvant properties, working as immune cell stimulators. Thus, nanovaccines have the potential to promote rapid as well as long-lasting humoral and cellular immunity. The nanovaccines have several possible benefits, including site-specific antigen delivery, increased antigen bioavailability, and a diminished adverse effect profile. To avail these benefits, several nanoparticle-based vaccines are being developed, including virus-like particles, liposomes, polymeric nanoparticles, nanogels, lipid nanoparticles, emulsion vaccines, exomes, and inorganic nanoparticles. Inspired by their distinctive properties, researchers are working on the development of nanovaccines for a variety of applications, such as cancer immunotherapy and infectious diseases. Although a few challenges still need to be overcome, such as modulation of the nanoparticle pharmacokinetics to avoid rapid elimination from the bloodstream by the reticuloendothelial system, The future prospects of this technology are also assuring, with multiple options such as personalised vaccines, needle-free formulations, and combination nanovaccines with several promising candidates.
Collapse
Affiliation(s)
- Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India
| | - Mai Abdel Haleem Abusalah
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Abhilasha Sharma
- Department of Life Science, Gujarat University, University School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Suhad Asad Mustafa
- Scientific Research Center/ Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India.
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, Punjab, India; Trafford College, Altrincham, Manchester WA14 5PQ, UK.
| | - Rajiv Khosla
- Department of Biotechnology, Doaba College, Jalandhar 144004, Punjab, India
| | - Aanchal Loshali
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ankush Sundriyal
- School of Pharmaceutical Sciences and Research, Sardar Bhagwan Singh University, Balawala, Dehradun 248001, India
| | - Jyoti Saini
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda 151103, Punjab, India
| |
Collapse
|
37
|
Sam G, Stenos J, Graves SR, Rehm BHA. Q fever immunology: the quest for a safe and effective vaccine. NPJ Vaccines 2023; 8:133. [PMID: 37679410 PMCID: PMC10484952 DOI: 10.1038/s41541-023-00727-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Q fever is an infectious zoonotic disease, caused by the Gram-negative bacterium Coxiella burnetii. Transmission occurs from livestock to humans through inhalation of a survival form of the bacterium, the Small Cell Variant, often via handling of animal parturition products. Q fever manifests as an acute self-limiting febrile illness or as a chronic disease with complications such as vasculitis and endocarditis. The current preventative human Q fever vaccine Q-VAX poses limitations on its worldwide implementation due to reactogenic responses in pre-sensitized individuals. Many strategies have been undertaken to develop a universal Q fever vaccine but with little success to date. The mechanisms of the underlying reactogenic responses remain only partially understood and are important factors in the development of a safe Q fever vaccine. This review provides an overview of previous and current experimental vaccines developed for use against Q fever and proposes approaches to develop a vaccine that establishes immunological memory while eliminating harmful reactogenic responses.
Collapse
Affiliation(s)
- Gayathri Sam
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - John Stenos
- Australian Rickettsial Reference Laboratory, University Hospital, Geelong, VIC, 3220, Australia
| | - Stephen R Graves
- Australian Rickettsial Reference Laboratory, University Hospital, Geelong, VIC, 3220, Australia
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, Menangle, NSW, 2567, Australia
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia.
| |
Collapse
|
38
|
He X, Wang J, Tang Y, Chiang ST, Han T, Chen Q, Qian C, Shen X, Li R, Ai X. Recent Advances of Emerging Spleen-Targeting Nanovaccines for Immunotherapy. Adv Healthc Mater 2023; 12:e2300351. [PMID: 37289567 DOI: 10.1002/adhm.202300351] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/19/2023] [Indexed: 06/10/2023]
Abstract
Vaccines provide a powerful tool to modulate the immune system for human disease prevention and treatment. Classical vaccines mainly initiate immune responses in the lymph nodes (LNs) after subcutaneous injection. However, some vaccines suffer from inefficient delivery of antigens to LNs, undesired inflammation, and slow immune induction when encountering the rapid proliferation of tumors. Alternatively, the spleen, as the largest secondary lymphoid organ with a high density of antigen-presenting cells (APCs) and lymphocytes, acts as an emerging target organ for vaccinations in the body. Upon intravenous administration, the rationally designed spleen-targeting nanovaccines can be internalized by the APCs in the spleen to induce selective antigen presentation to T and B cells in their specific sub-regions, thereby rapidly boosting durable cellular and humoral immunity. Herein, the recent advances of spleen-targeting nanovaccines for immunotherapy based on the anatomical architectures and functional zones of the spleen, as well as their limitations and perspectives for clinical applications are systematically summarized. The aim is to emphasize the design of innovative nanovaccines for enhanced immunotherapy of intractable diseases in the future.
