1
|
Javadi MM, Soleimani N, Zandi A. Enhancing breast Cancer immunotherapy using gold nanoparticles carrying tumor antigens. Sci Rep 2025; 15:16747. [PMID: 40369128 PMCID: PMC12078636 DOI: 10.1038/s41598-025-97343-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/03/2025] [Indexed: 05/16/2025] Open
Abstract
Cancer immunotherapy combined with standard treatments could provide an effective approach to enhancing anti-tumor responses. Activating antigen-presenting cells, such as dendritic cells (DCs), plays a central role in generating robust anti-tumor immune responses. Freund's adjuvant together with nanoparticles (NPs) and tumor antigens, promotes significant immune responses and shift antigen-specific T-cell activity from a Th2 to a Th1 response. Herein, Freund's adjuvant was combined with gold nanoparticles and tumor cell lysate (TCL). The AuNPs exhibited a spherical morphology. The in vitro release studies demonstrated a continuous and gradual release of AuNPs and TCL from Freund's adjuvant. The immunogenicity studies revealed high levels of cytokine secretion for IFN-γ, IL- 1, IL- 18, and TCD8+, along with reduced levels of IL- 4 cytokine in immunized mouse models in various treatment groups. In the prophylactic group, tumor growth was delayed, while in the therapeutic group, mouse models had more than 85% reduction within 31 days compared to the control group. The tumor size in the combination strategies, shrank to ~ 86% of its first size in just 17 days after treatment, while the control group tumor size increased by approximately 52%. These data suggest that the proposed drug system is an effective anti-tumor vaccine and also potentiate innate or adaptive immune responses for cancer therapy.
Collapse
Affiliation(s)
- Mahtab Moshref Javadi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Neda Soleimani
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Ashkan Zandi
- Nano-bioelectronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Eng, College of Engineering, Nano Electronic Center of Excellence, University of Tehran, P.O. Box: 14395 - 515, Tehran, Iran.
| |
Collapse
|
2
|
Chalesh A, Khaki P, Moradi Bidhendi S, Tebianian M, Tarnabi MT. Expression and Immunogenicity Analysis of Recombinant Leptospira Interrogans Surface Protein LigA in Mouse Model. Vet Med Sci 2025; 11:e70360. [PMID: 40388240 PMCID: PMC12087437 DOI: 10.1002/vms3.70360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/15/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Pathogenic strains of spirochetes of Leptospira spp. cause a globally distributed zoonotic disease called leptospirosis. The disease has several clinical manifestations, ranging from asymptomatic and subclinical infection to fatal and severe forms. HYPOTHESIS/OBJECTIVES The aim of this study was to produce a recombinant Leptospiral immunoglobulin-like surface protein-A (r-LigA) antigen of Leptospira interrogans in a prokaryotic expression system and to assess its efficacy in a mouse model. MATERIALS AND METHODS The optimal epitopes of the LigA protein were identified via bioinformatics studies. The pET32a+-LigA plasmid construct was cloned into E. coli Top10-DH5α, expressed in E. coli pLysS strains, and subjected to different IPTG concentrations at different times and temperatures. The expressed r-LigA was purified using nickel-affinity (Ni-NTA) chromatography from the insoluble fraction and reassessed by SDS-PAGE, western blotting, dot blotting, and Bradford assay. Female Balb/C mice were immunised subcutaneously with r-LigA alone or emulsified in Freund's adjuvant and subsequently boosted at 2 and 4 weeks. Specific antibody levels were evaluated by indirect ELISA. RESULTS Bioinformatics analysis identified the key antigenic region of LigA spanning amino acids 852 to 1210. Colony PCR and digestion confirmed the successful transformation. Induction using 0.5 mM IPTG at 30°C for 5 h was found to be optimal. Overexpression of r-LigA under optimised conditions accumulated proteins as inclusion bodies. Purification of r-LigA under native conditions using optimised Ni-NTA yielded 1050 µg/mL protein and high immunogenicity by effectively stimulating the immune system in female Balb/C mice. CONCLUSIONS These findings support r-LigA as a strong candidate for future leptospirosis diagnostic tools and subunit vaccine development.
Collapse
Affiliation(s)
- Aida Chalesh
- Department of Microbiology, North Tehran BranchIslamic Azad UniversityTehranIran
| | - Pejvak Khaki
- Department of MicrobiologyRazi Vaccine and Serum Research InstituteAgricultural Research, Education and Extension Organization (AREEO)KarajIran
| | | | - Majid Tebianian
- Department of ImmunologyRazi Vaccine and Serum Research InstituteAgricultural Research, Education and Extension Organization (AREEO)KarajIran
| | - Morteza Taghizadeh Tarnabi
- Department of Medical VaccineRazi Vaccine and Serum Research InstituteAgricultural Research, Education and Extension Organization (AREEO)KarajIran
| |
Collapse
|
3
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2025; 13:2164-2178. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [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: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
4
|
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
|
5
|
Babar M, Aslam B, Faisal MN, Malik A, Akhtar S, Fatima S, Majeed W, Umer A, Farooq MA. Phytochemical characterization and anti-arthritic potential of green-synthesized CuO nanoparticles derived from the Bistorta amplexicaulis root extract. Front Pharmacol 2024; 15:1474592. [PMID: 39741627 PMCID: PMC11685013 DOI: 10.3389/fphar.2024.1474592] [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: 08/01/2024] [Accepted: 11/22/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Rheumatoid arthritis is an autoimmune disease that mainly causes joint damage. The patient experiences loss of appetite, pain, fever, and fatigue. The present study was designed to phytochemically characterize and evaluate the anti-arthritic activity of green-synthesized copper oxide (CuO) nanoparticles (NPs) using the hydroalcoholic extract of Bistorta amplexicaulis roots in an adjuvant-induced arthritic rat model. Material and Methods For this purpose, crude powdered plant material was used for proximate analysis, and the plant extract was assessed for qualitative phytochemical analysis, mineral contents, and flavonoid and phenolic contents, as well as quantitative phytochemical analysis through reversed-phase high-performance liquid chromatography (RP-HPLC) and Fourier-transform infrared (FTIR) spectroscopy. The in vitro antioxidant activity of both extracts was determined by the 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay. The biosynthesized CuO NPs from the Bistorta amplexicaulis extract showed anti-arthritic activity due to the presence of flavonoids and phenols, which showed a pain reliever effect by blocking the cyclo-oxygenase enzyme and has immune suppressant activity, thus securing the joint from destruction. The nanoparticles were characterized by zeta size, zeta potential, scanning electron microscopy (SEM), and FTIR spectroscopy. Forty-eight albino rats were divided randomly into six treatment groups. Results and Disscussion The zeta size and zeta potential of the nanoparticles were 186.8 nm and -9.23 mV, respectively. Joint stiffness, spleen weight, thymus weight, and paw thickness showed a significant decrease after treatment with NPs. The hematological parameters such as red blood cells (RBCs) and hemoglobin showed a significant increase, while platelets and white blood cells (WBCs) showed a significant decrease in NP-treated groups. C-reactive protein (CRP), rheumatoid factor (RF), liver and kidney function biomarkers, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) levels showed a significant decrease at both low and high doses of green-synthesized CuO nanoparticles from the Bistorta amplexicaulis root extract. The final data were analyzed by one way and two-way analysis of variance (ANOVA) and Tukey's multi-comparison test. Conclusion So, from this study, it was concluded that both the plant root extract and green-synthesized CuO nanoparticles have anti-arthritic potential, but CuO NPs showed remarkable results.
