1
|
Yang BF, Jin J, He XR, Yang ZH, Qian X, Tong YQ, Ke CX, Li ZH, Li ZX, Zhong LF, Gan ZH, Zhang XF, Zeng Y. Immunogenicity and safety of SARS-CoV-2 recombinant protein vaccine (CHO cell) LYB001 as a heterologous booster following two- or three-dose inactivated COVID-19 vaccine in adults aged ≥18 years: interim results of a randomized, active-controlled, double-blinded, phase 3 trial. Expert Rev Vaccines 2025; 24:81-90. [PMID: 39720838 DOI: 10.1080/14760584.2024.2446288] [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: 06/12/2024] [Revised: 10/13/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND LYB001 is a recombinant protein COVID-19 vaccine displaying a receptor-binding domain (RBD) in a highly immunogenic array on virus-like particles (VLPs). This study assessed the immunogenicity and safety of LYB001 as a booster. RESEARCH DESIGN AND METHODS In this randomized, active-controlled, double-blinded, phase 3 trial, participants aged ≥ 18 years received a booster with LYB001 or ZF2001 (Recombinant COVID-19 Vaccine). The primary endpoint was to compare the geometric mean titer (GMT) of neutralizing antibodies against Omicron BA.4/5 at 14 days after the booster. RESULTS Overall, 1,200 participants aged ≥ 18 years were enrolled, 599 received LYB001, and 601 received ZF2001. Based on similar baseline level, the 14-day GMT ratio (LYB001/ZF2001) against Omicron BA.4/5 was 1.39 (95% CI: 1.25, 1.56), demonstrating superiority (95% CI lower limit > 1) of LYB001. The spike protein-binding IgG concentrations induced by LYB001 were significantly higher than those induced by ZF2001 on day 14 and day 28 after the booster (p-value <0.0001). LYB001 recipients reported more adverse reactions than ZF2001 recipients (21.4% vs. 15.0%); however, all adverse reactions in the LYB001 group were mild-to-moderate. CONCLUSIONS LYB001 is highly immunogenic and retains a well-characterized safety profile in adults aged ≥ 18 years. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov, identifier is NCT05664932.
Collapse
Affiliation(s)
- Bei-Fang Yang
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Jing Jin
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| | - Xi-Ran He
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| | - Zhong-Hua Yang
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| | - Xiao'ai Qian
- Yantai Patronus Biotech Co., Ltd., Yantai, China
| | - Ye-Qing Tong
- Hubei Institute for Infectious Disease Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Chang-Xian Ke
- Immunization Prevention Department Shiyan City Yunyang District Center for Disease Control and Prevention, Shiyan, China
| | - Zhao-Hong Li
- Immunization Prevention Department Shiyan City Yunyang District Center for Disease Control and Prevention, Shiyan, China
| | - Zhao-Xia Li
- Immunization Prevention Department, Gucheng Center for Disease Control and Prevention, Xiangyang, China
| | - Lin-Feng Zhong
- Immunization Prevention Department, Wuxue City Center for Disease Control and Prevention, Wuxue, China
| | - Ze-Hong Gan
- Immunization Prevention Department, Wuxue City Center for Disease Control and Prevention, Wuxue, China
| | - Xian-Feng Zhang
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Ying Zeng
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| |
Collapse
|
2
|
Edeling MA, Earnest L, Carrera Montoya J, Yap AHY, Mumford J, Roberts J, Wong CY, Hans D, Grima J, Bisset N, Bodle J, Rockman S, Torresi J. Development of Methods to Produce SARS CoV-2 Virus-Like Particles at Scale. Biotechnol Bioeng 2025; 122:1118-1129. [PMID: 39936889 PMCID: PMC11975197 DOI: 10.1002/bit.28937] [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: 05/14/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025]
Abstract
The devastating global toll precipitated by the SARS CoV-2 outbreak and the profound impact of vaccines in stemming that outbreak has established the need for molecular platforms capable of rapidly delivering effective, safe and accessible medical interventions in pandemic preparedness. We describe a simple, efficient and adaptable process to produce SARS CoV-2 virus-like particles (VLPs) that can be readily scaled for manufacturing. A rapid but gentle method of tangential flow filtration using a 100 kDa semi-permeable membrane concentrates and buffer exchanges 0.5 L of SARS CoV-2 VLP containing supernatant into low salt and optimal pH for anion exchange chromatography. VLPs are washed, eluted under high salt, dialyzed into physiological buffer, sterile filtered and aliquoted for storage at -80°C. Purification is completed in less than 2 days. A simple quality control process includes Western blot for coincident detection of Spike, Membrane and Envelope protein as a proxy for intact VLP, ELISA to detect conformationally sensitive Spike using readily available anti-Spike and/or anti-RBD antibodies, and negative stain and immunogold electron microscopy to validate particulate, Spike crowned VLPs. This process to produce SARS CoV-2 VLPs for preclinical studies serves as a roadmap for preparation of more distantly related VLPs for pandemic preparedness.
Collapse
Affiliation(s)
- Melissa A. Edeling
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Linda Earnest
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Julio Carrera Montoya
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Ashley Huey Yiing Yap
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Jamie Mumford
- Victorian Infectious Diseases Reference laboratoryRoyal Melbourne Hospital at the Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Jason Roberts
- Victorian Infectious Diseases Reference laboratoryRoyal Melbourne Hospital at the Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
- Department of Infectious DiseasesThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Chinn Yi Wong
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Dhiraj Hans
- Research, Innovation & Commercialisation, Faculty of Medicine, Dentistry & Health SciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Joseph Grima
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Nicole Bisset
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Jesse Bodle
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Steven Rockman
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Joseph Torresi
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| |
Collapse
|
3
|
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. [PMID: 40265996 DOI: 10.1021/acsnano.4c17269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
4
|
Jia J, Hao Y, Zhang L, Cao X, An L, Wang H, Ma Q, Jin X, Ma X. Development and validation of optimized lentivirus-like particles for gene editing tool delivery with Gag-Only strategy. Eur J Med Res 2025; 30:242. [PMID: 40186294 PMCID: PMC11969815 DOI: 10.1186/s40001-025-02499-2] [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: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The development of gene editing tools such as CRISPR-Cas9 and base editors (BE) is critical for genetic diseases and cancer. Lentivirus-like particles (LVLPs) grows into an auspicious platform for delivering mRNA or ribonucleic proteins (RNPs) due to it integrates the advantage of viral and non-viral vectors. Current LVLP systems predominantly utilize HIV-Gag and Pol proteins. However, the reverse transcriptase and integrase of Pol, pose risks of genomic integration and potential tumorigenesis. Enhancing the safety of VLP system is essential. This study focuses on improving the LVLP to minimize these risks. METHODS We implemented a Gag-Only strategy, constructing LVLPs with HIV-Gag protein, thereby eliminating the integration risks linked to Pol. By leveraging the interactions between MS2-MCP (MS2 coat protein), PP7 and PP7 BP (PP7 binding protein), and the psi (HIV packaging signal) with HIV-Gag, we encapsulated PAMless andesine base editor (CE-8e-SpRY) mRNA and sgRNA targeting the PD1 start codon (ATG) into the LVLP. Using recombinant lentiviral vector technology, we developed a stable PD1-expressing 293T cell line (PD1-293T) to assess the editing efficiency of LVLP. RESULTS The psi-LVLP demonstrated effective packaging capabilities, achieving 15% base editing efficiency in 293T cells. By optimizing plasmid ratios, we observed increased CE-8e-SpRY mRNA copy numbers, with 30% base editing efficiency. Additionally, the integration of HDVrz (hepatitis delta virus ribozyme) and psi into sgRNA (HDVrz-psi-LVLP) substantially enhanced sgRNA copy numbers, resulting in approximately 50% base editing efficiency in 293T cells and 20% base editing efficiency in Jurkat cells. Mendelian randomization analyses revealed significant genetic correlations between PD1, B2M, CIITA, and TIGIT genes with various cancer risks. Furthermore, HDVrz-psi-LVLPs targeting the start codons of B2M, CIITA, and TIGIT exhibited high base editing activity in both Jurkat and 293T cells. CONCLUSION In conclusion, this optimized platform effectively encapsulates CE-8e-SpRY mRNA and sgRNA, achieving high editing efficiencies across multiple genes and cell types. We introduce a safer and more efficient gene editing tool delivery system by constructing LVLPs based on the Gag-Only strategy. Our study presents a promising implication for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinlin Jia
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Yanzhe Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 100052, China.
| | - Lu Zhang
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Xiaofang Cao
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Lisha An
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Hu Wang
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Qi Ma
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 100052, China
| | - Xiaohua Jin
- National Research Institute for Family Planning, Beijing, 100081, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- National Human Genetic Resources Center, Beijing, 100052, China.
| | - Xu Ma
- National Research Institute for Family Planning, Beijing, 100081, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- National Human Genetic Resources Center, Beijing, 100052, China.
| |
Collapse
|
5
|
Ball J, Bradley A, Le A, Tisdale JF, Uchida N. Current and future treatments for sickle cell disease: From hematopoietic stem cell transplantation to in vivo gene therapy. Mol Ther 2025:S1525-0016(25)00190-X. [PMID: 40083162 DOI: 10.1016/j.ymthe.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Sickle cell disease (SCD) is a single-gene disorder caused by a point mutation of the β-globin gene, resulting in hemolytic anemia, acute pain, multiorgan damage, and early mortality. Hydroxyurea is a first-line drug therapy that switches sickle-globin to non-pathogenic γ-globin; however, it requires lifelong oral administration. Allogeneic hematopoietic stem cell (HSC) transplantation allows for a one-time cure for SCD, albeit with histocompatibility limitations. Therefore, autologous HSC gene therapy was developed to cure SCD in a single treatment, without HSC donors. Current HSC gene therapy is based on the ex vivo culture of patients' HSCs with lentiviral gene addition and gene editing, followed by autologous transplantation back to the patient. However, the complexity of the treatment process and high costs hinder the universal application of ex vivo gene therapy. Therefore, the development of in vivo HSC gene therapy, where gene therapy tools are directly administered to patients, is desirable to provide a more accessible, cost-effective solution that can cure SCD worldwide. In this review, we discuss current treatments, including drug therapies, HSC transplantation, and ex vivo gene therapy; the development of gene therapy tools; and progress toward curative in vivo gene therapy in SCD.