Collapse
Affiliation(s)
- Xuanyi He
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Jing Wang
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Yuqing Tang
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Seok Theng Chiang
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Tianzhen Han
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Qi Chen
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Chunxi Qian
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiaoshuai Shen
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Rongxiu Li
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiangzhao Ai
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| |
Collapse
|
39
|
Sadr S, Poorjafari Jafroodi P, Haratizadeh MJ, Ghasemi Z, Borji H, Hajjafari A. Current status of nano-vaccinology in veterinary medicine science. Vet Med Sci 2023; 9:2294-2308. [PMID: 37487030 PMCID: PMC10508510 DOI: 10.1002/vms3.1221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/11/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
Vaccination programmes provide a safe, effective and cost-efficient strategy for maintaining population health. In veterinary medicine, vaccination not only reduces disease within animal populations but also serves to enhance public health by targeting zoonoses. Nevertheless, for many pathogens, an effective vaccine remains elusive. Recently, nanovaccines have proved to be successful for various infectious and non-infectious diseases of animals. These novel technologies, such as virus-like particles, self-assembling proteins, polymeric nanoparticles, liposomes and virosomes, offer great potential for solving many of the vaccine production challenges. Their benefits include low immunotoxicity, antigen stability, enhanced immunogenicity, flexibility sustained release and the ability to evoke both humoral and cellular immune responses. Nanovaccines are more efficient than traditional vaccines due to ease of control and plasticity in their physio-chemical properties. They use a highly targeted immunological approach which can provide strong and long-lasting immunity. This article reviews the currently available nanovaccine technology and considers its utility for both infectious diseases and non-infectious diseases such as auto-immunity and cancer. Future research opportunities and application challenges from bench to clinical usage are also discussed.
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Clinical SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | | | | | - Zahra Ghasemi
- Department of Clinical SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Hassan Borji
- Department of PathobiologyFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Ashkan Hajjafari
- Department of PathobiologyFaculty of Veterinary MedicineIslamic Azad University, Science and Research BranchTehranIran
| |
Collapse
|
40
|
Ung T, Rutledge NS, Weiss AM, Esser-Kahn AP, Deak P. Cell-targeted vaccines: implications for adaptive immunity. Front Immunol 2023; 14:1221008. [PMID: 37662903 PMCID: PMC10468591 DOI: 10.3389/fimmu.2023.1221008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Recent advancements in immunology and chemistry have facilitated advancements in targeted vaccine technology. Targeting specific cell types, tissue locations, or receptors can allow for modulation of the adaptive immune response to vaccines. This review provides an overview of cellular targets of vaccines, suggests methods of targeting and downstream effects on immune responses, and summarizes general trends in the literature. Understanding the relationships between vaccine targets and subsequent adaptive immune responses is critical for effective vaccine design. This knowledge could facilitate design of more effective, disease-specialized vaccines.
Collapse
Affiliation(s)
- Trevor Ung
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Nakisha S. Rutledge
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Peter Deak
- Chemical and Biological Engineering Department, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
41
|
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
|
42
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
43
|
Vu MN, Pilkington EH, Lee WS, Tan H, Davis TP, Truong NP, Kent SJ, Wheatley AK. Engineered Ferritin Nanoparticle Vaccines Enable Rapid Screening of Antibody Functionalization to Boost Immune Responses. Adv Healthc Mater 2023; 12:e2202595. [PMID: 36786027 PMCID: PMC11469303 DOI: 10.1002/adhm.202202595] [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: 10/10/2022] [Revised: 01/16/2023] [Indexed: 02/15/2023]
Abstract
Employing monoclonal antibodies to target vaccine antigens to different immune cells within lymph nodes where adaptive immunity is initiated can provide a mechanism to fine-tune the magnitude or the quality of immune responses. However, studying the effects of different targeting antibodies head-to-head is challenging due to the lack of a feasible method that allows rapid screening of multiple antibodies for their impact on immunogenicity. Here self-assembling ferritin nanoparticles are prepared that co-display vaccine antigens and the Fc-binding domain of Staphylococcal protein A, allowing rapid attachment of soluble antibodies to the nanoparticle surface. Using this tunable system, ten antibodies targeting different immune cell subsets are screened, with targeting to Clec9a associated with higher serum antibody titers after immunization. Immune cell targeting using ferritin nanoparticles with anti-Clec9a antibodies drives concentrated deposition of antigens within germinal centers, boosting germinal center formation and robust antibody responses. However, the capacity to augment humoral immunity is antigen-dependent, with significant boosting observed for prototypic ovalbumin immunogens but reduced effectiveness with the SARS-CoV-2 RBD. This work provides a rapid platform for screening targeting antibodies, which will accelerate mechanistic insights into optimal delivery strategies for nanoparticle-based vaccines to maximize protective immunity.