Collapse
Affiliation(s)
- Mahrukh Babar
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Bilal Aslam
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Naeem Faisal
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Akhtar
- Department of Biochemistry, A.T. Still University of Health Sciences, Kirksville, MO, United States
| | - Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Wafa Majeed
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Asher Umer
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
6
|
Neves FMDL, Dos Santos KS, Dos Santos RC, de Lima Fontes M, Marcos CM, do Araujo VS, Fusco-Almeida AM, Mendes-Giannini MJS, Moroz A. Multiple Tolerization Subtractive Immunization in the Obtention of Specific Monoclonal Antibodies Against Paracoccidioides lutzii. Monoclon Antib Immunodiagn Immunother 2024; 43:160-170. [PMID: 39665994 DOI: 10.1089/mab.2024.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Paracoccidioidomycosis (PCM) is a chronic endemic mycosis in Latin America, predominantly caused by Paracoccidioides brasiliensis (Pb18) and Paracoccidioides lutzii (Pl01). Diagnosing PCM is challenging due to species-specific antigenic differences, therefore new biomarkers for accurate and rapid detection are needed. This study explores multiple tolerization subtractive immunization (MTSI) to generate monoclonal antibodies against rare or weakly expressed epitopes of Pb18 and Pl01, potentially improving PCM diagnosis. These strains were cultured to obtain cell-free antigens (CFA). MTSI involved immunizing BALB/c mice with CFA from Pb18 as a tolerogen and Pl01 as an immunogen, using Freund's adjuvant and cyclophosphamide to induce immune tolerance. The immune response was monitored via Enzyme-linked immunosorbent assay (ELISA) and Western blotting. Hybridomas were generated by fusing splenocytes from immunized mice with myeloma cells, after which clonal selection was conducted based on reactivity to Pl01 antigens. The study explores the presence of various proteins, including gp43 and Hsp60, in the protein profile of CFAs. Additionally, polyclonal antibody reactivity to Pb18 antigens was significantly reduced, suggesting that MTSI effectively promoted immunological tolerance. Followig the screening of hybridomas, clones with good reactivity to Pl01 and less reactive to Pb18 were selected. The monoclonal clones C1 and E6 exhibited potential specificity for Pl01 antigens. The effective generation of P. lutzii-specific antibodies by MTSI demonstrates this technology's promise for the development of accurate PCM diagnostic instruments. These antibodies have the potential to enhance patient outcomes and reduce the incidence of false-negative diagnoses, which could lead to better disease management.
Collapse
Affiliation(s)
| | | | | | - Marina de Lima Fontes
- BioSmart Nanotechnology LTDA, Araraquara, Brazil
- Department of Chemistry, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Caroline Maria Marcos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | | | | | | | - Andrei Moroz
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
7
|
Irnidayanti Y, Wisaksono SAJ, Darmanto W, Narayan M, Sarma H. Potential protection of resveratrol-tempeh against nephrotoxic and hepatotoxic histological damage in mice induced by aluminum. Tissue Cell 2024; 91:102589. [PMID: 39454472 DOI: 10.1016/j.tice.2024.102589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
Aluminum is a widely distributed metal that, while generally safe at low levels, can become toxic when accumulated in the body. Its exposure is daily through various sources, including food, water, and medications. High levels of aluminum have been shown to adversely affect the kidneys and liver, leading to significant organ damage. Resveratrol-tempeh is a safe protective agent against organ damage caused by aluminum. Here, we investigated the impact of resveratrol on liver and kidney toxicity and Al-induced levels of catalase and malondialdehyde. The mice group was the control group, Al-group, Al+REST5-group, and Al+REST10-group. Aluminum and resveratrol were administered intraperitoneally to mice for four weeks, but resveratrol was administered one hour after exposure to aluminum. Mice were killed by cervical dislocation; the liver and kidney were isolated for slide, and the level of an antioxidant enzyme of catalase and oxidant of malondialdehyde was measured. The results showed that administration of aluminum at a dose of 200 mg/kg body weight caused glomerular shrinkage and proximal tubule degeneration in the kidneys. In addition, it also caused liver tissue damage, with hepatocytes undergoing degeneration, sinusoids dilating, and decreased body weight in the mice. Administration of resveratrol-tempeh tended to decrease malondialdehyde levels and increase catalase activity, although the changes were not significant. It seems that resveratrol-tempeh can repair liver and kidney damage and restore them to normal conditions. Conclusion: Aluminum at 200 mg/kg is toxic to mice. Resveratrol-tempeh can be considered a potential candidate to protect kidney and liver damage caused by aluminum chloride toxicity.
Collapse
Affiliation(s)
- Yulia Irnidayanti
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Negeri Jakarta, Indonesia, Hasjim Asjarie Building, Rawamangun muka, Jakarta 13220, Indonesia.
| | - Salsabilla Audy Julieta Wisaksono
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Negeri Jakarta, Indonesia, Hasjim Asjarie Building, Rawamangun muka, Jakarta 13220, Indonesia.
| | - Win Darmanto
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya 60115, Indonesia.
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 West University Ave., El Paso, TX 79968, United States.
| | - Hemen Sarma
- Department of Botany, Bodoland University, Kokrajhar (BTR), Assam 783370, India.
| |
Collapse
|
8
|
Ratanabanangkoon K. Effective production of snake antivenom by targeting epidermal dendritic cells via the 'low dose, low volume, multi-site' immunization. Toxicon 2024; 251:108156. [PMID: 39490816 DOI: 10.1016/j.toxicon.2024.108156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Snakebite envenomation (SBE) is a serious neglected tropical disease that affects about 3 million people every year and causes over 100,000 deaths annually, mostly in developing countries. WHO has pledged to cut in half the morbidity and mortality due to SBE by 2030. Animal plasma-derived antivenoms, produced mostly in horses and sheep, are the main treatment modality. However, for over a century, equine plasma antivenom production has faced many problems. These include: low neutralizing potency, failure of horses to develop a satisfactory immune response, and a long immunization period. These problems have led to antivenom shortages and higher costs resulting in otherwise avoidable morbidity and mortality in snake bite victims. Attempts have been made to improve the antivenom production process. For example, a number of adjuvants designed to improve the immune response have been tested. In 1997, an immunization protocol involving the use of multi-site, low-volume and venom doses was developed and is currently used in antivenom production. This protocol constituted a significant innovation that has resulted in highly potent antivenoms within much shortened immunization periods, with all the immunized horses responding and with much less venom immunogen used. It has resulted in an ample antivenom supply for use in Thailand and neighboring countries and has led to no reported deaths from snakebite in Thailand in the past few years. The effectiveness of this immunization protocol was the result of a strategy based on targeting dendritic cells which play a pivotal role in the immune response process. This communication summarizes the basis and results of this immunization strategy.
Collapse
Affiliation(s)
- Kavi Ratanabanangkoon
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
9
|
Zeb MT, Dumont E, Khan MT, Shehzadi A, Ahmad I. Multi-Epitopic Peptide Vaccine Against Newcastle Disease Virus: Molecular Dynamics Simulation and Experimental Validation. Vaccines (Basel) 2024; 12:1250. [PMID: 39591153 PMCID: PMC11598688 DOI: 10.3390/vaccines12111250] [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: 10/03/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Newcastle disease virus (NDV) is a highly contagious and economically devastating pathogen affecting poultry worldwide, leading to significant losses in the poultry industry. Despite existing vaccines, outbreaks continue to occur, highlighting the need for more effective vaccination strategies. Developing a multi-epitopic peptide vaccine offers a promising approach to enhance protection against NDV. OBJECTIVES Here, we aimed to design and evaluate a multi-epitopic vaccine against NDV using molecular dynamics (MD) simulation. METHODOLOGY We retrieved NDV sequences for the fusion (F) protein and hemagglutinin-neuraminidase (HN) protein. Subsequently, B-cell and T-cell epitopes were predicted. The top potential epitopes were utilized to design the vaccine construct, which was subsequently docked against chicken TLR4 and MHC1 receptors to assess the immunological response. The resulting docked complex underwent a 1 microsecond (1000 ns) MD simulation. For experimental evaluation, the vaccine's efficacy was assessed in mice and chickens using a controlled study design, where animals were randomly divided into groups receiving either a local ND vaccine or the peptide vaccine or a control treatment. RESULTS The 40 amino acid peptide vaccine demonstrated strong binding affinity and stability within the TLR4 and MHC1 receptor-peptide complexes. The root mean square deviation of peptide vaccine and TLR4 receptor showed rapid stabilization after an initial repositioning. The root mean square fluctuation revealed relatively low fluctuations (below 3 Å) for the TLR4 receptor, while the peptide exhibited higher fluctuations. The overall binding energy of the peptide vaccine with TLR4 and MHC1 receptors amounted to -15.7 kcal·mol-1 and -36.8 kcal·mol-1, respectively. For experimental evaluations in mice and chicken, the peptide vaccine was synthesized using services of GeneScript Biotech® (Singapore) PTE Limited. Experimental evaluations showed a significant immune response in both mice and chickens, with the vaccine eliciting robust antibody production, as evidenced by increasing HI titers over time. Statistical analysis was performed using an independent t-test with Type-II error to compare the groups, calculating the p-values to determine the significance of the immune response between different groups. CONCLUSIONS Multi-epitopic peptide vaccine has demonstrated a good immunological response in natural hosts.