Collapse
Affiliation(s)
- Julia Ball
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Avery Bradley
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anh Le
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Yilmaz IC, Ipekoglu EM, Golcuklu BS, Bildik T, Aksoy AGB, Evcili I, Turay N, Surucu N, Bulbul A, Guvencli N, Yildirim M, Canavar Yildirim T, Atalay YA, Abras I, Ceylan Y, Ozsurekci Y, Tigen ET, Korten V, Gursel M, Gursel I. A phase I/II study of CpG/alum-adjuvanted mammalian-derived quadruple antigen carrying virus-like particle COVID-19 vaccine. Vaccine 2025; 49:126787. [PMID: 39892108 DOI: 10.1016/j.vaccine.2025.126787] [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: 09/29/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Waning Spike-elicited immunity and emerging COVID-19 variants underscore the need for vaccines leveraging multiple SARS-CoV-2 antigens, rapidly adaptable to evolving strains. Herein, we evaluated the safety and immunogenicity of a CpG ODN-adjuvanted, alum-adsorbed, virus-like particle (VLP) vaccine displaying the hexaproline stabilized Spike (S) protein and the Nucleocapsid, Membrane, and Envelope proteins of SARS-CoV-2. METHODS In phase 1 randomized, double-blind, placebo-controlled, dose-escalation trial, participants (N = 38, aged 18-59) received two subcutaneous injections of either 10 μg or 40 μg of VLP or placebo, 21 days apart. The primary and secondary objectives of the study was to evaluate the safety, reactogenicity and immunogenicity, respectively. In the double blind, multi-center phase-2 study, participants (N = 349, aged 18-55) were randomized into three cohorts receiving two doses of 40 μg VLPs displaying Wuhan-Spike, Alpha-Spike, or a combination. The primary and secondary objectives were humoral, and cell mediated immunogenicity (CMI) and safety, respectively. Antibody responses were analyzed using ELISA while ELIspot and CBA assays were used to assess the CMI. RESULTS The VLP vaccine demonstrated a good safety profile, with 255 non-serious adverse events in phase 1 and 308 in phase 2. Five serious AEs were reported in phase 2, all of which were resolved completely. The VLP vaccine, in phase 2, was well-tolerated, elicited moderate but sustained anti-S and anti-N antibody titers for 180 days and induced T-helper-1 biased cellular responses in participants. CONCLUSIONS The VLP platform is rapidly adaptable to accommodate stabilized Spike proteins from emerging variants and inclusion of other structural SARS-CoV-2 proteins could broaden the breadth of T cell-mediated immunity. CLINICALTRIALS gov; NCT04818281 and NCT04962893.
Collapse
MESH Headings
- Humans
- Adult
- Female
- Male
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/adverse effects
- Middle Aged
- Double-Blind Method
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/adverse effects
- SARS-CoV-2/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Young Adult
- COVID-19/prevention & control
- COVID-19/immunology
- Adolescent
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Immunogenicity, Vaccine
- Adjuvants, Immunologic/administration & dosage
- Alum Compounds/administration & dosage
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Adjuvants, Vaccine/administration & dosage
Collapse
Affiliation(s)
- Ismail Cem Yilmaz
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Emre Mert Ipekoglu
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | | | - Tugce Bildik
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | | | - Irem Evcili
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Nilsu Turay
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye
| | - Naz Surucu
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Artun Bulbul
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Nese Guvencli
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Muzaffer Yildirim
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye
| | - Tugce Canavar Yildirim
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Yagmur Aydin Atalay
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkiye
| | - Irem Abras
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Yasemin Ceylan
- Bilkent University, Molecular Biology and Genetics Department, Bilkent, Ankara, Turkiye
| | - Yasemin Ozsurekci
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye; Hacettepe University, Faculty of Medicine, Department of Pediatric Infectious Diseases, Hacettepe University, Faculty of Medicine, Ankara, Turkiye
| | - Elif Tukenmez Tigen
- Marmara University, Infectious Disease and Clinical Microbiology Department, Istanbul, Turkiye
| | - Volkan Korten
- Marmara University, Infectious Disease and Clinical Microbiology Department, Istanbul, Turkiye
| | - Mayda Gursel
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye.
| | - Ihsan Gursel
- Izmir Biomedicine and Genome Center, DEU Saglik Kampusu, Balcova, Izmir, Turkiye.
| |
Collapse
|
7
|
Liu X. Opportunities and challenges of mRNA technologies in development of dengue virus vaccine. Front Immunol 2025; 16:1520968. [PMID: 40109333 PMCID: PMC11919880 DOI: 10.3389/fimmu.2025.1520968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Dengue virus (DENV) is a mosquito-borne virus with a significant human health concern. With 390 million infections annually and 96 million showing clinical symptoms, severe dengue can lead to life-threatening conditions like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). The only FDA-approved vaccine, Dengvaxia, has limitations due to antibody-dependent enhancement (ADE), necessitating careful administration. The recent pre-approval of TAK-003 by WHO in 2024 highlights ongoing efforts to improve vaccine options. This review explores recent advancements in dengue vaccine development, emphasizing potential utility of mRNA-based vaccines. By examining current clinical trial data and innovations, we aim to identify promising strategies to address the limitations of existing vaccines and enhance global dengue prevention efforts.
Collapse
Affiliation(s)
- Xiaoyang Liu
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
8
|
Maphis NM, Hulse J, Peabody J, Dadras S, Whelpley MJ, Kandath M, Wilson C, Hobson S, Thompson J, Poolsup S, Beckman D, Ott SP, Watanabe JW, Usachenko JL, Van Rompay KK, Morrison J, Selwyn R, Rosenberg G, Knoefel J, Chackerian B, Bhaskar K. Targeting of phosphorylated tau at threonine 181 by a Qβ virus-like particle vaccine is safe, highly immunogenic, and reduces disease severity in mice and rhesus macaques. Alzheimers Dement 2025; 21:e70101. [PMID: 40145301 PMCID: PMC11947757 DOI: 10.1002/alz.70101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Pathological accumulation of tau (pTau) contributes to various tauopathies, including Alzheimer's disease (AD), and correlates with cognitive decline. A rapid surge in tau-targeted approaches via anti-sense oligonucleotides, active/passive immunotherapies suggests that targeting p-Tau is a viable strategy against tauopathies. METHOD We describe a multi-species validation of our previously described Qß virus-like particle (VLP)-based vaccine technology targeting phosphorylated tau on threonine 181 (pT181-Qß). RESULTS Two vaccine doses of pT181-Qß, without any adjuvants, elicited robust antibody responses in two different mouse models of tauopathy (PS19 and hTau) and rhesus macaques. In mouse models, vaccination reduced AT180+ hyperphosphorylated, Sarkosyl insoluble, Gallyas silver positive tau, inflammasomes/neuroinflammation, and improved recognition memory and motor function without inducing adverse T-cell activation. Anti-pT181 antibodies are reactive to pTau in human AD brains, engage pT181+ tau in human brain lysates, and are central nervous system bioavailable. DISCUSSION Our results suggest the translational utility of pT181-Qß against tauopathies. HIGHLIGHTS Icosahedral display of phosphorylated tau at threonine 181 (pT181) Qß virus-like particle surface ("pT181-Qß" vaccine) induces a robust immune response in mice and in non-human primates (NHPs) pT181-Qß vaccination reduces pathological tau (pTau) and brain atrophy, and improves memory and motor function in PS19 and hTau mice. pT181-Qß vaccination-induced immunoglobulin Gs (IgGs) are safe, Th2 skewed (anti-inflammatory), specific to pTau in human AD brain, and efficiently engage pT181 in NHPs and human brain lysate. pT181+ tau in human plasma correlates with the neurofilament light in subjects with mild cognitive impairment (MCI)-suggesting the presence of pT181-Qß vaccine target in the early disease state.
Collapse
Affiliation(s)
- Nicole M. Maphis
- Department of Neuroscience1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Jonathan Hulse
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Julianne Peabody
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Somayeh Dadras
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Madelin J Whelpley
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Manas Kandath
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Colin Wilson
- Department of Radiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Sasha Hobson
- Center for Memory and Aging1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Jeff Thompson
- Center for Memory and Aging1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Suttinee Poolsup
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Danielle Beckman
- California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Sean P Ott
- California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Jennifer W. Watanabe
- California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Jodie L. Usachenko
- California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Koen K Van Rompay
- California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - John Morrison
- California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| | - Reed Selwyn
- Department of Radiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Gary Rosenberg
- Center for Memory and Aging1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Janice Knoefel
- Center for Memory and Aging1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Bryce Chackerian
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology1 University of New MexicoAlbuquerqueNew MexicoUSA
| |
Collapse
|
9
|
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
|
10
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Gallet S, Hannani D, Dergan-Dylon S, Vassal-Stermann E, Bally I, Chevillard C, Fenel D, Epaulard O, Poignard P, Fender P. Pre-Existing Anti-Vector Immunity to Adenovirus-Inspired VLP Vaccines Shows an Adjuvant-Dependent Antagonism. Vaccines (Basel) 2025; 13:238. [PMID: 40266128 PMCID: PMC11946745 DOI: 10.3390/vaccines13030238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: The use of virus-like particles (VLPs) in vaccinology has expanded significantly in recent years. VLPs have the advantage of being non-infectious while effectively stimulating B cell responses through the repetitive presentation of epitope motifs on their surface. Since VLPs are often derived from human-infecting viruses, preexisting immunity may influence the immune response they elicit, warranting further investigation. Methods: We have developed a 60-mer VLP derived from human adenovirus type 3, a common pathogen. We investigated the impact of pre-existing adenovirus immunity on the immunization outcome against the linear S14P5 epitope of SARS-CoV-2, which was engineered into the particle (Ad-VLP-S14P5). To this end, antibody responses to S14P5 were evaluated following immunization with Ad-VLP-S14P5 in either naive or vector-primed mice. Results: Mice with pre-existing anti-vector immunity exhibited significantly greater anti-S14P5 antibody responses compared to vector-naive animals, demonstrating a beneficial impact of prior anti-adenovirus responses. However, the addition of an oil-in-water adjuvant for the immunizations abolished this positive impact, even leading to a deleterious effect of the pre-existing anti-vector immunity. Conclusions: The data suggest that the immune status against immunizing VLPs must be taken into consideration when designing immunization protocols. Importantly, the effects of prior immunity may vary depending on the nature of the protocol, including factors such as adjuvant use.
Collapse
Affiliation(s)
- Salomé Gallet
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
- Clinical Infectious Disease Unit, Grenoble-Alpes University Hospital, F-38000 Grenoble, France
- Groupe de Recherche en Infectiologie Clinique, CIC 1406—Inserm—Université Grenoble Alpes, F-38000 Grenoble, France
| | - Dalil Hannani
- Université Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, F-38400 Grenoble, France
| | - Sebastian Dergan-Dylon
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| | - Emilie Vassal-Stermann
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| | - Isabelle Bally
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| | - Christopher Chevillard
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| | - Daphna Fenel
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| | - Olivier Epaulard
- Clinical Infectious Disease Unit, Grenoble-Alpes University Hospital, F-38000 Grenoble, France
- Groupe de Recherche en Infectiologie Clinique, CIC 1406—Inserm—Université Grenoble Alpes, F-38000 Grenoble, France
| | - Pascal Poignard
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| | - Pascal Fender
- CNRS, CEA, IBS, Université Grenoble Alpes, F-38000 Grenoble, France; (S.G.)