Collapse
Affiliation(s)
- Mai N. Vu
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Faculty of Pharmaceutics and Pharmaceutical TechnologyHanoi University of Pharmacy10000HanoiVietnam
| | - Emily H. Pilkington
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Wen Shi Lee
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
| | - Hyon‐Xhi Tan
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
| | - Thomas P. Davis
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Australia Institute of Bioengineering & NanotechnologyUniversity of QueenslandBrisbaneQLD4072Australia
| | - Nghia P. Truong
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Stephen J. Kent
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Melbourne Sexual Health Centre and Department of Infectious DiseasesAlfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVIC3004Australia
| | - Adam K. Wheatley
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
| |
Collapse
|
44
|
Hao H, Wu S, Lin J, Zheng Z, Zhou Y, Zhang Y, Guo Q, Tian F, Zhao M, Chen Y, Xu X, Hou L, Wang X, Tang R. Immunization against Zika by entrapping live virus in a subcutaneous self-adjuvanting hydrogel. Nat Biomed Eng 2023; 7:928-942. [PMID: 36959404 DOI: 10.1038/s41551-023-01014-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 02/20/2023] [Indexed: 03/25/2023]
Abstract
The threat of new viral outbreaks has heightened the need for ready-to-use vaccines that are safe and effective. Here we show that a subcutaneous vaccine consisting of live Zika virus electrostatically entrapped in a self-adjuvanting hydrogel recruited immune cells at the injection site and provided mice with effective protection against a lethal viral challenge. The hydrogel prevented the escape of the viral particles and upregulated pattern recognition receptors that activated innate antiviral immunity. The local inflammatory niche facilitated the engulfment of the virus by immune cells infiltrating the hydrogel, the processing and cross-presentation of antigens and the expansion of germinal centre B cells and induced robust antigen-specific adaptive responses and immune memory. Inflammatory immune niches entrapping live viruses may facilitate the rapid development of safe and efficacious vaccines.
Collapse
Affiliation(s)
- Haibin Hao
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Jiake Lin
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Zitong Zheng
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Yuemin Zhou
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Ying Zhang
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Qiang Guo
- Beijing Institute of Biotechnology, Beijing, China
| | - Fengchao Tian
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Mengsu Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Xurong Xu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China.
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China.
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China.
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
45
|
Huang Y, Zhu X, Guo X, Zhou Y, Liu D, Mao J, Xiong Y, Deng Y, Gao X. Advances in mRNA vaccines for viral diseases. J Med Virol 2023; 95:e28924. [PMID: 37417396 DOI: 10.1002/jmv.28924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
Since the onset of the pandemic caused by severe acute respiratory syndrome coronavirus 2, messenger RNA (mRNA) vaccines have demonstrated outstanding performance. mRNA vaccines offer significant advantages over conventional vaccines in production speed and cost-effectiveness, making them an attractive option against other viral diseases. This article reviewed recent advances in viral mRNA vaccines and their delivery systems to provide references and guidance for developing mRNA vaccines for new viral diseases.