Collapse
Affiliation(s)
- Muhammad Tariq Zeb
- Department of Molecular Biology and Genetics, Institute of Basic Medical Sciences, Khyber Medical University, Phase-V, Hayatabad Peshawar, Peshawar 25100, Pakistan;
- Genomic Laboratory, Veterinary Research Institute, Bacha Khan Chowk, Charsadda Road, Peshawar 25100, Pakistan
| | - Elise Dumont
- Institut de Chimie de Nice, Université Côte d’Azur, CNRS, UMR 7272, 06108 Nice, France;
- Institut Universitaire de France, 5 Rue Descartes, 75005 Paris, France
| | - Muhammad Tahir Khan
- Institute of Molecular Biology & Biotechnology (IMBB), The University of Lahore, KM Defence Road, Lahore 54000, Pakistan;
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Clinical Laboratory, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou 510180, China
- Qihe Laboratory, Qishui Guang East, Qibin District, Hebi 458030, China
| | - Aroosa Shehzadi
- Institute of Molecular Biology & Biotechnology (IMBB), The University of Lahore, KM Defence Road, Lahore 54000, Pakistan;
| | - Irshad Ahmad
- Department of Molecular Biology and Genetics, Institute of Basic Medical Sciences, Khyber Medical University, Phase-V, Hayatabad Peshawar, Peshawar 25100, Pakistan;
| |
Collapse
|
10
|
Alsakini KAMH, Sanci E, Buhur A, Yavasoglu A, Karabay Yavasoglu NÜ, Nalbantsoy A. Single and repeat-dose toxicity and local tolerance assessment of newly developed oil emulsion adjuvant formulations for veterinary purposes. Drug Chem Toxicol 2024; 47:827-838. [PMID: 38093608 DOI: 10.1080/01480545.2023.2291985] [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: 06/07/2023] [Accepted: 11/21/2023] [Indexed: 11/21/2024]
Abstract
Adjuvants are components of vaccines that boost the intensity, duration, and breadth of the immune response. Insight into the mechanisms responsible for the immunotoxicity of both local and systemic adverse reactions following the use of adjuvants has been gained through research over the past twenty years. In the present study, single and repeated-dose toxicity and local tolerance of newly developed Water-in-Oil (W/O) and Water-in-Oil-in-Water (W/O/W) Emulsion adjuvants (Coralvac RZ 528, Coralvac RZ 506, Coralvac AT 318, Coralvac AT 318 SIS and Coralvac 252) by Coral Biotechnology Industry and Trade Incorporated Company were demonstrated after intramuscular injection in mice. In both toxicity studies, no adverse reactions such as death, general appearance, behavior, or weight loss were observed in the mice in the experimental groups. The results indicate that clinical chemistry parameters demonstrated normal function of the major organs and no irreversible damage to the mice in all adjuvant groups compared to the control group. In histopathologic investigation of single dose toxicity study, inflammation, edema, and large amounts of lipid droplets were observed on the 7th day in all experimental groups. On the 14th day, when the control group and the experimental groups were compared, it was seen that inflammation and edema had decreased considerably. Similarly, repeated dose toxicity study showed mild inflammation and edema in the control group, while quite widespread and severe inflammation, edema, and diffuse lipid droplets of varying sizes were observed in all adjuvant groups compared to the control group. These observations would be useful for the future development of oil-based adjuvants and their use in veterinary inactive vaccines.
Collapse
Affiliation(s)
| | - Ebru Sanci
- Center for Drug Development and Pharmacokinetic Applications, Ege University, Izmir, Turkey
| | - Aylin Buhur
- Department of Histology and Embryology, Ege University, Izmir, Turkey
| | - Altuğ Yavasoglu
- Department of Histology and Embryology, Ege University, Izmir, Turkey
| | | | | |
Collapse
|
11
|
Mitsuyama H, Iizasa E, Kukita A, Toda S, Yoshida H, Inoue H, Hara H. Deletion of Card9 eliminates the detrimental facets of mycobacterial adjuvants. Heliyon 2024; 10:e38139. [PMID: 39386804 PMCID: PMC11462255 DOI: 10.1016/j.heliyon.2024.e38139] [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: 11/25/2023] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
Although mycobacterial adjuvants are capable of eliciting a strong adaptive humoral and cellular immunity, they also sometimes provoke detrimental outcomes, including autoimmune/inflammatory syndromes. Here, we show that the deletion of caspase recruitment domain family member 9 (Card9), a signaling adaptor of a set of innate immune receptors, can eliminate the detrimental effects of mycobacterial adjuvants. Long-lasting tissue-destructive skin inflammation at the site of complete Freund's adjuvant (CFA) injection, lung granuloma formation induced by intratracheal Mycobacterium bovis Bacillus Calmette-Guérin infection, and the incidence and severity of experimental autoimmune encephalomyelitis and collagen-induced arthritis induced by autoantigen immunization with CFA were considerably attenuated in Card9-deficient (Card9 -/- ) mice compared to control wild-type mice. Card9 -/- mice showed impaired development of Th17, but not Th1, in the early phase after autoimmune induction, due to the impaired development of IL-6-producing Sirpαhigh dendritic cells, which are essential for priming pathigenic Th17, in the draining lymph nodes. However, Card9 deletion did not affect overall adaptive antibody production or delayed-type hypersensitivity following immunization with CFA, indicating that humoral and type 1 immune responses remained intact. These results suggest that avoiding the activation of Card9 signaling during vaccination with mycobacteria-containing vaccines may mitigate the risk of detrimental type 3 immune responses, while preserving type 1 immune responses that are effective against intracellular pathogens and cancers.
Collapse
Affiliation(s)
- Hideo Mitsuyama
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| | - Ei'ichi Iizasa
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
- Department of Psychosomatic Internal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| | - Akiko Kukita
- Research Center of Arthroplasty, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-City, Saga. 849-8501, Japan
| | - Shuji Toda
- Division of Pathology, Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-City, Saga, 849-8501, Japan
- Department of Pathology, Takagi Hospital, Okawa, Fukuoka, 831-8501, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-City, Saga, 849-8501, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-City, Kagoshima, 890-8544, Japan
| |
Collapse
|
12
|
Benazzouz SM, Benlouahmia N, Bouhadida K, Benlamara M, Arezki N, Sadeddine OEK, Issad M, Attal N, Mansouri K, Derrar F, Djidjik R. Evaluation of the immunoprotective power of a multiple antigenic peptide against Aah II toxin of Androctonus australis hector scorpion. Vaccine X 2024; 19:100503. [PMID: 38868522 PMCID: PMC11167365 DOI: 10.1016/j.jvacx.2024.100503] [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: 10/28/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
Scorpion envenoming (SE) is a public health problem in developing countries. In Algeria, the population exposed to the risk of SE was estimated at 86.45% in 2019. Thus, the development of a vaccine to protect the exposed population against scorpion toxins would be a major advance in the fight against this disease. This work aimed to evaluate the immunoprotective effect of a Multiple Antigenic Peptide against the Aah II toxin of Androctonus australis hector scorpion, the most dangerous scorpion species in Algeria. The immunogen MAP1Aah2 was designed and tested accordingly. This molecule contains a B epitope, derived from Aah II toxin, linked by a spacer to a universal T epitope, derived from the tetanus toxin. The results showed that MAP1Aah2 was non-toxic despite the fact that its sequence was derived from Aah II toxin. The immunoenzymatic assay revealed that the 3 immunization regimens tested generated specific anti-MAP1Aah2 antibodies and cross-reacted with the toxin. Mice immunized with this immunogen were partially protected against mortality caused by challenge doses of 2 and 3 LD50 of the toxin. The survival rate and developed symptoms varied depending on the adjuvant and the challenge dose used. In the in vitro neutralization test, the immune sera of mice having received the immunogen with incomplete Freund's adjuvant neutralized a challenge dose of 2 LD50. Hence, the concept of using peptide dendrimers, based on linear epitopes of scorpion toxins, as immunogens against the parent toxin was established. However, the protective properties of the tested immunogen require further optimizations.