- Institut de Biologie Structurale, 71 rue des Martyrs, F-38042 Grenoble, France
| |
Collapse
|
12
|
Gallinaro A, Falce C, Pirillo MF, Borghi M, Grasso F, Canitano A, Cecchetti S, Baratella M, Michelini Z, Mariotti S, Chiantore MV, Farina I, Di Virgilio A, Tinari A, Scarlatti G, Negri D, Cara A. Simian Immunodeficiency Virus-Based Virus-like Particles Are an Efficient Tool to Induce Persistent Anti-SARS-CoV-2 Spike Neutralizing Antibodies and Specific T Cells in Mice. Vaccines (Basel) 2025; 13:216. [PMID: 40266067 PMCID: PMC11945333 DOI: 10.3390/vaccines13030216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Virus-like particles (VLPs) represent an attractive platform for delivering vaccine formulations, combining a high biosafety profile with a potent immune-stimulatory ability. VLPs are non-infectious, non-replicating, self-assembling nanostructures that can be exploited to efficiently expose membrane-tethered glycoproteins such as the SARS-CoV-2 Spike (S) protein, the main target of approved preventive vaccines. Here, we describe the development and preclinical validation of Simian Immunodeficiency Virus (SIV)-based GFP-labeled VLPs displaying S from the B.1.617.2 (Delta) variant (VLP/S-Delta) for inducing persistent anti-SARS-CoV-2 neutralizing antibodies (nAbs) and S-specific T cell responses in mice. Methods: SIV-derived VLP/S-Delta were produced by co-transfecting a plasmid expressing SIVGag-GFP, required for VLP assembly and quantification by flow virometry, a plasmid encoding the Delta S protein deleted in the cytoplasmic tail (CT), to improve membrane binding, and a VSV.G-expressing plasmid, to enhance VLP uptake. Recovered VLPs were titrated by flow virometry and characterized in vitro by transmission electron microscopy (TEM) and confocal microscopy (CLSM). BALB/c mice were immunized intramuscularly with VLP/S-Delta following a prime-boost regimen, and humoral and cellular immune responses were assessed. Results: VLP/S-Delta were efficiently pseudotyped with CT-truncated S-Delta. After BALB/c priming, VLP/S-Delta elicited both specific anti-RBD IgGs and anti-Delta nAbs that significantly increased after the boost and were maintained over time. The prime-boost vaccination induced similar levels of cross-nAbs against the ancestral Wuhan-Hu-1 strain as well as cross-nAbs against Omicron BA.1, BA.2 and BA.4/5 VoCs, albeit at lower levels. Moreover, immunization with VLP/S-Delta induced S-specific IFNγ-producing T cells. Conclusions: These data suggest that SIV-based VLPs are an appropriate delivery system for the elicitation of efficient and sustained humoral and cellular immunity in mice, paving the way for further improvements in the immunogen design to enhance the quality and breadth of immune responses against different viral glycoproteins.
Collapse
Affiliation(s)
- Alessandra Gallinaro
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Chiara Falce
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Maria Franca Pirillo
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Felicia Grasso
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Serena Cecchetti
- Confocal Microscopy Unit NMR, Confocal Microscopy Area Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Marco Baratella
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (M.B.); (G.S.)
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Sabrina Mariotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Maria Vincenza Chiantore
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Iole Farina
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Antonio Di Virgilio
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Antonella Tinari
- Center for Gender Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (M.B.); (G.S.)
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| |
Collapse
|
13
|
Zhou Y, Wei Y, Tian X, Wei X. Cancer vaccines: current status and future directions. J Hematol Oncol 2025; 18:18. [PMID: 39962549 PMCID: PMC11834487 DOI: 10.1186/s13045-025-01670-w] [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: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Cancer continues to be a major global health burden, with high morbidity and mortality. Building on the success of immune checkpoint inhibitors and adoptive cellular therapy, cancer vaccines have garnered significant interest, but their clinical success remains modest. Benefiting from advancements in technology, many meticulously designed cancer vaccines have shown promise, warranting further investigations to reach their full potential. Cancer vaccines hold unique benefits, particularly for patients resistant to other therapies, and they offer the ability to initiate broad and durable T cell responses. In this review, we highlight the antigen selection for cancer vaccines, introduce the immune responses induced by vaccines, and propose strategies to enhance vaccine immunogenicity. Furthermore, we summarize key features and notable clinical advances of various vaccine platforms. Lastly, we delve into the mechanisms of tumor resistance and explore the potential benefits of combining cancer vaccines with standard treatments and other immunomodulatory approaches to improve vaccine efficacy.
Collapse
Affiliation(s)
- Yingqiong Zhou
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
14
|
Sun B, Li R, Ji N, Liu H, Wang H, Chen C, Bai L, Su J, Chen J. Brain-targeting drug delivery systems: The state of the art in treatment of glioblastoma. Mater Today Bio 2025; 30:101443. [PMID: 39866779 PMCID: PMC11759563 DOI: 10.1016/j.mtbio.2025.101443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant brain tumor, characterized by a high mortality rate and a poor prognosis. The blood-brain barrier (BBB) and the blood-tumor barrier (BTB) present significant obstacles to the efficacy of tumor-targeted pharmacotherapy, thereby impeding the therapeutic potential of numerous candidate drugs. Targeting delivery of adequate doses of drug across the BBB to treat GBM has become a prominent research area in recent years. This emphasis has driven the exploration and evaluation of diverse technologies for GBM pharmacotherapy, with some already undergoing clinical trials. This review provides a thorough overview of recent advancements and challenges in targeted drug delivery for GBM treatment. It specifically emphasizes systemic drug administration strategies to assess their potential and limitations in GBM treatment. Furthermore, this review highlights promising future research directions in the development of intelligent drug delivery systems aimed at overcoming current challenges and enhancing therapeutic efficacy against GBM. These advancements not only support foundational research on targeted drug delivery systems for GBM but also offer methodological approaches for future clinical applications.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Rong Li
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chao Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
15
|
Ruzzi F, Riccardo F, Conti L, Tarone L, Semprini MS, Bolli E, Barutello G, Quaglino E, Lollini PL, Cavallo F. Cancer vaccines: Target antigens, vaccine platforms and preclinical models. Mol Aspects Med 2025; 101:101324. [PMID: 39631227 DOI: 10.1016/j.mam.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
This review provides a comprehensive overview of the evolving landscape of cancer vaccines, highlighting their potential to revolutionize tumor prevention. Building on the success of vaccines against virus-related cancers, such as HPV- and HBV-associated cervical and liver cancers, the current challenge is to extend these achievements to the prevention of non-viral tumors and the treatment of preneoplastic or early neoplastic lesions. This review analyzes the critical aspects of preventive anti-cancer vaccination, focusing on the choice of target antigens, the development of effective vaccine platforms and technologies, and the use of various model systems for preclinical testing, from laboratory rodents to companion animals.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Federica Riccardo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Laura Conti
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Lidia Tarone
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Elisabetta Bolli
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Giuseppina Barutello
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Elena Quaglino
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy; IRCCS Azienda Ospedaliera Universitaria di Bologna, 40138, Bologna, Italy.
| | - Federica Cavallo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy.
| |
Collapse
|
16
|
Pham JA, Coronel MM. Unlocking Transplant Tolerance with Biomaterials. Adv Healthc Mater 2025; 14:e2400965. [PMID: 38843866 PMCID: PMC11834385 DOI: 10.1002/adhm.202400965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/31/2024] [Indexed: 07/04/2024]
Abstract
For patients suffering from organ failure due to injury or autoimmune disease, allogeneic organ transplantation with chronic immunosuppression is considered the god standard in terms of clinical treatment. However, the true "holy grail" of transplant immunology is operational tolerance, in which the recipient exhibits a sustained lack of alloreactivity toward unencountered antigen presented by the donor graft. This outcome is resultant from critical changes to the phenotype and genotype of the immune repertoire predicated by the activation of specific signaling pathways responsive to soluble and mechanosensitive cues. Biomaterials have emerged as a medium for interfacing with and reprogramming these endogenous pathways toward tolerance in precise, minimally invasive, and spatiotemporally defined manners. By viewing seminal and contemporary breakthroughs in transplant tolerance induction through the lens of biomaterials-mediated immunomodulation strategies-which include intrinsic material immunogenicity, the depot effect, graft coatings, induction and delivery of tolerogenic immune cells, biomimicry of tolerogenic immune cells, and in situ reprogramming-this review emphasizes the stunning diversity of approaches in the field and spotlights exciting future directions for research to come.
Collapse
Affiliation(s)
- John‐Paul A. Pham
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Elizabeth Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - María M. Coronel
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Elizabeth Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
17
|
Adeleke RA, Sahler J, Choi A, Roth K, Upadhye V, Ezzatpour S, Imbiakha B, Khomandiak S, Diaz A, Whittaker GR, Jager MC, August A, Buchholz DW, Aguilar HC. Replication-incompetent VSV-based vaccine elicits protective responses against SARS-CoV-2 and influenza virus. SCIENCE ADVANCES 2025; 11:eadq4545. [PMID: 39879304 PMCID: PMC11777205 DOI: 10.1126/sciadv.adq4545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses lead to severe respiratory illnesses and death in humans, exacerbated in individuals with underlying health conditions, remaining substantial global public health concerns. Here, we developed a bivalent replication-incompetent single-cycle pseudotyped vesicular stomatitis virus vaccine that incorporates both a prefusion-stabilized SARS-CoV-2 spike protein lacking a furin cleavage site and a full-length influenza A virus neuraminidase protein. Vaccination of K18-hACE2 or C57BL/6J mouse models generated durable levels of neutralizing antibodies, T cell responses, and protection from morbidity and mortality upon challenge with either virus. Furthermore, the vaccine provided heterologous protection upon challenge with a different influenza virus strain, supporting the advantage of using NA to increase the breadth of vaccine protection. Now, no bivalent vaccine is approved for use against both SARS-CoV-2 and influenza virus. Our study supports using this platform to develop safe and efficient vaccines against multiple viruses.
Collapse
Affiliation(s)
- Richard A. Adeleke
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Julie Sahler
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Kyle Roth
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Viraj Upadhye
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Brian Imbiakha
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Solomiia Khomandiak
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Annika Diaz
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Mason C. Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - David W. Buchholz
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| |
Collapse
|
18
|
Peralta-Cuevas E, Garcia-Atutxa I, Huerta-Saquero A, Villanueva-Flores F. The Role of Plant Virus-like Particles in Advanced Drug Delivery and Vaccine Development: Structural Attributes and Application Potential. Viruses 2025; 17:148. [PMID: 40006903 PMCID: PMC11861432 DOI: 10.3390/v17020148] [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: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 02/27/2025] Open
Abstract
Plant virus-like particles (pVLPs) present distinct research advantages, including cost-effective production and scalability through plant-based systems, making them a promising yet underutilized alternative to traditional VLPs. Human exposure to plant viruses through diet for millions of years supports their biocompatibility and safety, making them suitable for biomedical applications. This review offers a practical guide to selecting pVLPs based on critical design factors. It begins by examining how pVLP size and shape influence cellular interactions, such as uptake, biodistribution, and clearance, key for effective drug delivery and vaccine development. We also explore how surface charge affects VLP-cell interactions, impacting binding and internalization, and discuss the benefits of surface modifications to enhance targeting and stability. Additional considerations include host range and biosafety, ensuring safe, effective pVLP applications in clinical and environmental contexts. The scalability of pVLP production across different expression systems is also reviewed, noting challenges and opportunities in large-scale manufacturing. Concluding with future perspectives, the review highlights the innovation potential of pVLPs in vaccine development, targeted therapies, and diagnostics, positioning them as valuable tools in biotechnology and medicine. This guide provides a foundation for selecting optimal pVLPs across diverse applications.