Collapse
Affiliation(s)
- Yukai Huang
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xuerui Zhu
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiao Guo
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuancheng Zhou
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
| | - Dongying Liu
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingrui Mao
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yongai Xiong
- Department of Pharmaceutics, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Youcai Deng
- Department of Hematology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xinghong Gao
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Provincial Department of Education, Key Laboratory of Infectious Disease & Bio-Safety, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
46
|
Sheng Y, Li Z, Lin X, Ma Y, Ren Y, Su Z, Ma G, Zhang S. The position of Spy Tag/Catcher system in hepatitis B core protein particles affects the immunogenicity and stability of the synthetic vaccine. Vaccine 2023:S0264-410X(23)00759-4. [PMID: 37391312 DOI: 10.1016/j.vaccine.2023.06.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023]
Abstract
Presenting exogenous antigens on virus-like particles (VLPs) through "plug-and-display" decoration strategies based on SpyTag/SpyCatcher isopeptide bonding have emerged as attractive technology for vaccine synthesis. However, whether the position of ligation site in VLPs will impose effects on immunogenicity and physiochemical properties of the synthetic vaccine remains rarely investigated. Here in the present work, the well-established hepatitis B core (HBc) protein was used as chassis to construct dual-antigen influenza nanovaccines, with the conserved epitope peptides derived from extracellular domain of matrix protein M2 (M2e) and hemagglutinin (HA) as target antigens. The M2e antigen was genetically fused to the HBc in the MIR region, together with the SpyTag peptide, which was fused either in the MIR region or at the N-terminal of the protein, so that a recombinant HA antigen (rHA) linked to SpyCatcher can be displayed on it, at two different localizations. Both synthetic nanovaccines showed ability in inducing strong M2e and rHA-specific antibodies and cellular immunogenicity; nevertheless, the one in which rHA was conjugated by N-terminal Tag ligation, was superior to another one synthesized by linking the rHA to MIR region SpyTagged-HBc in all aspects, including higher antigen-specific immunogenicity responses, lower anti-HBc carrier antibody, as well as better dispersion stability. Surface charge and hydrophobicity properties of the two synthetic nanovaccines were analyzed, results revealed that linking the rHA to MIR region SpyTagged-HBc lead to more significant and disadvantageous alteration in physiochemical properties of the HBc chassis. This study will expand our knowledge on "plug-and-display" decoration strategies and provide helpful guidance for the rational design of HBc-VLPs based modular vaccines by using SpyTag/Catcher synthesis.
Collapse
Affiliation(s)
- Yanan Sheng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xuan Lin
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Yanyan Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Ying Ren
- State Key Laboratory of Multiphase Complex Systems, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiguo Su
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Songping Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
47
|
Wamhoff EC, Ronsard L, Feldman J, Knappe GA, Hauser BM, Romanov A, Lam E, Denis KS, Boucau J, Barczak AK, Balazs AB, Schmidt A, Lingwood D, Bathe M. Enhancing antibody responses by multivalent antigen display on thymus-independent DNA origami scaffolds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2022.08.16.504128. [PMID: 36032975 PMCID: PMC9413718 DOI: 10.1101/2022.08.16.504128] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multivalent antigen display is a well-established principle to enhance humoral immunity. Protein-based virus-like particles (VLPs) are commonly used to spatially organize antigens. However, protein-based VLPs are limited in their ability to control valency on fixed scaffold geometries and are thymus-dependent antigens that elicit neutralizing B cell memory themselves, which can distract immune responses. Here, we investigated DNA origami as an alternative material for multivalent antigen display in vivo, applied to the receptor binding domain (RBD) of SARS-CoV2 that is the primary antigenic target of neutralizing antibody responses. Icosahedral DNA-VLPs elicited neutralizing antibodies to SARS-CoV-2 in a valency-dependent manner following sequential immunization in mice, quantified by pseudo- and live-virus neutralization assays. Further, induction of B cell memory against the RBD required T cell help, but the immune sera did not contain boosted, class-switched antibodies against the DNA scaffold. This contrasted with protein-based VLP display of the RBD that elicited B cell memory against both the target antigen and the scaffold. Thus, DNA-based VLPs enhance target antigen immunogenicity without generating off-target, scaffold-directed immune memory, thereby offering a potentially important alternative material for particulate vaccine design.