Collapse
Affiliation(s)
- Safouane M. Benazzouz
- Laboratoire de Pharmacologie, Faculté de Pharmacie d’Alger, Université d’Alger 1. 8 Rue du Lieutenant Mohamed Benarfa, El Biar, Alger 16000, Algeria
- Laboratoire des Sérums Thérapeutiques, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Nesrine Benlouahmia
- Laboratoire des Sérums Thérapeutiques, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Karima Bouhadida
- Laboratoire des Sérums Thérapeutiques, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Meriem Benlamara
- Laboratoire des Sérums Thérapeutiques, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Naziha Arezki
- Laboratoire des Sérums Thérapeutiques, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Oum El Kheir Sadeddine
- Laboratoire des Sérums Thérapeutiques, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Mourad Issad
- Laboratoire des Vaccins Viraux Humains, Département des Produits Biologiques Humains, Direction de la Production, Institut Pasteur d’Algérie, Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Nabila Attal
- Laboratoire d’Immunologie, Faculté de Pharmacie d’Alger, Université d’Alger 1. 8 Rue du Lieutenant Mohamed Benarfa, El Biar, Alger 16000, Algeria
- Département d’Immunologie, Direction des Laboratoires, de la Recherche et du Développement, Institut Pasteur d’Algérie, Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
| | - Kamel Mansouri
- Laboratoire de Pharmacologie, Faculté de Pharmacie d’Alger, Université d’Alger 1. 8 Rue du Lieutenant Mohamed Benarfa, El Biar, Alger 16000, Algeria
| | - Fawzi Derrar
- Laboratoire de la Grippe et autres Virus Respiratoires, Département de Virologie, Direction des Laboratoires, de la Recherche et du Développement, Institut Pasteur d’Algérie. Rue du Petit Staoueli, Dely Ibrahim, Alger 16320, Algeria
- Laboratoire de Microbiologie, Faculté de Médecine d’Alger, Université d’Alger 1. 8 Rue du Lieutenant Mohamed Benarfa, El Biar, Alger 16000, Algeria
| | - Reda Djidjik
- Laboratoire d’Immunologie, Faculté de Pharmacie d’Alger, Université d’Alger 1. 8 Rue du Lieutenant Mohamed Benarfa, El Biar, Alger 16000, Algeria
- Laboratoire d’Immunologie Médicale, CHU Issaad Hassani, Rue Ibrahim Hadjeras, Beni Messous, Alger 16206, Algeria
| |
Collapse
|
13
|
Slepenkin A, Pal S, Rasley A, Coleman MA, de la Maza LM. Safety and efficacy of C. muridarum vaccines adjuvanted with CpG-1826 and four concentrations of Montanide-ISA-720-VG. NPJ Vaccines 2024; 9:104. [PMID: 38858418 PMCID: PMC11164897 DOI: 10.1038/s41541-024-00880-6] [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: 11/30/2023] [Accepted: 04/19/2024] [Indexed: 06/12/2024] Open
Abstract
It is recommended that the adjuvant Montanide ISA 720 VG be used at a concentration of 70% v/v. At this concentration, Montanide causes at the site of immunization a local granuloma that can last for several weeks. To determine the safety and protective efficacy of a Chlamydia muridarum MOMP vaccine, formulated with CpG-1826 and four different concentrations of Montanide (70%, 50%, 30% and 10%), BALB/c (H-2d) female mice were immunized twice intramuscularly. Local reactogenicity was significant for vaccines formulated with 70% or 50% Montanide but not for those inoculated with 30% or 10% Montanide. Robust humoral and cell mediated memory immune responses were elicited by the 70%, 50% and 30% Montanide formulations. Mice were challenged intranasally with 104 C. muridarum inclusion forming units (IFU). Based on changes in body weight, lungs's weight and number of IFU recovered, mice vaccinated with the 70%, 50% and 30% Montanide formulations were significantly protected, but not mice receiving 10% Montanide. To conclude, we recommend the 30% Montanide concentration to be tested in humans and animal models to determine its safety and efficacy, in comparison to the 70% Montanide concentration currently used. The 30% Montanide formulation could significantly facilitate licensing of this adjuvant for human use.
Collapse
Affiliation(s)
- Anatoli Slepenkin
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA, 92697-4800, USA
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA, 92697-4800, USA
| | - Amy Rasley
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550-9234, USA
| | - Matthew A Coleman
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550-9234, USA
- University of California Davis, School of Medicine, Department of Radiation Oncology, Sacramento, CA, 95616, USA
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA, 92697-4800, USA.
| |
Collapse
|
14
|
Jordan C, Siebold K, Priegue P, Seeberger PH, Gilmour R. A Fluorinated Sialic Acid Vaccine Lead Against Meningitis B and C. J Am Chem Soc 2024; 146:15366-15375. [PMID: 38768956 PMCID: PMC11157539 DOI: 10.1021/jacs.4c03179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024]
Abstract
Inspired by the specificity of α-(2,9)-sialyl epitopes in bacterial capsular polysaccharides (CPS), a doubly fluorinated disaccharide has been validated as a vaccine lead against Neisseria meningitidis serogroups C and/or B. Emulating the importance of fluorine in drug discovery, this molecular editing approach serves a multitude of purposes, which range from controlling α-selective chemical sialylation to mitigating competing elimination. Conjugation of the disialoside with two carrier proteins (CRM197 and PorA) enabled a semisynthetic vaccine to be generated; this was then investigated in six groups of six mice. The individual levels of antibodies formed were compared and classified as highly glycan-specific and protective. All glycoconjugates induced a stable and long-term IgG response and binding to the native CPS epitope was achieved. The generated antibodies were protective against MenC and/or MenB; this was validated in vitro by SBA and OPKA assays. By merging the fluorinated glycan epitope of MenC with an outer cell membrane protein of MenB, a bivalent vaccine against both serogroups was created. It is envisaged that validation of this synthetic, fluorinated disialoside bioisostere as a potent antigen will open new therapeutic avenues.
Collapse
Affiliation(s)
- Christina Jordan
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| | - Kathrin Siebold
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| | - Patricia Priegue
- Department
of Biomolecular Systems, Max Planck Institute
for Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
- Freie
Universität Berlin, Institute of
Chemistry and Biochemistry, Arnimallee 22, Berlin 14195, Germany
| | - Peter H. Seeberger
- Department
of Biomolecular Systems, Max Planck Institute
for Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
- Freie
Universität Berlin, Institute of
Chemistry and Biochemistry, Arnimallee 22, Berlin 14195, Germany
| | - Ryan Gilmour
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| |
Collapse
|
15
|
Nakkala JR, Li Y, Akter L, Kang X, Chen X. Differential Regulation of DC Function, Adaptive Immunity, and MyD88 Dependence by Two Squalene Emulsion-Based Vaccine Adjuvants. Vaccines (Basel) 2024; 12:531. [PMID: 38793782 PMCID: PMC11125884 DOI: 10.3390/vaccines12050531] [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: 04/01/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
MF59 and AS03 are squalene emulsion-based vaccine adjuvants with similar compositions and droplet sizes. Despite their broad use in licensed influenza vaccines, few studies compared their adjuvant effects and action mechanisms side by side. Considering the majority of adjuvants act on dendritic cells (DCs) to achieve their adjuvant effects, this study compared AddaVax and AddaS03 with similar compositions to MF59 and AS03 adjuvants to enhance antigen uptake, DC maturation, ovalbumin (OVA), and seasonal influenza vaccine-induced immune responses. Considering MF59 was reported to activate MyD88 to mediate its adjuvant effects, this study also investigated whether the above-explored adjuvant effects of AddaVax and AddaS03 depended on MyD88. We found AddaVax more potently enhanced antigen uptake at the local injection site, while AddaS03 more potently enhanced antigen uptake in the draining lymph nodes. AddaS03 but not AddaVax stimulated DC maturation. Adjuvant-enhanced antigen uptake was MyD88 independent, while AddaS03-induced DC maturation was MyD88 dependent. AddaVax and AddaS03 similarly enhanced OVA-induced IgG and subtype IgG1 antibody responses as well as influenza vaccine-induced hemagglutination inhibition antibody titers, whileAddaS03 more potently enhanced OVA-specific IgG2c antibody responses. Both adjuvants depended on MyD88 to enhance vaccine-induced antibody responses, while AddaVax depended more on MyD88 to achieve its adjuvant effects. Our study reveals similarities and differences of the two squalene emulsion-based vaccine adjuvants, contributing to our improved understanding of their action mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Xinyuan Chen
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI 02881, USA; (J.R.N.); (Y.L.); (L.A.); (X.K.)