Collapse
Affiliation(s)
- Esperanza Peralta-Cuevas
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA), Unidad Morelos del Instituto Politécnico Nacional (IPN), Boulevard de la Tecnología No. 1036, Xochitepec 62790, Mexico;
| | - Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Av. de los Jerónimos, 135, 30107 Murcia, Spain;
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Km. 107 Carretera Tijuana-Ensenada, Ensenada 22860, Mexico;
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA), Unidad Morelos del Instituto Politécnico Nacional (IPN), Boulevard de la Tecnología No. 1036, Xochitepec 62790, Mexico;
| |
Collapse
|
19
|
Sergeeva OV, Luo L, Guiseppi-Elie A. Cancer theragnostics: closing the loop for advanced personalized cancer treatment through the platform integration of therapeutics and diagnostics. Front Bioeng Biotechnol 2025; 12:1499474. [PMID: 39898278 PMCID: PMC11782185 DOI: 10.3389/fbioe.2024.1499474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Cancer continues to be one of the leading causes of death worldwide, and conventional cancer therapies such as chemotherapy, radiation therapy, and surgery have limitations. RNA therapy and cancer vaccines hold considerable promise as an alternative to conventional therapies for their ability to enable personalized therapy with improved efficacy and reduced side effects. The principal approach of cancer vaccines is to induce a specific immune response against cancer cells. However, a major challenge in cancer immunotherapy is to predict which patients will respond to treatment and to monitor the efficacy of the vaccine during treatment. Theragnostics, an integration of diagnostic and therapeutic capabilities into a single hybrid platform system, has the potential to address these challenges by enabling real-time monitoring of treatment response while allowing endogenously controlled personalized treatment adjustments. In this article, we review the current state-of-the-art in theragnostics for cancer vaccines and RNA therapy, including imaging agents, biomarkers, and other diagnostic tools relevant to cancer, and their application in cancer therapy development and personalization. We also discuss the opportunities and challenges for further development and clinical translation of theragnostics in cancer vaccines.
Collapse
Affiliation(s)
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Anthony Guiseppi-Elie
- Bioelectronics, Biosensors and Biochips (C3B), Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Full Affiliate Member, Houston Methodist Research Institute, Houston, TX, United States
- ABTECH Scientific, Inc., Biotechnology Research Park, Richmond, VA, United States
| |
Collapse
|
20
|
Zhang M, Wang C, Pan J, Cui H, Zhao X. Advancing novel veterinary vaccines: From comprehensive antigen and adjuvant design to preparation process optimization. Int Immunopharmacol 2025; 145:113784. [PMID: 39672026 DOI: 10.1016/j.intimp.2024.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
Vaccination stands as the paramount and cost-effective strategy for the prevention and management of animal infectious diseases. With the advances in biological technology, materials science and industrial optimization, substantial progress has been made in the development of innovative veterinary vaccines. A majority of the novel vaccines under current investigation tend to stimulate multiple immune pathways and to achieve long-term resistance against infectious diseases, yet it remains imperative to concentrate research efforts on the efficient utilization of vaccines, mitigating toxic side effects, and ensuring safe production processes. This article presents an overview of research progress in veterinary vaccines, encompassing comprehensive antigen design, adjuvant formulation advancements, preparation process optimization, and rigorous immune efficacy evaluation. It summarizes cutting-edge vaccines derived from in vitro synthesis and in vivo application, emphasizing immunogenic components and immune response mechanisms. It also highlights novel biological adjuvants that enhance immune efficacy, diversify raw materials, and possess targeted functions, while comprehensively exploring advancements in production methodologies and compatible vaccine products. By establishing a foundation for the integrated use of these innovative veterinary vaccines, this work facilitates future interdisciplinary cooperation in their advancement, aiming to accelerate the achievement of herd immunity through concerted efforts.
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chunxin Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Junqian Pan
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haixin Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
21
|
Liang J, Yao L, Liu Z, Chen Y, Lin Y, Tian T. Nanoparticles in Subunit Vaccines: Immunological Foundations, Categories, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407649. [PMID: 39501996 DOI: 10.1002/smll.202407649] [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: 08/28/2024] [Revised: 10/12/2024] [Indexed: 01/11/2025]
Abstract
Subunit vaccines, significant in next-generation vaccine development, offer precise targeting of immune responses by focusing on specific antigens. However, this precision often comes at the cost of eliciting strong and durable immunity, posing a great challenge to vaccine design. To address this limitation, recent advancements in nanoparticles (NPs) are utilized to enhance antigen delivery efficiency and boost vaccine efficacy. This review examines how the physicochemical properties of NPs influence various stages of the immune response during vaccine delivery and analyzes how different NP types contribute to immune activation and enhance vaccine performance. It then explores the unique characteristics and immune activation mechanisms of these NPs, along with their recent advancements, and highlights their application in subunit vaccines targeting infectious diseases and cancer. Finally, it discusses the challenges in NP-based vaccine development and proposes future directions for innovation in this promising field.
Collapse
Affiliation(s)
- Jiale Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Taoran Tian
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
22
|
Grant M, Ni Lee L, Chinnakannan S, Tong O, Kwok J, Cianci N, Tillman L, Saha A, Pereira Almeida V, Leung C. Unlocking cancer vaccine potential: What are the key factors? Hum Vaccin Immunother 2024; 20:2331486. [PMID: 38564321 PMCID: PMC11657071 DOI: 10.1080/21645515.2024.2331486] [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: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer is a global health challenge, with changing demographics and lifestyle factors producing an increasing burden worldwide. Screening advancements are enabling earlier diagnoses, but current cancer immunotherapies only induce remission in a small proportion of patients and come at a high cost. Cancer vaccines may offer a solution to these challenges, but they have been mired by poor results in past decades. Greater understanding of tumor biology, coupled with the success of vaccine technologies during the COVID-19 pandemic, has reinvigorated cancer vaccine development. With the first signs of efficacy being reported, cancer vaccines may be beginning to fulfill their potential. Solid tumors, however, present different hurdles than infectious diseases. Combining insights from previous cancer vaccine clinical development and contemporary knowledge of tumor immunology, we ask: who are the 'right' patients, what are the 'right' targets, and which are the 'right' modalities to maximize the chances of cancer vaccine success?
Collapse
|
23
|
Waugh S, Cameron CE. Syphilis vaccine development: Aligning vaccine design with manufacturing requirements. Hum Vaccin Immunother 2024; 20:2399915. [PMID: 39262177 PMCID: PMC11404580 DOI: 10.1080/21645515.2024.2399915] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Syphilis, caused by Treponema pallidum subsp. pallidum, is a global health concern with increasing rates worldwide. Current prevention strategies, including screen-and-treat approaches, are not sufficient to resolve rising infection rates, emphasizing the need for a vaccine. Developing a syphilis vaccine necessitates a range of cross-disciplinary considerations, including essential disease-specific protection, technical requirements, economic feasibility, manufacturing constraints, public acceptance, equitable vaccine access, alignment with global public vaccination programs, and identification of essential populations to be vaccinated to achieve herd immunity. Central to syphilis vaccine development is prioritization of global vaccine availability, including access in low- to middle-income settings. Various vaccine platforms, including subunit, virus-like particle (VLP), mRNA, and outer membrane vesicle (OMV) vaccines, present both advantages and challenges. The proactive consideration of both manufacturing feasibility and efficacy throughout the pre-clinical research and development stages is essential for producing an efficacious, inexpensive, and scalable syphilis vaccine to address the growing global health burden caused by this disease.
Collapse
Affiliation(s)
- Sean Waugh
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Caroline E. Cameron
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Department of Medicine, Division of Allergy and Infectious Disease, University of Washington, Seattle, WA, USA
| |
Collapse
|
24
|
Huang S, Que H, Wang M, Wei X. mRNA vaccines as cancer therapies. Chin Med J (Engl) 2024; 137:2979-2995. [PMID: 39668413 PMCID: PMC11706586 DOI: 10.1097/cm9.0000000000003455] [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: 10/14/2024] [Indexed: 12/14/2024] Open
Abstract
ABSTRACT Cancer remains a major global health challenge, with conventional treatments like chemotherapy and radiotherapy often hindered by significant side effects, lack of specificity, and limited efficacy in advanced cases. Among emerging therapeutic strategies, mRNA vaccines have shown remarkable potential due to their adaptability, rapid production, and capability for personalized cancer treatment. This review provides an in-depth analysis of messenger RNA (mRNA) vaccines as a therapeutic approach for cancer immunotherapy, focusing on their molecular biology, classification, mechanisms, and clinical studies. Derived from reported literature and data on clinicaltrials.gov, it examines studies on mRNA vaccines encoding tumor-specific antigens (TSAs), tumor-associated antigens (TAAs), immunomodulators, and chimeric antigen receptors (CARs) across various cancer types. The review highlights the ability of mRNA vaccines to encode TSAs and TAAs, enabling personalized cancer treatments, and classifies these vaccines into non-replicating and self-amplifying types. It further explores their mechanisms of action, including antigen presentation and immune activation, while emphasizing findings from clinical studies that demonstrate the potential of mRNA vaccines in cancer therapy. Despite their promise, challenges remain in enhancing delivery systems, improving immunogenicity, and addressing tumor heterogeneity. Overcoming these obstacles will require further investigation to fully harness the potential of mRNA vaccines in personalized cancer treatment.
Collapse
Affiliation(s)
- Shaoxiong Huang
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
25
|
Sun X, Tian T, Lian Y, Cui Z. Current Advances in Viral Nanoparticles for Biomedicine. ACS NANO 2024; 18:33827-33863. [PMID: 39648920 DOI: 10.1021/acsnano.4c13146] [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: 12/10/2024]
Abstract
Viral nanoparticles (VNPs) have emerged as crucial tools in the field of biomedicine. Leveraging their biological and physicochemical properties, VNPs exhibit significant advantages in the prevention, diagnosis, and treatment of human diseases. Through techniques such as chemical bioconjugation, infusion, genetic engineering, and encapsulation, these VNPs have been endowed with multifunctional capabilities, including the display of functional peptides or proteins, encapsulation of therapeutic drugs or inorganic particles, integration with imaging agents, and conjugation with bioactive molecules. This review provides an in-depth analysis of VNPs in biomedicine, elucidating their diverse types, distinctive features, production methods, and complex design principles behind multifunctional VNPs. It highlights recent innovative research and various applications, covering their roles in imaging, drug delivery, therapeutics, gene delivery, vaccines, immunotherapy, and tissue regeneration. Additionally, the review provides an assessment of their safety and biocompatibility and discusses challenges and future opportunities in the field, underscoring the vast potential and evolving nature of VNP research.
Collapse
Affiliation(s)
- Xianxun Sun
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Tao Tian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yindong Lian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
26
|
Meng X, Yan F, Wang W, Wang S, Cong H, Li J, Zhao Y, Wang T, Li N, Gao Y, Wang J, Feng N, Xia X. A single dose of an ALVAC vector-based RABV virus-like particle candidate vaccine induces a potent immune response in mice, cats and dogs. Emerg Microbes Infect 2024; 13:2406280. [PMID: 39295522 PMCID: PMC11443554 DOI: 10.1080/22221751.2024.2406280] [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: 05/13/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 09/21/2024]
Abstract
Rabies, caused by the Rabies virus (RABV), is a highly fatal zoonotic disease. Existing rabies vaccines have demonstrated good immune efficacy, but the complexity of immunization procedures and high cost has impeded the elimination of RABV, particularly in the post-COVID-19 era. There is a pressing need for safer and more effective rabies vaccines that streamline vaccination protocols and reduce expense. To meet this need, we have developed a potential rabies vaccine candidate called ALVAC-RABV-VLP, utilizing CRISPR/Cas9 gene editing technology. This vaccine employs a canarypox virus vector (ALVAC) to generate RABV virus-like particles (VLPs). In mice, a single dose of ALVAC-RABV-VLP effectively activated dendritic cells (DCs), follicular helper T cells (Tfh), and the germinal centre (GC)/plasma cell axis, resulting in durable and effective humoral immune responses. The survival rate of mice challenged with lethal RABV was 100%. Similarly, in dogs and cats, a single immunization with ALVAC-RABV-VLP elicited a stronger and longer-lasting antibody response. ALVAC-RABV-VLP induced superior cellular and humoral immunity in both mice and beagles compared to the commercial inactivated rabies vaccine. In conclusion, ALVAC-RABV-VLP induced robust protective immune responses in mice, dogs and cats, offering a novel, cost-effective, efficient, and promising approach for herd prevention of rabies.