Collapse
Affiliation(s)
- Eike-Christian Wamhoff
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Larance Ronsard
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Jared Feldman
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Grant A. Knappe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Blake M. Hauser
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Anna Romanov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Evan Lam
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Kerri St. Denis
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Julie Boucau
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Amy K Barczak
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Alejandro B. Balazs
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Aaron Schmidt
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, United States
| | - Daniel Lingwood
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, United States
| | - Mark Bathe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, United States
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
48
|
Mao L, Ma P, Luo X, Cheng H, Wang Z, Ye E, Loh XJ, Wu YL, Li Z. Stimuli-Responsive Polymeric Nanovaccines Toward Next-Generation Immunotherapy. ACS NANO 2023; 17:9826-9849. [PMID: 37207347 DOI: 10.1021/acsnano.3c02273] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The development of nanovaccines that employ polymeric delivery carriers has garnered substantial interest in therapeutic treatment of cancer and a variety of infectious diseases due to their superior biocompatibility, lower toxicity and reduced immunogenicity. Particularly, stimuli-responsive polymeric nanocarriers show great promise for delivering antigens and adjuvants to targeted immune cells, preventing antigen degradation and clearance, and increasing the uptake of specific antigen-presenting cells, thereby sustaining adaptive immune responses and improving immunotherapy for certain diseases. In this review, the most recent advances in the utilization of stimulus-responsive polymer-based nanovaccines for immunotherapeutic applications are presented. These sophisticated polymeric nanovaccines with diverse functions, aimed at therapeutic administration for disease prevention and immunotherapy, are further classified into several active domains, including pH, temperature, redox, light and ultrasound-sensitive intelligent nanodelivery systems. Finally, the potential strategies for the future design of multifunctional next-generation polymeric nanovaccines by integrating materials science with biological interface are proposed.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Panqin Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xi Luo
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhanxiang Wang
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Enyi Ye
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| |
Collapse
|
49
|
Tsai HH, Huang PH, Lin LC, Yao BY, Liao WT, Pai CH, Liu YH, Chen HW, Hu CMJ. Lymph Node Follicle-Targeting STING Agonist Nanoshells Enable Single-Shot M2e Vaccination for Broad and Durable Influenza Protection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2206521. [PMID: 37092580 DOI: 10.1002/advs.202206521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/14/2023] [Indexed: 05/03/2023]
Abstract
The highly conserved matrix protein 2 ectodomain (M2e) of influenza viruses presents a compelling vaccine antigen candidate for stemming the pandemic threat of the mutation-prone pathogen, yet the low immunogenicity of the diminutive M2e peptide renders vaccine development challenging. A highly potent M2e nanoshell vaccine that confers broad and durable influenza protectivity under a single vaccination is shown. Prepared via asymmetric ionic stabilization for nanoscopic curvature formation, polymeric nanoshells co-encapsulating high densities of M2e peptides and stimulator of interferon genes (STING) agonists are prepared. Robust and long-lasting protectivity against heterotypic influenza viruses is achieved with a single administration of the M2e nanoshells in mice. Mechanistically, molecular adjuvancy by the STING agonist and nanoshell-mediated prolongation of M2e antigen exposure in the lymph node follicles synergistically contribute to the heightened anti-M2e humoral responses. STING agonist-triggered T cell helper functions and extended residence of M2e peptides in the follicular dendritic cell network provide a favorable microenvironment that induces Th1-biased antibody production against the diminutive antigen. These findings highlight a versatile nanoparticulate design that leverages innate immune pathways for enhancing the immunogenicity of weak immunogens. The single-shot nanovaccine further provides a translationally viable platform for pandemic preparedness.
Collapse
Affiliation(s)
- Hsiao-Han Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
| | - Ping-Han Huang
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Leon Cw Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Bing-Yu Yao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Wan-Ting Liao
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Chen-Hsueh Pai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yu-Han Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Che-Ming J Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
50
|
Zhang YN, Paynter J, Antanasijevic A, Allen JD, Eldad M, Lee YZ, Copps J, Newby ML, He L, Chavez D, Frost P, Goodroe A, Dutton J, Lanford R, Chen C, Wilson IA, Crispin M, Ward AB, Zhu J. Single-component multilayered self-assembling protein nanoparticles presenting glycan-trimmed uncleaved prefusion optimized envelope trimmers as HIV-1 vaccine candidates. Nat Commun 2023; 14:1985. [PMID: 37031217 PMCID: PMC10082823 DOI: 10.1038/s41467-023-37742-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Uncleaved prefusion-optimized (UFO) design can stabilize diverse HIV-1 envelope glycoproteins (Envs). Single-component, self-assembling protein nanoparticles (1c-SApNP) can display 8 or 20 native-like Env trimers as vaccine candidates. We characterize the biophysical, structural, and antigenic properties of 1c-SApNPs that present the BG505 UFO trimer with wildtype and modified glycans. For 1c-SApNPs, glycan trimming improves recognition of the CD4 binding site without affecting broadly neutralizing antibodies (bNAbs) to major glycan epitopes. In mice, rabbits, and nonhuman primates, glycan trimming increases the frequency of vaccine responders (FVR) and steers antibody responses away from immunodominant glycan holes and glycan patches. The mechanism of vaccine-induced immunity is examined in mice. Compared with the UFO trimer, the multilayered E2p and I3-01v9 1c-SApNPs show 420 times longer retention in lymph node follicles, 20-32 times greater presentation on follicular dendritic cell dendrites, and up-to-4 times stronger germinal center reactions. These findings can inform future HIV-1 vaccine development.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jennifer Paynter
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Aleksandar Antanasijevic
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Joel D Allen
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Mor Eldad
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Maddy L Newby
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Deborah Chavez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Pat Frost
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Anna Goodroe
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - John Dutton
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Robert Lanford
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Christopher Chen
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Max Crispin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|