| |
Collapse
|
16
|
Peralta Alvarez MP, Jones H, Redondo Azema H, Davis C, White AD, Sarfas C, Dennis M, Li S, Wright D, Puentes E, Kimuda S, Belij-Rammerstorfer S, Aguilo N, Martin C, Sharpe S, McShane H, Tanner R. Low-dose M.tb infection but not BCG or MTBVAC vaccination enhances heterologous antibody titres in non-human primates. Front Immunol 2024; 15:1387454. [PMID: 38799468 PMCID: PMC11116990 DOI: 10.3389/fimmu.2024.1387454] [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: 02/17/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Mycobacteria are known to exert a range of heterologous effects on the immune system. The mycobacteria-based Freund's Complete Adjuvant is a potent non-specific stimulator of the immune response used in immunization protocols promoting antibody production, and Mycobacterium bovis Bacille Calmette Guérin (BCG) vaccination has been linked with decreased morbidity and mortality beyond the specific protection it provides against tuberculosis (TB) in some populations and age groups. The role of heterologous antibodies in this phenomenon, if any, remains unclear and under-studied. Methods We set out to evaluate antibody responses to a range of unrelated pathogens following infection with Mycobacterium tuberculosis (M.tb) and vaccination with BCG or a candidate TB vaccine, MTBVAC, in non-human primates. Results We demonstrate a significant increase in the titer of antibodies against SARS-CoV-2, cytomegalovirus, Epstein-Barr virus, tetanus toxoid, and respiratory syncytial virus antigens following low-dose aerosol infection with M.tb. The magnitude of some of these responses correlated with TB disease severity. However, vaccination with BCG administered by the intradermal, intravenous or aerosol routes, or intradermal delivery of MTBVAC, did not increase antibody responses against unrelated pathogens. Discussion Our findings suggest that it is unlikely that heterologous antibodies contribute to the non-specific effects of these vaccines. The apparent dysregulation of B cell responses associated with TB disease warrants further investigation, with potential implications for risk of B cell cancers and novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Holly Jones
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hugo Redondo Azema
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Chloe Davis
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Andrew D. White
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Charlotte Sarfas
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Mike Dennis
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Shuailin Li
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Daniel Wright
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Eugenia Puentes
- Clinical Research Department y Research and Development Department, Biofabri, Grupo Zendal, Pontevedra, Spain
| | - Simon Kimuda
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, London, United Kingdom
| | | | - Nacho Aguilo
- University of Zaragoza, Spanish Network for Research on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martin
- University of Zaragoza, Spanish Network for Research on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Sally Sharpe
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Helen McShane
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Rachel Tanner
- Department of Biology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Lv X, Martin J, Hoover H, Joshi B, Wilkens M, Ullisch DA, Leibold T, Juchum JS, Revadkar S, Kalinovska B, Keith J, Truby A, Liu G, Sun E, Haserick J, DeGnore J, Conolly J, Hill AV, Baldoni J, Kensil C, Levey D, Spencer AJ, Gorr G, Findeis M, Tanne A. Chemical and biological characterization of vaccine adjuvant QS-21 produced via plant cell culture. iScience 2024; 27:109006. [PMID: 38361610 PMCID: PMC10867646 DOI: 10.1016/j.isci.2024.109006] [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: 04/05/2023] [Revised: 09/07/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Many vaccines, including those using recombinant antigen subunits, rely on adjuvant(s) to enhance the efficacy of the host immune responses. Among the few adjuvants clinically approved, QS-21, a saponin-based immunomodulatory molecule isolated from the tree bark of Quillaja saponaria (QS) is used in complex formulations in approved effective vaccines. High demand of the QS raw material as well as manufacturing scalability limitation has been barriers here. We report for the first-time successful plant cell culture production of QS-21 having structural, chemical, and biologic, properties similar to the bark extracted product. These data ensure QS-21 and related saponins are broadly available and accessible to drug developers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John S. Juchum
- Phyton Biotech LLC, 1503 Cliveden Avenue, Delta, BC V3M 6P7, Canada
| | | | | | | | - Adam Truby
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | - Adrian V.S. Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | - Alexandra J. Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, College of Health, Medicine & Wellbeing; Immune Health Program, New Lambton Heights, NSW, Australia
| | | | | | | |
Collapse
|
18
|
Huang Z, Gong H, Sun Q, Yang J, Yan X, Xu F. Research progress on emulsion vaccine adjuvants. Heliyon 2024; 10:e24662. [PMID: 38317888 PMCID: PMC10839794 DOI: 10.1016/j.heliyon.2024.e24662] [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: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Vaccination is the most cost-effective method for preventing various infectious diseases. Compared with conventional vaccines, new-generation vaccines, especially recombinant protein or synthetic peptide vaccines, are safer but less immunogenic than crude inactivated microbial vaccines. The immunogenicity of these vaccines can be enhanced using suitable adjuvants. This is the main reason why adjuvants are of great importance in vaccine development. Several novel human emulsion-based vaccine adjuvants (MF59, AS03) have been approved for clinical use. This paper reviews the research progress on emulsion-based adjuvants and focuses on their mechanism of action. An outlook can be provided for the development of emulsion-based vaccine adjuvants.
Collapse
Affiliation(s)
- Zhuanqing Huang
- Department of Ophthalmology, The No. 944 Hospital of Joint Logistic Support Force of PLA, Gansu 735000, China
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| | - Hui Gong
- Medical School of Chinese PLA, Beijing 100853, China
| | - Qi Sun
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| | - Jinjin Yang
- The Fifth medical center of Chinese PLA General Hospital, Beijing 100071, China
| | - Xiaochuan Yan
- Department of Ophthalmology, The No. 944 Hospital of Joint Logistic Support Force of PLA, Gansu 735000, China
| | - Fenghua Xu
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| |
Collapse
|
19
|
Açık MN, Karagülle B, Yakut S, Öztürk Y, Kutlu MA, Kalın R, Çetinkaya B. Production, characterization and therapeutic efficacy of egg yolk antibodies specific to Nosema ceranae. PLoS One 2024; 19:e0297864. [PMID: 38335158 PMCID: PMC10857605 DOI: 10.1371/journal.pone.0297864] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 01/05/2024] [Indexed: 02/12/2024] Open
Abstract
Nosema disease, caused by Nosema ceranae, one of the single-celled fungal microsporidian parasites, is one of the most important and common diseases of adult honey bees. Since fumagillin, which has been used for decades in the control of Nosema disease in honey bees (Apis mellifera), poses a toxic threat and its efficacy against N. ceranae is uncertain, there is an urgent need to develop alternative prophylactic and curative strategies for the treatment of this disease. The main aim of this study was to investigate the therapeutic potential of specific egg yolk immunoglobulins (IgY) on Nosema disease. For this purpose, the presence of N. ceranae was determined by microscopic and PCR methods in honey bees collected from Nosema suspicious colonies by conducting a field survey. Layered Ataks chickens, divided into four groups each containing 20 animals, were vaccinated with live and inactivated vaccines prepared from field isolates of N. ceranae. Eggs were collected weekly for 10 weeks following the last vaccination. IgY extraction was performed using the PEG precipitation method from egg yolks collected from each group, and the purity of the antibodies was determined by SDS-PAGE and Western Blot. The presence of N. ceranae-specific IgYs was investigated by Western Blot and indirect ELISA methods. It was determined that specific IgYs showed high therapeutic efficacy on Nosema disease in naturally infected bee colonies. In addition, honey bees collected from infected colonies were brought to the laboratory and placed in cages with 30 bees each, and the effectiveness of IgYs was investigated under controlled conditions. It was detected that specific IgY reduced the Nosema spore load and the number of infected bees significantly in both the field and experimental study groups treated for seven days. It was concluded that chicken IgYs, an innovative and eco-friendly method, had a significant potential for use as an alternative to antifungal drugs.
Collapse
Affiliation(s)
- Mehmet Nuri Açık
- Department of Microbiology, Faculty of Veterinary Medicine, University of Bingol, Bingol, Turkiye
| | - Burcu Karagülle
- Department of Microbiology, Faculty of Veterinary Medicine, University of Firat, Elazig, Turkiye
| | - Seda Yakut
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Bingol, Bingol, Turkiye
| | - Yasin Öztürk
- Department of Pharmacology, Faculty of Veterinary Medicine, University of Necmettin Erbakan, Konya, Turkiye
| | - Mehmet Ali Kutlu
- Department of Plant and Animal Production, Vocational School of Food, Agriculture and Livestock, University of Bingol, Bingol, Turkiye
| | - Recep Kalın
- Department of Microbiology, Faculty of Veterinary Medicine, Cumhuriyet University, Sivas, Turkiye
| | - Burhan Çetinkaya
- Department of Microbiology, Faculty of Veterinary Medicine, University of Firat, Elazig, Turkiye
| |
Collapse
|
20
|
Alsakini KAMH, Çöven FO, Nalbantsoy A. Adjuvant effects of novel water/oil emulsion formulations on immune responses against infectious bronchitis (IB) vaccine in mice. Biologicals 2024; 85:101736. [PMID: 38101004 DOI: 10.1016/j.biologicals.2023.101736] [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: 06/20/2023] [Revised: 11/13/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Vaccines have long made use of adjuvants to boost the immune response of the body and reduce the amount of vaccine needed as well as the expense of producing the vaccine. Many vaccine adjuvants are in development, but their application in veterinary vaccinations is restricted due to their lack of efficacy or undesirable side effects. For this reason, it is essential to develop novel adjuvants. To address the issue that the currently available infectious bronchitis (IB) vaccine often fails to produce sufficient immune responses, Coral Biotechnology tested two of their newly developed water-in-oil (W/O) type emulsion adjuvants (Coralvac RZ 528 and Coralvac RZ 506) in the IB vaccine. These adjuvants were tested in a mouse model to determine whether it worked with an inactive IBV H120 vaccine. Vaccine formulations were prepared by combining a virus concentration of 1 × 106 EID50/0.1 ml with an emulsion of the W/O type in a specific ratio. Once the formulations were ready, it was injected intramuscularly as a single dosage, and the mice were monitored for 21 days afterwards. The results showed that anti-IB antibody titer (IgG and IgG1), CD3+ CD8+ T cell responses as well as IFN- γ cytokine production, and splenocyte proliferation were all considerably higher in the IBV H120 with Coralvac RZ 528 and IBV H120 with Coralvac RZ 506 formulation groups than in the viral control group. According to our findings, the humoral and cellular immune responses of mice were significantly enhanced by these novel vaccine adjuvants. Thus, our results provide evidence that the W/O type emulsion adjuvants developed by Coral Biotechnology may be a useful adjuvant in IBV vaccines.