Collapse
Affiliation(s)
- Xianyong Meng
- College of Veterinary Medicine, Jilin agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Haiyang Cong
- College of Veterinary Medicine, Jilin agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Jiaqi Li
- College of Veterinary Medicine, Jilin agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin agricultural University, Changchun, People’s Republic of China
| | - Na Feng
- College of Veterinary Medicine, Jilin agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
27
|
Griffo A. Evolving virus-like particles. Nat Biomed Eng 2024; 8:1512. [PMID: 39681706 DOI: 10.1038/s41551-024-01330-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
|
28
|
Li HX, Gong YW, Yan PJ, Xu Y, Qin G, Wen WP, Teng FY. Revolutionizing head and neck squamous cell carcinoma treatment with nanomedicine in the era of immunotherapy. Front Immunol 2024; 15:1453753. [PMID: 39676875 PMCID: PMC11638222 DOI: 10.3389/fimmu.2024.1453753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a prevalent malignant tumor globally. Despite advancements in treatment methods, the overall survival rate remains low due to limitations such as poor targeting and low bioavailability, which result in the limited efficacy of traditional drug therapies. Nanomedicine is considered to be a promising strategy in tumor therapy, offering the potential for maximal anti-tumor effects. Nanocarriers can overcome biological barriers, enhance drug delivery efficiency to targeted sites, and minimize damage to normal tissues. Currently, various nano-carriers for drug delivery have been developed to construct new nanomedicine. This review aims to provide an overview of the current status of HNSCC treatment and the necessity of nanomedicine in improving treatment outcomes. Moreover, it delves into the research progress of nanomedicine in HNSCC treatment, with a focus on enhancing radiation sensitivity, improving the efficacy of tumor immunotherapy, effectively delivering chemotherapy drugs, and utilizing small molecule inhibitors. Finally, this article discussed the challenges and prospects of applying nanomedicine in cancer treatment.
Collapse
Affiliation(s)
- Hong-Xia Li
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Yu-Wen Gong
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pi-Jun Yan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Yong Xu
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei-Ping Wen
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fang-Yuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| |
Collapse
|
29
|
Huang S, He Y, Madow A, Peng H, Griffin M, Qi J, Huang M, Amoroso H, Abrashoff R, Heldman N, Belcher AM. Reprogramming the genome of M13 bacteriophage for all-in-one personalized cancer vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624916. [PMID: 39605543 PMCID: PMC11601592 DOI: 10.1101/2024.11.22.624916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Peptide-based vaccines face limitations in immunogenicity and stability, and challenges in co-delivering antigens and adjuvants effectively. Virus-based nanoparticles, particularly M13 bacteriophage, present a promising solution due to their genetic modifiability, intrinsic adjuvanticity, and efficient antigen presentation capabilities. Here we developed a programmable M13 phage-based personalized cancer vaccine enabling single-step antigen-adjuvant assembly. Specifically, we designed a reprogrammed (RP) phage platform that precisely regulates Toll-like receptor 9 activation by programming its genome sequence and modulates antigen density through genetic engineering. Vaccination studies with RP phages demonstrated that the immune response could be modulated by fine-tuning the adjuvanticity and antigen density, revealing an optimal antigen dose and adjuvanticity for maximum vaccine efficacy. The RP phage induced a remarkable 24-fold increase in neoantigen-specific CD8 + T cells and eradicated established MC-38 tumors when combined with anti-PD-1 therapy. These findings highlight the RP phage's potential as a powerful nanovaccine platform for personalized cancer vaccines.
Collapse
|
30
|
Li J, Ding J, Guo C, Xu X, Shan C, Qian J, Ding Z. Development of a Novel Chimeric ND-GP cVLPs Vaccine for the Prevention of Goose-Derived Newcastle Disease and Gosling Plague. Microorganisms 2024; 12:2266. [PMID: 39597655 PMCID: PMC11596917 DOI: 10.3390/microorganisms12112266] [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: 09/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Goose-derived Newcastle disease (ND) and gosling plague (GP) are serious threats to the goose industry. Conventional vaccines have made significant contributions to preventing GP and ND. Nevertheless, the renewal of conventional vaccines and the application of novel vaccines are urgently needed to align with eco-friendly and efficient breeding concepts and achieve the final goal of epidemic purification. Therefore, based on the Newcastle disease virus-like particles (ND VLPs) vector platform, we developed novel chimeric ND-GP bivalent cVLPs (ND-GP cVLPs) displaying the NDV HN protein and the GPV VP3 protein. In vivo, immunization experiments revealed that geese immunized with 30 µg, 50 µg, or 70 µg of the ND-GP cVLPs and commercial vaccines produced highly effective hemagglutination inhibitory antibodies against NDV and neutralizing antibodies against GPV, respectively. Furthermore, 70 µg of the ND-GP cVLPs effectively protected against virulent NDV and GPV, reducing tissue damage from viral infection and virus shedding in the oropharynx and cloaca. In conclusion, we provide eco-friendly and efficient novel ND-GP cVLPs for preventing goose-derived ND and GP. Our findings provide the basis for using ND VLPs as foreign protein carriers for the developing of multi-conjugate vaccines.
Collapse
Affiliation(s)
- Jindou Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (J.L.); (J.D.); (C.G.); (X.X.); (C.S.)
| | - Jiaxin Ding
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (J.L.); (J.D.); (C.G.); (X.X.); (C.S.)
| | - Chunhong Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (J.L.); (J.D.); (C.G.); (X.X.); (C.S.)
| | - Xiaohong Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (J.L.); (J.D.); (C.G.); (X.X.); (C.S.)
| | - Chunhui Shan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (J.L.); (J.D.); (C.G.); (X.X.); (C.S.)
| | - Jing Qian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Zhuang Ding
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (J.L.); (J.D.); (C.G.); (X.X.); (C.S.)
| |
Collapse
|
31
|
Aragão Tejo Dias V, Oliveira Guardalini LG, Leme J, Consoni Bernardino T, da Silveira SR, Tonso A, Attie Calil Jorge S, Fernández Núñez EG. Different modeling approaches for inline biochemical monitoring over the VLP-making upstream stages using Raman spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 320:124638. [PMID: 38880076 DOI: 10.1016/j.saa.2024.124638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/22/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024]
Abstract
This work aimed to set inline Raman spectroscopy models to monitor biochemically (viable cell density, cell viability, glucose, lactate, glutamine, glutamate, and ammonium) all upstream stages of a virus-like particle-making process. Linear (Partial least squares, PLS; Principal components regression, PCR) and nonlinear (Artificial neural networks, ANN; supported vector machine, SVM) modeling approaches were assessed. The nonlinear models, ANN and SVM, were the more suitable models with the lowest absolute errors. The mean absolute error of the best models within the assessed parameter ranges for viable cell density (0.01-8.83 × 106 cells/mL), cell viability (1.3-100.0 %), glucose (5.22-10.93 g/L), lactate (18.6-152.7 mg/L), glutamine (158-1761 mg/L), glutamate (807.6-2159.7 mg/L), and ammonium (62.8-117.8 mg/L) were 1.55 ± 1.37 × 106 cells/mL (ANN), 5.01 ± 4.93 % (ANN), 0.27 ± 0.22 g/L (SVM), 4.7 ± 2.6 mg/L (SVM), 51 ± 49 mg/L (ANN), 57 ± 39 mg/L (SVM) and 2.0 ± 1.8 mg/L (ANN), respectively. The errors achieved, and best-fitted models were like those for the same bioprocess using offline data and others, which utilized inline spectra for mammalian cell lines as a host.
Collapse
Affiliation(s)
- Vinícius Aragão Tejo Dias
- Laboratório de Engenharia de Bioprocessos, Escola de Artes, Ciências e Humanidades (EACH), Universidade de São Paulo, Rua Arlindo Béttio, 1000, CEP 03828-000, São Paulo, SP, Brazil
| | | | - Jaci Leme
- Laboratório de Biotecnologia Viral, Instituto Butantan, Av Vital Brasil 1500, CEP 05503-900 São Paulo, SP, Brazil
| | - Thaissa Consoni Bernardino
- Laboratório de Biotecnologia Viral, Instituto Butantan, Av Vital Brasil 1500, CEP 05503-900 São Paulo, SP, Brazil
| | | | - Aldo Tonso
- Laboratório de Células Animais, Departamento de Engenharia Química, Escola Politécnica, Universidade de São Paulo, Av. Prof. Luciano Gualberto, travessa do Politécnico, 380, 05508-010 São Paulo, SP, Brazil
| | - Soraia Attie Calil Jorge
- Laboratório de Biotecnologia Viral, Instituto Butantan, Av Vital Brasil 1500, CEP 05503-900 São Paulo, SP, Brazil
| | - Eutimio Gustavo Fernández Núñez
- Laboratório de Engenharia de Bioprocessos, Escola de Artes, Ciências e Humanidades (EACH), Universidade de São Paulo, Rua Arlindo Béttio, 1000, CEP 03828-000, São Paulo, SP, Brazil.
| |
Collapse
|
32
|
Sun X, Lian Y, Tian T, Cui Z. Virus-like particle encapsulation of functional proteins: advances and applications. Theranostics 2024; 14:7604-7622. [PMID: 39659581 PMCID: PMC11626933 DOI: 10.7150/thno.103127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024] Open
Abstract
Proteins face several challenges in biomedicine, including issues with antibody production, degradation by proteases, rapid clearance by the kidneys, and short half-lives. To address these problems, various nano delivery systems have been developed, with virus-like particles (VLPs) emerging as a leading solution. VLPs, which are self-assembled protein complexes, offer effective encapsulation and transport of proteins. They provide enhanced stability, extended circulation time, preserved biological activity, improved targeting for therapies or imaging, and reduced side effects due to minimized systemic exposure. This review explores various methods for encapsulating proteins within VLPs. It assesses the benefits and limitations of each method and their applications in imaging, therapeutic enzyme delivery, vaccines, immunotherapy, nanoreactors, and biosensors. Future advancements in VLPs will depend on improving packaging methods, controlling protein loading, optimizing assembly techniques, and enhancing capsid design. The review also discusses current challenges and proposes solutions to advance the use of VLPs in various applications.