Collapse
Affiliation(s)
| | - Furkan Ozan Çöven
- Department of Bioengineering, Natural and Applied Sciences Institute, Ege University, 35100, İzmir, Turkey.
| | - Ayse Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100, Bornova, Izmir, Turkey.
| |
Collapse
|
21
|
Hirose M, Leliavski A, de Assis LVM, Matveeva O, Skrum L, Solbach W, Oster H, Heyde I. Chronic Inflammation Disrupts Circadian Rhythms in Splenic CD4+ and CD8+ T Cells in Mice. Cells 2024; 13:151. [PMID: 38247842 PMCID: PMC10814081 DOI: 10.3390/cells13020151] [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: 12/07/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
Internal circadian clocks coordinate 24 h rhythms in behavior and physiology. Many immune functions show daily oscillations, and cellular circadian clocks can impact immune functions and disease outcome. Inflammation may disrupt circadian clocks in peripheral tissues and innate immune cells. However, it remains elusive if chronic inflammation impacts adaptive immune cell clock, e.g., in CD4+ and CD8+ T lymphocytes. We studied this in the experimental autoimmune encephalomyelitis (EAE), a mouse model for multiple sclerosis, as an established experimental paradigm for chronic inflammation. We analyzed splenic T cell circadian clock and immune gene expression rhythms in mice with late-stage EAE, CFA/PTx-treated, and untreated mice. In both treatment groups, clock gene expression rhythms were altered with differential effects for baseline expression and peak phase compared with control mice. Most immune cell marker genes tested in this study did not show circadian oscillations in either of the three groups, but time-of-day- independent alterations were observed in EAE and CFA/PTx compared to control mice. Notably, T cell effects were likely independent of central clock function as circadian behavioral rhythms in EAE mice remained intact. Together, chronic inflammation induced by CFA/PTx treatment and EAE immunization has lasting effects on circadian rhythms in peripheral immune cells.
Collapse
Affiliation(s)
- Misa Hirose
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (L.S.)
- Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
- Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | | | - Leonardo Vinícius Monteiro de Assis
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (L.S.)
- Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Olga Matveeva
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (L.S.)
| | - Ludmila Skrum
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (L.S.)
- Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Werner Solbach
- Institute for Medical Microbiology and Hygiene, University of Lübeck, 23562 Lübeck, Germany;
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (L.S.)
- Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Isabel Heyde
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (L.S.)
- Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
22
|
Greenblatt CL, Lathe R. Vaccines and Dementia: Part I. Non-Specific Immune Boosting with BCG: History, Ligands, and Receptors. J Alzheimers Dis 2024; 98:343-360. [PMID: 38393912 PMCID: PMC10977417 DOI: 10.3233/jad-231315] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2024] [Indexed: 02/25/2024]
Abstract
Vaccines such as Bacille Calmette-Guérin (BCG) can apparently defer dementia onset with an efficacy better than all drugs known to date, as initially reported by Gofrit et al. (PLoS One14, e0224433), now confirmed by other studies. Understanding how and why is of immense importance because it could represent a sea-change in how we manage patients with mild cognitive impairment through to dementia. Given that infection and/or inflammation are likely to contribute to the development of dementias such as Alzheimer's disease (Part II of this work), we provide a historical and molecular background to how vaccines, adjuvants, and their component molecules can elicit broad-spectrum protective effects against diverse agents. We review early studies in which poxvirus, herpes virus, and tuberculosis (TB) infections afford cross-protection against unrelated pathogens, a concept known as 'trained immunity'. We then focus on the attenuated TB vaccine, BCG, that was introduced to protect against the causative agent of TB, Mycobacterium tuberculosis. We trace the development of BCG in the 1920 s through to the discovery, by Freund and McDermott in the 1940 s, that extracts of mycobacteria can themselves exert potent immunostimulating (adjuvant) activity; Freund's complete adjuvant based on mycobacteria remains the most potent immunopotentiator reported to date. We then discuss whether the beneficial effects of BCG require long-term persistence of live bacteria, before focusing on the specific mycobacterial molecules, notably muramyl dipeptides, that mediate immunopotentiation, as well as the receptors involved. Part II addresses evidence that immunopotentiation by BCG and other vaccines can protect against dementia development.
Collapse
Affiliation(s)
- Charles L. Greenblatt
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel–Canada (IMRIC), Hebrew University of Jerusalem, Jerusalem, Israel
| | - Richard Lathe
- Division of Infection Medicine, University of Edinburgh Medical School, Edinburgh, UK
| |
Collapse
|
23
|
Cwiklinski K, McEvoy A, López Corrales J, Jewhurst H, Calvani NED, De Marco Verissimo C, Dorey AL, Keane OM, Dalton JP, Lalor R. Fasciola hepatica antioxidant and protease-inhibitor cocktail recombinant vaccines administered five times elicit potent and sustained immune responses in sheep but do not confer protection. Vet Parasitol 2023; 323:110049. [PMID: 37826973 DOI: 10.1016/j.vetpar.2023.110049] [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: 06/20/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/14/2023]
Abstract
Our laboratory's vaccine development strategy against the livestock parasite Fasciola hepatica centres around disrupting key biological processes by combining groups of antigens with similar/complementary functional actions into a single vaccine cocktail. In this study the focus was on antioxidant protein vaccines and a protease inhibitor vaccine aimed at disrupting the parasite's ability to defend against oxidative stress and protease-inhibitor balance, respectively. Two combinations of recombinantly expressed antioxidants were assessed, namely peroxiredoxin (rFhPrx), thioredoxin (rFhTrx) and thioredoxin-glutathione reductase (rFhTGR) (Group 1) and rFhPrx, rFhTrx, and two superoxide dismutases (rFhSOD1 and rFhSOD3) (Group 2). The protease inhibitor vaccine cocktail included representatives of each of the key secreted protease inhibitor families, namely a Kunitz-type inhibitor (rFhKT1), a serpin (rFhSrp1) and a stefin, (rFhStf1) (Group 3). The vaccine combinations were formulated in adjuvant Montanide 61VG administered at five timepoints; two before experimental challenge with 60 F. hepatica metacercariae and three after infection. The vaccine combinations did not reduce the liver fluke burden, and only Group 2 displayed a marginal reduction in egg viability (8.2%). Despite previous results showing an effect of liver fluke vaccines on overall weight gain in infected animals, no significant (P value >0.05) impact on weight gain was observed in this study. Antibodies were elicited against all the vaccine antigens within the cocktails and were maintained at high levels to the end of the trial, due to our strategy of continuing vaccine administration after infection. However, these responses were not boosted by the challenge F. hepatica infection. A comparative analysis with previous vaccine data using a protease inhibitor vaccine found no repeat of the promising outcomes associated with this vaccine, indicating that the addition of rFhSrp1 to the vaccine cocktail did not improve vaccine efficacy. Assessment of liver pathology across the two trials using a modified liver enzyme score (glutamate dehydrogenase to platelet ratio) at eight weeks post infection suggests an association with liver fluke burden above 45 flukes, which could be used to predict liver pathology in future trials. The results reported in this study highlight the ambiguousness in liver fluke vaccine development and the difficulty in obtaining consistent and repeatable protection. This work stresses the need for repetition of trials and the use of sufficiently sized groups to assess vaccine efficacy with adequate statistical power.
Collapse
Affiliation(s)
- Krystyna Cwiklinski
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, UK.
| | - Amanda McEvoy
- Animal & Bioscience Department, Teagasc Mellows Campus, Athenry, Co., Galway, Ireland
| | - Jesús López Corrales
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| | - Heather Jewhurst
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| | - Nichola Eliza Davies Calvani
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| | - Carolina De Marco Verissimo
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| | - Amber Louise Dorey
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| | - Orla M Keane
- Animal & Bioscience Department, Teagasc Grange, Dunsany, Co. Meath, Ireland
| | - John Pius Dalton
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| | - Richard Lalor
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, University of Galway, Galway, Ireland
| |
Collapse
|
24
|
Liu X, Liu Y, Yang X, Lu X, Xu XN, Zhang J, Chen R. Potentiating the Immune Responses of HBsAg-VLP Vaccine Using a Polyphosphoester-Based Cationic Polymer Adjuvant. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48871-48881. [PMID: 37816068 PMCID: PMC10614196 DOI: 10.1021/acsami.3c07491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
Virus-like particle (VLP)-based vaccines are required to be associated with a suitable adjuvant to potentiate their immune responses. Herein, we report a novel, biodegradable, and biocompatible polyphosphoester-based amphiphilic cationic polymer, poly(ethylene glycol)-b-poly(aminoethyl ethylene phosphate) (PEG-PAEEP), as a Hepatitis B surface antigen (HBsAg)-VLP vaccine adjuvant. The polymer adjuvant effectively bound with HBsAg-VLP through electrostatic interactions to form a stable vaccine nanoformulation with a net positive surface charge. The nanoformulations exhibited enhanced cellular uptake by macrophages. HBsAg-VLP/PEG-PAEEP induced a significantly higher HBsAg-specific IgG titer in mice than HBsAg-VLP alone after second immunization, indicative of the antigen-dose sparing advantage of PEG-PAEEP. Furthermore, the nanoformulations exhibited a favorable biocompatibility and in vivo tolerability. This work presents the PEG-PAEEP copolymer as a promising vaccine adjuvant and as a potentially effective alternative to aluminum adjuvants.
Collapse
Affiliation(s)
- Xuhan Liu
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
- Department
of Emergency Medicine, Shenzhen University
General Hospital, Shenzhen University, Shenzhen 518051, China
| | - Yifan Liu
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Xiaoyu Yang
- AIM
Honesty Biopharmaceutical Co., Ltd, Dalian 116620, China
| | - Xinyu Lu
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Xiao-Ning Xu
- Department
of Infectious Diseases, Imperial College
London, London W12 0NN, U.K.
| | - Jiancheng Zhang
- AIM
Honesty Biopharmaceutical Co., Ltd, Dalian 116620, China
| | - Rongjun Chen
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
25
|
Meseko C, Ameji NO, Kumar B, Culhane M. Rational approach to vaccination against highly pathogenic avian influenza in Nigeria: a scientific perspective and global best practice. Arch Virol 2023; 168:263. [PMID: 37775596 DOI: 10.1007/s00705-023-05888-2] [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: 05/23/2023] [Accepted: 08/21/2023] [Indexed: 10/01/2023]
Abstract
Since 2006, highly pathogenic avian influenza (HPAI) subtypes H5Nx have adversely affected poultry production in Nigeria. Successive waves of infections in the last two decades have raised concerns about the ability to contain infections by biosecurity alone, and evidence of recurrent outbreaks suggests a need for adoption of additional control measures such as vaccination. Although vaccination can be used to control virus spread and reduce the morbidity and mortality caused by HPAI, no country using vaccination alone as a control measure against HPAI has been able to eliminate or prevent re-infection. To inform policy in Nigeria, we examined the intricacies of HPAI vaccination, government regulations, and scientific data regarding what kind of vaccines can be used based on subtype, whether inactivated or live attenuated should be used, when to deliver vaccine either proactively or reactively, where to apply vaccination either in disease control zones, regionally, or nationally, and how to vaccinate the targeted poultry population for optimum success. A resurgence of HPAI outbreaks in Nigeria since 2018, after the country was declared free of the epidemic following the first outbreak in 2006, has led to enhanced intervention. Controlled vaccination entails monitoring the application of vaccines, the capacity to differentiate vaccinated from infected (DIVA) flocks, and assessing seroconversion or other immune correlates of protection. Concurrent surveillance for circulating avian influenza virus (AIV) and analyzing AIV isolates obtained via surveillance efforts for genetic and/or antigenic mismatch with vaccine strains are also important. Countries with high investment in commercial poultry farms like Nigeria may identify and zone territories where vaccines can be applied. This may include ring vaccination to control HPAI in areas or production systems at risk of infection. Before adoption of vaccination as an additional control measure on commercial poultry farms, two outcomes must be considered. First, vaccination is an admission of endemicity. Secondly, vaccinated flocks may no longer be made accessible to international poultry markets in accordance with WOAH trade regulations. Vaccination must therefore be approached with utmost caution and be guided by science-based evidence throughout the implementation strategy after thorough risk assessment. Influenza vaccine research, development, and controlled application in addition to biosecurity may be a precautionary measure in the evolving HPAI scenario in Nigeria.
Collapse
Affiliation(s)
- Clement Meseko
- Regional Laboratory for Animal Influenza and Transboundary Diseases, National Veterinary Research Institute, vom plateau, Nigeria.
- Department of Veterinary Public Health and Preventive Medicine, Faculty of Veterinary Medicine, University of Jos, Jos, Nigeria.
| | - Negedu Onogu Ameji
- Department of Veterinary Medicine, Surgery and Radiology, University of Jos, Jos, Nigeria
| | - Binod Kumar
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - Marie Culhane
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Minnesota, USA
| |
Collapse
|
26
|
Lin HH, Wang CY, Hsieh FJ, Liao FZ, Su YK, Pham MD, Lee CY, Chang HC, Hsu HH. Nanodiamonds-in-oil emulsions elicit potent immune responses for effective vaccination and therapeutics. Nanomedicine (Lond) 2023; 18:1045-1059. [PMID: 37610004 DOI: 10.2217/nnm-2023-0179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Background: The use of nanodiamonds (NDs) and fluorescent nanodiamonds (FNDs) as nonallergenic biocompatible additives in incomplete Freund's adjuvant (IFA) to elicit immune responses in vivo was investigated. Methods: C57BL/6 mice were immunized with chicken egg ovalbumin (OVA) in IFA and also OVA-conjugated NDs (or OVA-conjugated FNDs) in IFA to produce antibodies. OVA-expressing E.G7 lymphoma cells and OVA-negative EL4 cells were inoculated in mice to induce tumor formation. Results: The new formulation significantly enhanced immune responses and thus disease resistance. It exhibited specific therapeutic activities, effectively inhibiting the growth of E.G7 tumor cells in mice over 35 days. Conclusion: The high biocompatibility and multiple functionalities of NDs/FNDs render them applicable as active and trackable vaccine adjuvants and antitumor agents.
Collapse
Affiliation(s)
- Hsin-Hung Lin
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Chih-Yen Wang
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Feng-Jen Hsieh
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Fang-Zhen Liao
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Yu-Kai Su
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Minh Dinh Pham
- Institute of Biotechnology, Vietnam Academy of Science & Technology, Ha Noi 100000, Vietnam
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital & College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Huan-Cheng Chang
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
- Department of Chemical Engineering, National Taiwan University of Science & Technology, Taipei City 106, Taiwan
- Department of Chemistry, National Taiwan Normal University, Taipei City 106, Taiwan
| | - Hsao-Hsun Hsu
- Department of Surgery, National Taiwan University Hospital & College of Medicine, National Taiwan University, Taipei 100, Taiwan
- National Taiwan University Cancer Center, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
27
|
Sharanya CS, Abhithaj J, Arun KG, Eeda KR, Bhat V, Variyar EJ, Sabu A, Haridas M. Lipoxygenase inhibitory synthetic derivatives of methyl gallate regulate gene expressions of COX-2 and cytokines to reduce animal model arthritis. Sci Rep 2023; 13:10644. [PMID: 37391468 PMCID: PMC10313808 DOI: 10.1038/s41598-023-37613-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 06/24/2023] [Indexed: 07/02/2023] Open
Abstract
Mammalian lipoxygenases (LOXs) are involved in the biosynthesis of mediators of anaphylactic reactions and have been implicated in cell maturation, the pathogenesis of bronchial asthma, atherosclerosis, rheumatoid arthritis, cardiovascular diseases, Alzheimer's disease and osteoporosis. Hence LOX inhibition in chronic conditions can lead to reducing the disease progression, which can be a good target for treating these diseases. The present study deals with designing methyl gallate derivatives and their anti-inflammatory effect by in silico, in vitro and in vivo methods. Designed derivatives were docked against LOX enzyme, and molecular dynamic simulations were carried out. Following the synthesis of derivatives, in vitro LOX inhibition assay, enzyme kinetics and fluorescence quenching studies were performed. One of the derivatives of methyl gallate (MGSD 1) was demonstrated as an anti-inflammatory agent for the treatment of rheumatoid arthritis in the animal model. Amelioration of Freund's complete adjuvant (FCA)-induced arthritis by methyl gallate and its derivative with a concentration of 10-40 mg.kg-1 has been assessed in vivo in a 28-day-long study. TNF-α and COX-2 gene expression were also studied. Methyl gallate synthetic derivatives (MGSDs) inhibited LOX with an IC50 of 100 nM, 304 nM, and 226 nM for MGSD 1, MGSD 2, and MGSD 3, respectively. Fluorescence quenching methods also prove their binding characteristics, and 200 ns simulations studies showed that the RMSDs for the entire complex were less than 2.8 Å. The in vivo results showed that methyl gallate was required approximately five times diclofenac for the same level of effect, and the synthesised (MGSD 1) compound required only approximately 1/12 of diclofenac for the same level of effect in in-vivo studies. The preeminent expression of COX-2 and TNF-α genes was significantly decreased after the treatment of the methyl gallate derivative. Hence, the in vivo results showed that the referenced synthetic derivative might have more arthritis-reducing properties than the parent compound methyl gallate and is more potent than the standard drug diclofenac, with no apparent induced toxicity.