Collapse
Affiliation(s)
- Xianxun Sun
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Yindong Lian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Tao Tian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
33
|
Yan W, Huang S, Zhang L, Yang Q, Liu S, Wang Z, Chu Q, Tian M, Zhao L, Sun Y, Lei C, Wang H, Yang X. Virus-like Particles vaccine based on co-expression of G5 Porcine rotavirus VP2-VP6-VP7 induces a powerful immune protective response in mice. Vet Microbiol 2024; 298:110241. [PMID: 39226763 DOI: 10.1016/j.vetmic.2024.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Porcine rotavirus (PoRV), a member of the Reoviridae family, constitutes a principal etiological agent of acute diarrhea in piglets younger than eight weeks of age, and it is associated with considerable morbidity and mortality within the swine industry. The G5 genotype rotavirus strain currently predominates in circulation. To develop a safe and effective porcine rotavirus vaccine, we generated an insect cell-baculovirus expression system, and successfully expressed these three viral proteins and assembled them into virus-like particles (VLPs) co-displaying VP2, VP6, and VP7. Transmission electron microscopy (TEM) analysis revealed that the VP2-VP6-VP7 VLPs exhibited a "wheeled" morphology resembling that of native rotavirus particles, with an estimated diameter of approximately 65 nm. To evaluate the immunogenicity and protective efficacy of these VP2-VP6-VP7 VLPs, we immunized BALB/C mice with four escalating doses of the VLPs, ranging from 5 to 40 μg of VLP protein per dose. ELISA-based assessments of PoRV-specific antibodies and T cell cytokines, including IL-4, IL-2, and IFN-γ, demonstrate that immunization with VP2-VP6-VP7 VLPs can effectively elicit both humoral and cellular immune responses in mice, resulting in a notable induction of neutralizing antibodies. On days 4, 6, 8, and 10 post-infection (dpi), the VLP-vaccinated group exhibited significantly reduced levels of PoRV RNA copy numbers when compared to the PBS controls. Histological examination of the duodenum, ileum, and kidneys revealed that VP2-VP6-VP7 VLPs provided effective protection against PoRV induced intestinal injury. Collectively, these findings indicate that the VLPs generated in this study possess strong immunogenicity and suggest the considerable promise of the VLP-based vaccine candidate in the prevention and containment of Porcine Rotavirus infections.
Collapse
Affiliation(s)
- Wenjun Yan
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Siyu Huang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China; Sichuan Animal Science Academy (SASA), Chengdu 610066, China
| | - Lan Zhang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Qingcheng Yang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Song Liu
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Zheng Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Qinyuan Chu
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Mingyue Tian
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Lijun Zhao
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yue Sun
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Changwei Lei
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hongning Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Xin Yang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science. Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| |
Collapse
|
34
|
Yu Q, Zhu Q, Huang X, Wu J, Zhou Q, Chen T, Zhu C, Ding L, Deng G, Wang Y, Zhang Z, Zhang B. Preparation of bovine coronavirus virus-like particles and its immunogenicity in mice and cattle. Microb Pathog 2024; 197:107062. [PMID: 39442811 DOI: 10.1016/j.micpath.2024.107062] [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: 08/21/2024] [Revised: 10/07/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
The widespread prevalence of bovine coronavirus (BCoV) disease worldwide has impacted the livestock industry economically. No effective vaccine is available in China. In this study, we produced BCoV virus-like particles (VLPs) containing E, M, N, S, and hemagglutinin-esterase (HE) proteins using a baculovirus expression system. Five recombinant baculoviruses were co-infected with Sf9 cells, and the VLPs were assembled and characterized. Mice and cattle were immunized by VLPs mixed with MF59 and CpG 55.2 adjuvants. Two doses of the VLPs/MF59/CpG vaccine were administered in mice and cattle. The immune effect of the VLPs/MF59/CpG vaccine was measured using indirect ELISA and neutralization assays. After immunization, the serum IgG-specific antibody titer against S protein and neutralization antibody titer increased to 1:1.28 × 104 (p < 0.01) and 1:128 (p < 0.01) in mice, respectively. Interestingly, the high IgG antibody and neutralizing antibody titers were maintained for seven days in mice. In addition, the serum IgG-specific antibody titer against S proteins and neutralization antibody titer increased to 1:1.024 × 105 and 1:512 (p < 0.05) in cattle, respectively. The high IgG antibody and neutralizing antibody titers were maintained for 21 days in cattle. Notably, BCoV VLPs group interferon-gamma (IFN-γ) lymphocytes in spleens were significantly increased (p < 0.01). These findings suggest that BCoV VLPs induced strong cellular and humoral immune responses in mice and cattle. These findings suggest that BCoV VLPs could serve as a potent immunogen for vaccine development.
Collapse
Affiliation(s)
- Qisheng Yu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Qing Zhu
- Center for Animal Disease Control and Prevention, Ganzi Tibetan Autonomous Prefecture, Kangding, 626000, China.
| | - Xiangyue Huang
- Animal Husbandry Science Institute of Aba Autonomous Prefecture, Hongyuan, 624400, China.
| | - Jinbo Wu
- Animal Husbandry Science Institute of Aba Autonomous Prefecture, Hongyuan, 624400, China.
| | - Qun Zhou
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Taoyun Chen
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Chenxi Zhu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Lu Ding
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Gunan Deng
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Yi Wang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
| | - Zhaohui Zhang
- Center for Animal Disease Control and Prevention, Ganzi Tibetan Autonomous Prefecture, Kangding, 626000, China.
| | - Bin Zhang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China.
| |
Collapse
|
35
|
Li YZ, Ji RR. Gene therapy for chronic pain management. Cell Rep Med 2024; 5:101756. [PMID: 39366385 PMCID: PMC11513853 DOI: 10.1016/j.xcrm.2024.101756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/20/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Despite significant advances in identifying molecular targets for chronic pain over the past two decades, many remain difficult to target with traditional methods. Gene therapies such as antisense oligonucleotides (ASOs), RNA interference (RNAi), CRISPR, and virus-based delivery systems have played crucial roles in discovering and validating new pain targets. While there has been a surge in gene therapy-based clinical trials, those focusing on pain as the primary outcome remain uncommon. This review examines various gene therapy strategies, including ASOs, small interfering RNA (siRNAs), optogenetics, chemogenetics, and CRISPR, and their delivery methods targeting primary sensory neurons and non-neuronal cells, including glia and chondrocytes. We also explore emerging gene therapy tools and highlight gene therapy's clinical potential in pain management, including trials targeting pain-related diseases. Advances in single-cell analysis of sensory neurons and non-neuronal cells, along with the development of new delivery tools, are poised to accelerate the application of gene therapy in pain medicine.
Collapse
Affiliation(s)
- Yi-Ze Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
36
|
Tian T, Zhu Y, Shi J, Shang K, Yin Z, Shi H, He Y, Ding J, Zhang F. The development of a human Brucella mucosal vaccine: What should be considered? Life Sci 2024; 355:122986. [PMID: 39151885 DOI: 10.1016/j.lfs.2024.122986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Brucellosis is a chronic infectious disease that is zoonotic in nature. Brucella can infect humans through interactions with livestock, primarily via the digestive tract, respiratory tract, and oral cavity. This bacterium has the potential to be utilized as a biological weapon and is classified as a Category B pathogen by the Centers for Disease Control and Prevention. Currently, there is no approved vaccine for humans against Brucella, highlighting an urgent need for the development of a vaccine to mitigate the risks posed by this pathogen. Brucella primarily infects its host by adhering to and penetrating mucosal surfaces. Mucosal immunity plays a vital role in preventing local infections, clearing microorganisms from mucosal surfaces, and inhibiting the spread of pathogens. As mucosal vaccine strategies continue to evolve, the development of a safe and effective mucosal vaccine against Brucella appears promising.This paper reviews the immune mechanism of mucosal vaccines, the infection mechanism of Brucella, successful Brucella mucosal vaccines in animals, and mucosal adjuvants. Additionally, it elucidates targeting and optimization strategies for mucosal vaccines to facilitate the development of human vaccines against Brucella.
Collapse
Affiliation(s)
- Tingting Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Yuejie Zhu
- Reproductive Fertility Assistance Center, First Afffliated Hospital of Xinjiang Medical University, China
| | - Juan Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Kaiyu Shang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Zhengwei Yin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Huidong Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Yueyue He
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Jianbing Ding
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Fengbo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China; Department of Clinical laboratory, The First Affiliated hospital of Xinjiang Medical University, China.
| |
Collapse
|
37
|
Mandviwala AS, Huckriede ALW, Arankalle VA, Patil HP. Mucosal delivery of a prefusogenic-F, glycoprotein, and matrix proteins-based virus-like particle respiratory syncytial virus vaccine induces protective immunity as evidenced by challenge studies in mice. Virology 2024; 598:110194. [PMID: 39096774 DOI: 10.1016/j.virol.2024.110194] [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: 06/04/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.
Collapse
Affiliation(s)
- Ahmedali S Mandviwala
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anke L W Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vidya A Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Harshad P Patil
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India.
| |
Collapse
|
38
|
Zhang Q, Sun Y, Sun Y, Zhang H, Yang R. Expression of VP2 protein of novel goose parvovirus in baculovirus and evaluation of its immune effect. Microb Pathog 2024; 195:106751. [PMID: 38880314 DOI: 10.1016/j.micpath.2024.106751] [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: 01/25/2024] [Revised: 05/02/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Short-beak and dwarfism syndrome (SBDS) is a new disease caused by a genetic variant of goose parvovirus in ducks that results in enormous economic losses for the waterfowl industry. Currently, there is no commercial vaccine for this disease, so it is urgent to develop a safer and more effective vaccine to prevent this disease. In this study, we optimized the production conditions to enhance the expression of the recombinant VP2 protein and identified the optimal conditions for subsequent large-scale expression. Furthermore, the protein underwent purification via nickel column affinity chromatography, followed by concentration using ultrafiltration tube. Subsequently, it was observed by transmission electron microscopy (TEM) that the NGPV recombinant VP2 protein assembled into virus-like particles (VLPs) resembling those of the original virus. Finally, the ISA 78-VG adjuvant was mixed with the NGPV-VP2 VLPs to be prepared as a subunit vaccine. Furthermore, both agar gel precipitation test (AGP) and serum neutralization test demonstrated that NGPV VLP subunit vaccine could induce the increase of NGPV antibody in breeding ducks. The ducklings were also challenged with the NGPV, and the results showed that the maternal antibody level could provide sufficient protection to the ducklings. These results indicated that the use of the NGPV VLP subunit vaccine based on the baculovirus expression system could facilitate the large-scale development of a reliable vaccine in the future.
Collapse
Affiliation(s)
- Qing Zhang
- Qingdao agriculture university, Qingdao, 266109, China
| | | | - Yudian Sun
- Qingdao agriculture university, Qingdao, 266109, China
| | | | - Ruimei Yang
- Qingdao agriculture university, Qingdao, 266109, China.
| |
Collapse
|
39
|
Ren M, Abdullah SW, Pei C, Guo H, Sun S. Use of virus-like particles and nanoparticle-based vaccines for combating picornavirus infections. Vet Res 2024; 55:128. [PMID: 39350170 PMCID: PMC11443892 DOI: 10.1186/s13567-024-01383-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
Picornaviridae are non-enveloped ssRNA viruses that cause diseases such as poliomyelitis, hand-foot-and-mouth disease (HFMD), hepatitis A, encephalitis, myocarditis, and foot-and-mouth disease (FMD). Virus-like particles (VLPs) vaccines mainly comprise particles formed through the self-assembly of viral capsid proteins (for enveloped viruses, envelope proteins are also an option). They do not contain the viral genome. On the other hand, the nanoparticles vaccine (NPs) is mainly composed of self-assembling biological proteins or nanomaterials, with viral antigens displayed on the surface. The presentation of viral antigens on these particles in a repetitive array can elicit a strong immune response in animals. VLPs and NPs can be powerful platforms for multivalent antigen presentation. This review summarises the development of virus-like particle vaccines (VLPs) and nanoparticle vaccines (NPs) against picornaviruses. By detailing the progress made in the fight against various picornaviruses such as poliovirus (PV), foot-and-mouth disease virus (FMDV), enterovirus (EV), Senecavirus A (SVA), and encephalomyocarditis virus (EMCV), we in turn highlight the significant strides made in vaccine technology. These advancements include diverse construction methods, expression systems, elicited immune responses, and the use of various adjuvants. We see promising prospects for the continued development and optimisation of VLPs and NPs vaccines. Future research should focus on enhancing these vaccines' immunogenicity, stability, and delivery methods. Moreover, expanding our understanding of the interplay between these vaccines and the immune system will be crucial. We hope these insights will inspire and guide fellow researchers in the ongoing quest to combat picornavirus infections more effectively.
Collapse
Affiliation(s)
- Mei Ren
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gembloux Agro-Biotech, University of Liege, Gembloux, Belgium
| | - Sahibzada Waheed Abdullah
- Livestock and dairy development department peshawar, Government of Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
40
|
Huang Z, Zhu Z, Liu L, Song W, Chen X. Preparation of viromimetic rod-like nanoparticle vaccines (RLNVax) and study of their humoral immune activation efficacy. Biomater Sci 2024; 12:5115-5122. [PMID: 39225616 DOI: 10.1039/d4bm00827h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Virus-like nanoparticle vaccines can efficiently activate the humoral immune response by cross-linking B cell receptors with their surface multivalent antigen arrays. This structurally dependent mechanism makes it crucial to regulate and optimize structural parameters to enhance the efficacy of nanoparticle vaccines. In this study, we prepared nanoparticle vaccines with different aspect ratios by chemically modifying antigen proteins onto the surfaces of poly(amino acid) nanoparticles of various shapes (spherical, ellipsoidal, and rod-like). This allowed us to investigate the impact of structural anisotropy on the humoral immune activation efficacy of nanoparticle vaccines. Furthermore, the end-group molecules of poly(amino acid) materials possess aggregation-induced emission (AIE) properties, which facilitate monitoring the dynamics of nano-assemblies within the body. Results showed that rod-like nanoparticle vaccines (RLNVax) with a higher aspect ratio (AR = 5) exhibited greater lymph node draining efficiency and could elicit more effective B cell activation compared to conventional isotropic spherical nanoparticle vaccines. In a murine subcutaneous immunization model using ovalbumin (OVA) as a model antigen, RLNVax elicited antigen-specific antibody titers that were about 64 times and 4.6 times higher than those induced by free antigen proteins and spherical nanoparticle vaccines, respectively. Additionally, when combined with an aluminum adjuvant, antibody titers elicited by RLNVax were further enhanced by 4-fold. These findings indicate that the anisotropic rod-like structure is advantageous for improving the humoral immune activation efficacy of nanoparticle vaccines, providing significant insights for the design and optimization of next-generation nanoparticle vaccines.
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
| | - 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
| | - 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
| | - 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
|
41
|
Edge RJ, Marriott AE, Stars EL, Patel RN, Wilkinson MC, King LDW, Slagboom J, Tan CH, Ratanabanangkoon K, Draper SJ, Ainsworth S. Plug and play virus-like particles for the generation of anti-toxin antibodies. Toxicon X 2024; 23:100204. [PMID: 39280983 PMCID: PMC11401359 DOI: 10.1016/j.toxcx.2024.100204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/08/2024] [Accepted: 08/18/2024] [Indexed: 09/18/2024] Open
Abstract
Snakebite is a major global health concern, for which antivenom remains the only approved treatment to neutralise the harmful effects of the toxins. However, some medically important toxins are poorly immunogenic, resulting in reduced efficacy of the final product. Boosting the immunogenicity of these toxins in the commercial antivenom immunising mixtures could be an effective strategy to improve the final dose efficacy, and displaying snake antigens on Virus-like particles (VLPs) is one method for this. However, despite some applications in the field of snakebite, VLPs have yet to be explored in methods that could be practical at an antivenom manufacturing scale. Here we describe the utilisation of a "plug and play" VLP system to display immunogenic linear peptide epitopes from three finger toxins (3FTxs) and generate anti-toxin antibodies. Rabbits were immunised with VLPs displaying individual consensus linear epitopes and their antibody responses were characterised by immunoassay. Of the three experimental consensus sequences, two produced antibodies capable of recognising the consensus peptides, whilst only one of these could also recognise native whole toxins. Further characterisation of antibodies raised against this peptide demonstrated a sub-class specific response, and that these were able to elicit partially neutralising antibody responses, resulting in increased survival times in a murine snakebite envenoming model.
Collapse
Affiliation(s)
- Rebecca J Edge
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L3 5RF, United Kingdom
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| | - Amy E Marriott
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L3 5RF, United Kingdom
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| | - Emma L Stars
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| | - Rohit N Patel
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| | - Mark C Wilkinson
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, United Kingdom
| | - Julien Slagboom
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Choo Hock Tan
- School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300, Taiwan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kavi Ratanabanangkoon
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, United Kingdom
| | - Stuart Ainsworth
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L3 5RF, United Kingdom
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| |
Collapse
|
42
|
Zhu J, Tao P, Chopra AK, Rao VB. Bacteriophage T4 as a Protein-Based, Adjuvant- and Needle-Free, Mucosal Pandemic Vaccine Design Platform. Annu Rev Virol 2024; 11:395-420. [PMID: 38768614 PMCID: PMC11690488 DOI: 10.1146/annurev-virology-111821-111145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The COVID-19 pandemic has transformed vaccinology. Rapid deployment of mRNA vaccines has saved countless lives. However, these platforms have inherent limitations including lack of durability of immune responses and mucosal immunity, high cost, and thermal instability. These and uncertainties about the nature of future pandemics underscore the need for exploring next-generation vaccine platforms. Here, we present a novel protein-based, bacteriophage T4 platform for rapid design of efficacious vaccines against bacterial and viral pathogens. Full-length antigens can be displayed at high density on a 120 × 86 nm phage capsid through nonessential capsid binding proteins Soc and Hoc. Such nanoparticles, without any adjuvant, induce robust humoral, cellular, and mucosal responses when administered intranasally and confer sterilizing immunity. Combined with structural stability and ease of manufacture, T4 phage provides an excellent needle-free, mucosal pandemic vaccine platform and allows equitable vaccine access to low- and middle-income communities across the globe.
Collapse
Affiliation(s)
- Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, USA; ,
| | - Pan Tao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ashok K Chopra
- Department of Microbiology and Immunology, Sealy Institute for Vaccine Sciences, Institute for Human Infections and Immunity, and Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Venigalla B Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, USA; ,
| |
Collapse
|
43
|
Zhou Q, Song X, Li Y, Huang J, Yu QS, Den GN, Zhang JQ, Zhu CX, Zhang B. Preparation of a novel type I feline coronavirus virus-like particle vaccine and its immunogenicity in mice and cats. Microb Pathog 2024; 194:106795. [PMID: 39019122 DOI: 10.1016/j.micpath.2024.106795] [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: 12/09/2023] [Revised: 06/10/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Feline coronavirus (FCoV) infection is a leading cause of death in cats. In this study, we produced FCoV-I virus-like particles (VLPs) containing E, M, N, and S proteins using a baculovirus expression system and mixed VLPs with the adjuvants MF59 and CpG 55.2 to prepare an VLP/MF59/CpG vaccine. After immunization of mice with the vaccine, IgG specific antibodies titers against S and N proteins increased to 1:12,800, and IFN-γ+ and IL-4+ splenocytes were significantly increased. Following immunization of FCoV-negative cats, the S protein antibodies in immunized cats (5/5) increased significantly, with a peak of 1:12,800. Notably, after booster vaccination in FCoV-positive cats, a significant reduction in viral load was observed in the feces of partial cats (4/5), and the FCoV-I negative conversion was found in two immunized cats (2/5). Therefore, the VLP/MF59/CpG vaccine is a promising candidate vaccine to prevent the FCoV infection.
Collapse
MESH Headings
- Animals
- Cats
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Mice
- Coronavirus, Feline/immunology
- Immunoglobulin G/blood
- Adjuvants, Immunologic/administration & dosage
- Viral Load
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Interleukin-4/metabolism
- Interferon-gamma/metabolism
- Mice, Inbred BALB C
- Feces/virology
- Adjuvants, Vaccine
- Polysorbates/administration & dosage
- Female
- Coronavirus Infections/prevention & control
- Coronavirus Infections/immunology
- Coronavirus Infections/veterinary
- Immunogenicity, Vaccine
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spleen/immunology
- Cat Diseases/prevention & control
- Cat Diseases/immunology
- Cat Diseases/virology
- Baculoviridae/genetics
- Vaccination
- Immunization, Secondary
- Squalene
Collapse
Affiliation(s)
- Qun Zhou
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Xin Song
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Yan Li
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China; Key laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China
| | - Jian Huang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China; Key laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China
| | - Qi-Sheng Yu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Gu-Nan Den
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Jia-Qi Zhang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Chen-Xi Zhu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Bin Zhang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China; Key laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China.
| |
Collapse
|
44
|
Mi Y, Xu K, Wang W, Kong W, Xu X, Rong X, Tan J. Sequential Immunization with Vaccines Based on SARS-CoV-2 Virus-like Particles Induces Broadly Neutralizing Antibodies. Vaccines (Basel) 2024; 12:927. [PMID: 39204050 PMCID: PMC11359007 DOI: 10.3390/vaccines12080927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Although many people have been vaccinated against COVID-19, infections with SARS-CoV-2 seem hard to avoid. There is a need to develop more effective vaccines and immunization strategies against emerging variants of infectious diseases. To understand whether different immunization strategies using variants sequence-based virus-like particles (VLPs) vaccines could offer superior immunity against future SARS-CoV-2 variants, our team constructed VLPs for the original Wuhan-Hu-1 strain (prototype), Delta (δ) variant, and Omicron (ο) variant of SARS-CoV-2, using baculovirus-insect expression system. Then we used these VLPs to assess the immune responses induced by homologous prime-boost, heterologous prime-boost, and sequential immunizations strategies in a mouse model. Our results showed that the pro+δ+ο sequential strategies elicited better neutralizing antibody responses. These sequential strategies also take advantage of inducing CD4+ T and CD8+ T lymphocytes proliferation and tendency to cytokine of Th1. Currently, our data suggest that sequential immunization with VLPs of encoding spike protein derived from SARS-CoV-2 variants of concern may be a potential vaccine strategy against emerging diseases, such as "Disease X".
Collapse
Affiliation(s)
- Youjun Mi
- Department of Pathophysiology, School of BasicMedical Sciences, Lanzhou University, Lanzhou 730000, China;
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (W.W.); (W.K.); (X.X.); (X.R.)
| | - Kun Xu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Department of Immunology, School of Basic Medicine Sciences, Lanzhou University, Lanzhou 730000, China;
- People’s Hospital of Qianxinan Prefecture, Xingyi 562400, China
| | - Wenting Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (W.W.); (W.K.); (X.X.); (X.R.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Department of Immunology, School of Basic Medicine Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Weize Kong
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (W.W.); (W.K.); (X.X.); (X.R.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Department of Immunology, School of Basic Medicine Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Xiaonan Xu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (W.W.); (W.K.); (X.X.); (X.R.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Department of Immunology, School of Basic Medicine Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Xifeng Rong
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (W.W.); (W.K.); (X.X.); (X.R.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Department of Immunology, School of Basic Medicine Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Jiying Tan
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (W.W.); (W.K.); (X.X.); (X.R.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Department of Immunology, School of Basic Medicine Sciences, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
45
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
46
|
Hussain T, Zhao Z, Murphy B, Taylor ZE, Gudorf JA, Klein S, Barnes LF, VanNieuwenhze M, Jarrold MF, Zlotnick A. Chemically Tagging Cargo for Specific Packaging inside and on the Surface of Virus-like Particles. ACS NANO 2024; 18:21024-21037. [PMID: 39087909 PMCID: PMC11503556 DOI: 10.1021/acsnano.4c02056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Virus-like particles (VLPs) have untapped potential for packaging and delivery of macromolecular cargo. To be a broadly useful platform, there needs to be a strategy for attaching macromolecules to the inside or the outside of the VLP with minimal modification of the platform or cargo. Here, we repurpose antiviral compounds that bind to hepatitis B virus (HBV) capsids to create a chemical tag to noncovalently attach cargo to the VLP. Our tag consists of a capsid assembly modulator, HAP13, connected to a linker terminating in maleimide. Our cargo is a green fluorescent protein (GFP) with a single addressable cysteine, a feature that can be engineered in many proteins. The HAP-GFP construct maintained HAP's intrinsic ability to bind HBV capsids and accelerate assembly. We investigated the capacity of HAP-GFP to coassemble with HBV capsid protein and bind to preassembled capsids. HAP-GFP binding was concentration-dependent, sensitive to capsid stability, and dependent on linker length. Long linkers had the greatest activity to bind capsids, while short linkers impeded assembly and damaged intact capsids. In coassembly reactions, >20 HAP-GFP molecules were presented on the outside and inside of the capsid, concentrating the cargo by more than 100-fold compared to bulk solution. We also tested an HAP-GFP with a cleavable linker so that external GFP molecules could be removed, resulting in exclusive internal packaging. These results demonstrate a generalizable strategy for attaching cargo to a VLP, supporting development of HBV as a modular VLP platform.
Collapse
Affiliation(s)
- Tariq Hussain
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhongchao Zhao
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Brennan Murphy
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Zachary E Taylor
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jessica A Gudorf
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Shelby Klein
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Lauren F Barnes
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Michael VanNieuwenhze
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Martin F Jarrold
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Adam Zlotnick
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
47
|
Shen X, Yang YB, Gao Y, Wang S, Wang H, Sun M, Meng F, Tang YD, Tu Y, Kong Q, An TQ, Cai XH. Lipid A-modified Escherichia coli can produce porcine parvovirus virus-like particles with high immunogenicity and minimal endotoxin activity. Microb Cell Fact 2024; 23:222. [PMID: 39118114 PMCID: PMC11308658 DOI: 10.1186/s12934-024-02497-9] [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: 06/17/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND A cost-effective Escherichia coli expression system has gained popularity for producing virus-like particle (VLP) vaccines. However, the challenge lies in balancing the endotoxin residue and removal costs, as residual endotoxins can cause inflammatory reactions in the body. RESULTS In this study, porcine parvovirus virus-like particles (PPV-VLPs) were successfully assembled from Decreased Endotoxic BL21 (BL21-DeE), and the effect of structural changes in the lipid A of BL21 on endotoxin activity, immunogenicity, and safety was investigated. The lipopolysaccharide purified from BL21-DeE produced lower IL-6 and TNF-α than that from wild-type BL21 (BL21-W) in both RAW264.7 cells and BALB/c mice. Additionally, mice immunized with PPV-VLP derived form BL21-DeE (BL21-DeE-VLP) showed significantly lower production of inflammatory factors and a smaller increase in body temperature within 3 h than those immunized with VLP from BL21-W (BL21-W-VLP) and endotoxin-removed VLP (ReE-VLP). Moreover, mice in the BL21-DeE-VLP immunized group had similar levels of serum antibodies as those in the BL21-W-VLP group but significantly higher levels than those in the ReE-VLP group. Furthermore, the liver, lungs, and kidneys showed no pathological damage compared with the BL21-W-VLP group. CONCLUSION Overall, this study proposes a method for producing VLP with high immunogenicity and minimal endotoxin activity without chemical or physical endotoxin removal methods. This method could address the issue of endotoxin residues in the VLP and provide production benefits.
Collapse
Affiliation(s)
- Xuegang Shen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Yong-Bo Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
- Heilongjiang Veterinary Biopharmaceutical Engineering Technology Research Center, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yanfei Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Shujie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Mingxia Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Fandan Meng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Yan-Dong Tang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Yabin Tu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, Beibei District, Chongqing, 400715, China.
| | - Tong-Qing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China.
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Xue-Hui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China.
- Heilongjiang Veterinary Biopharmaceutical Engineering Technology Research Center, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| |
Collapse
|
48
|
Siebart JC, Chan CS, Yao X, Su FY, Kwong GA. In vivo gene delivery to immune cells. Curr Opin Biotechnol 2024; 88:103169. [PMID: 38972172 PMCID: PMC11316639 DOI: 10.1016/j.copbio.2024.103169] [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: 02/13/2023] [Revised: 11/16/2023] [Accepted: 06/14/2024] [Indexed: 07/09/2024]
Abstract
Immune cell therapies are an emerging class of living drugs that rely on the delivery of therapeutic transgenes to enhance, modulate, or restore cell function, such as those that encode for tumor-targeting receptors or replacement proteins. However, many cellular immunotherapies are autologous treatments that are limited by high manufacturing costs, typical vein-to-vein time of 3-4 weeks, and severe immune-related adverse effects. To address these issues, different classes of gene delivery vehicles are being developed to target specific immune cell subsets in vivo to address the limitations of ex vivo manufacturing, modulate therapeutic responses in situ, and reduce on- and off-target toxicity. The success of in vivo gene delivery to immune cells - which is being tested at the preclinical and clinical stages of development for the treatment of cancer, infectious diseases, and autoimmunity - is paramount for the democratization of cellular immunotherapies.
Collapse
Affiliation(s)
- Jamison C Siebart
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Ching S Chan
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Xinyi Yao
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Fang-Yi Su
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Gabriel A Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA 30332, USA; Georgia ImmunoEngineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
49
|
Josi R, Pardini A, Haindrich A, Marar SV, Vogt ACS, Gessler A, Rentsch D, Cherubini P, Bachmann MF, Mohsen MO. Green Routes: Exploring Protein-Based Virus-like Nanoparticle Transport and Immune Activation in Nicotiana benthamiana for Biotechnological Applications. Vaccines (Basel) 2024; 12:831. [PMID: 39203957 PMCID: PMC11358932 DOI: 10.3390/vaccines12080831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/03/2024] Open
Abstract
Viral, bacterial, fungal, and nematode infections cause significant agricultural losses, with limited treatment options, necessitating novel approaches to enhance plant defense systems and protection against pathogens. Virus-like nanoparticles (VLPs), extensively used in animal and human therapies (e.g., vaccines and immune enhancers), hold potential for novel agricultural solutions and advancing plant nanotechnology. This study employed various methodologies, including VLP production, confocal microscopy, and real-time qPCR. Our findings demonstrated the presence of 30 nm Qβ-VLPs, fluorescently labeled, within the intercellular space of Nicotiana benthamiana leaves one hour post-infiltration. Furthermore, infiltration with Qβ-VLPs led to an upregulation of key defense genes (NbPR1a, NbPR5, NbNPR, NbERF1, NbMYC2, and NbLRR2) in treated plants. Using RT-qPCR, a significant increase in the relative expression levels of defense genes was observed, with sustained high levels of NbERF1 and NbLRR2 even after 24 h. These findings suggest that Qβ-VLPs effectively upregulate genes crucial for pathogen defense in N. benthamiana, initiating PAMP-triggered immunity and launching signaling cascades that enhance defense mechanisms. This innovative application of VLPs to activate plant defense programs advances plant nanobiotechnology, offering new agricultural solutions.
Collapse
Affiliation(s)
- Romano Josi
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland (M.O.M.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), 3012 Bern, Switzerland
| | - Alessandro Pardini
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland (M.O.M.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), 3012 Bern, Switzerland
| | | | - Sanjana V. Marar
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland (M.O.M.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
| | - Anne-Cathrine S. Vogt
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland (M.O.M.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), 3012 Bern, Switzerland
| | - Arthur Gessler
- WSL, Swiss Federal Institute for Forest Snow and Landscape Research, 8903 Birmensdorf, Switzerland
- Institute of Terrestrial Ecosystems, ETH Zürich, 8092 Zürich, Switzerland
| | - Doris Rentsch
- Institute of Plant Sciences, University of Bern, 3013 Bern, Switzerland
| | - Paolo Cherubini
- WSL, Swiss Federal Institute for Forest Snow and Landscape Research, 8903 Birmensdorf, Switzerland
- Deptartment of Forest and Conservation Sciences, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Martin F. Bachmann
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland (M.O.M.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
| | - Mona O. Mohsen
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland (M.O.M.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
- Tajarub Research & Development, Doha P.O. Box 12627, Qatar
| |
Collapse
|
50
|
Choi W, Cha S, Kim K. Navigating the CRISPR/Cas Landscape for Enhanced Diagnosis and Treatment of Wilson's Disease. Cells 2024; 13:1214. [PMID: 39056796 PMCID: PMC11274827 DOI: 10.3390/cells13141214] [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: 05/27/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) system continues to evolve, thereby enabling more precise detection and repair of mutagenesis. The development of CRISPR/Cas-based diagnosis holds promise for high-throughput, cost-effective, and portable nucleic acid screening and genetic disease diagnosis. In addition, advancements in transportation strategies such as adeno-associated virus (AAV), lentiviral vectors, nanoparticles, and virus-like vectors (VLPs) offer synergistic insights for gene therapeutics in vivo. Wilson's disease (WD), a copper metabolism disorder, is primarily caused by mutations in the ATPase copper transporting beta (ATP7B) gene. The condition is associated with the accumulation of copper in the body, leading to irreversible damage to various organs, including the liver, nervous system, kidneys, and eyes. However, the heterogeneous nature and individualized presentation of physical and neurological symptoms in WD patients pose significant challenges to accurate diagnosis. Furthermore, patients must consume copper-chelating medication throughout their lifetime. Herein, we provide a detailed description of WD and review the application of novel CRISPR-based strategies for its diagnosis and treatment, along with the challenges that need to be overcome.
Collapse
Affiliation(s)
- Woong Choi
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea;
| | - Seongkwang Cha
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea;
- Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kyoungmi Kim
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea;
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|