Collapse
Affiliation(s)
- C S Sharanya
- Department of Biotechnology and Microbiology and IUCB, Dr Janaki Ammal Campus, Kannur University, Palayad, Thalassery, Kannur, Kerala, 670661, India
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - J Abhithaj
- Department of Biotechnology and Microbiology and IUCB, Dr Janaki Ammal Campus, Kannur University, Palayad, Thalassery, Kannur, Kerala, 670661, India
| | - K G Arun
- Department of Biotechnology and Microbiology and IUCB, Dr Janaki Ammal Campus, Kannur University, Palayad, Thalassery, Kannur, Kerala, 670661, India
| | - Koti Reddy Eeda
- Department of Chemistry, Vignan Foundation for Science Technology and Research, Vignan University (Deemed to be University), Vadlamudi, Guntur, Andhra Pradesh, 522 213, India
| | - Vignesh Bhat
- Department of Chemistry, Mangalore University, Mangalagangothri, Karnataka, 574 199, India
| | - E J Variyar
- Department of Biotechnology and Microbiology and IUCB, Dr Janaki Ammal Campus, Kannur University, Palayad, Thalassery, Kannur, Kerala, 670661, India
| | - A Sabu
- Department of Biotechnology and Microbiology and IUCB, Dr Janaki Ammal Campus, Kannur University, Palayad, Thalassery, Kannur, Kerala, 670661, India
| | - M Haridas
- Department of Biotechnology and Microbiology and IUCB, Dr Janaki Ammal Campus, Kannur University, Palayad, Thalassery, Kannur, Kerala, 670661, India.
| |
Collapse
|
28
|
Kumari A, Singh K. Preventive role of cinnamaldehyde against tenuazonic acid- and Freund's adjuvant-induced histopathological and biochemical alterations in the mouse model. Front Microbiol 2023; 14:1159881. [PMID: 37426034 PMCID: PMC10325827 DOI: 10.3389/fmicb.2023.1159881] [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: 02/06/2023] [Accepted: 05/31/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction This study was designed to assess the protective role of cinnamaldehyde (Cin) against the synergistic effect of tenuazonic acid (TeA) and Freund's adjuvant on different organs of Swiss albino mice. Methods TeA was administered singly and in combination with Freund's adjuvant intra-peritoneally. The mice were divided into control (vehicle treated), mycotoxicosis-induced (MI) groups, and treatment groups. The route of administration of TeA was intra-peritoneal. The treatment group (FAICT) received Cin orally as a protective agent against TeA-induced mycotoxicosis. The effects on performance, differential leukocyte counts (DLC), and pathological measurements in eight organs (liver, lungs, kidney, spleen, stomach, heart, brain, and testis) were taken into consideration. Results The body weight and feed consumption decreased significantly in the MI groups, which were reversed in the FAICT group. The necropsy observations revealed an increase in the relative organ-to-body weight percentage in the MI groups, which was restored to normal in the FAICT group. Freund's adjuvant enhanced the effects of TeA on DLC. The antioxidant enzymes SOD and CAT decreased, while MDA increased in the MI groups. Caspase-3 activity was reduced in all organs and remained stable in the treatment group. TeA elevated the ALT concentration in the liver and kidneys and the AST in the liver, kidney, heart, and brain tissues. The oxidative stress induced by TeA in the MI groups was ameliorated in the treatment group. Histopathological observations consisted of NASH, pulmonary oedema and fibrosis, renal crystals and inflammation, splenic hyperplasia, gastric ulceration and cyst, cerebral axonopathy, testicular hyperplasia, and vacuolation in the MI groups. However, no such pathology was recorded in the treatment group. Discussions Thus, it can be concluded that the toxicity of TeA was found to be enhanced when combined with Freund's adjuvant. However, Cin exhibited promising protective effects against TeA + Freund's adjuvant toxicity and reverted the pathological alterations caused by them. Additionally, this study emphasizes Freund's adjuvant's ability to increase mycotoxicity rather than just acting as an immunopotentiator.
Collapse
|
29
|
de Oliveira NR, Santos FDS, Dos Santos VAC, Maia MAC, Oliveira TL, Dellagostin OA. Challenges and Strategies for Developing Recombinant Vaccines against Leptospirosis: Role of Expression Platforms and Adjuvants in Achieving Protective Efficacy. Pathogens 2023; 12:787. [PMID: 37375478 DOI: 10.3390/pathogens12060787] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
The first leptospiral recombinant vaccine was developed in the late 1990s. Since then, progress in the fields of reverse vaccinology (RV) and structural vaccinology (SV) has significantly improved the identification of novel surface-exposed and conserved vaccine targets. However, developing recombinant vaccines for leptospirosis faces various challenges, including selecting the ideal expression platform or delivery system, assessing immunogenicity, selecting adjuvants, establishing vaccine formulation, demonstrating protective efficacy against lethal disease in homologous challenge, achieving full renal clearance using experimental models, and reproducibility of protective efficacy against heterologous challenge. In this review, we highlight the role of the expression/delivery system employed in studies based on the well-known LipL32 and leptospiral immunoglobulin-like (Lig) proteins, as well as the choice of adjuvants, as key factors to achieving the best vaccine performance in terms of protective efficacy against lethal infection and induction of sterile immunity.
Collapse
Affiliation(s)
- Natasha Rodrigues de Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | - Francisco Denis Souza Santos
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | | | - Mara Andrade Colares Maia
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | - Thaís Larré Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | - Odir Antônio Dellagostin
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| |
Collapse
|
30
|
Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards Adjuvant Development: Focus on Antiviral Therapy. Int J Mol Sci 2023; 24:9225. [PMID: 37298177 PMCID: PMC10253057 DOI: 10.3390/ijms24119225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In recent decades, vaccines have been extraordinary resources to prevent pathogen diffusion and cancer. Even if they can be formed by a single antigen, the addition of one or more adjuvants represents the key to enhance the response of the immune signal to the antigen, thus accelerating and increasing the duration and the potency of the protective effect. Their use is of particular importance for vulnerable populations, such as the elderly or immunocompromised people. Despite their importance, only in the last forty years has the search for novel adjuvants increased, with the discovery of novel classes of immune potentiators and immunomodulators. Due to the complexity of the cascades involved in immune signal activation, their mechanism of action remains poorly understood, even if significant discovery has been recently made thanks to recombinant technology and metabolomics. This review focuses on the classes of adjuvants under research, recent mechanism of action studies, as well as nanodelivery systems and novel classes of adjuvants that can be chemically manipulated to create novel small molecule adjuvants.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Federica Poggialini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Pasqualini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Immacolata Trivisani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Chiara Vagaggini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Elena Dreassi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| |
Collapse
|
31
|
Adeoye B, Nakiyingi L, Moreau Y, Nankya E, Olson AJ, Zhang M, Jacobson KR, Gupta A, Manabe YC, Hosseinipour MC, Kumwenda J, Sagar M, AIDS Clinical Trials Group A5274 (REMEMBER) Study Team. Mycobacterium tuberculosis disease associates with higher HIV-1-specific antibody responses. iScience 2023; 26:106631. [PMID: 37168567 PMCID: PMC10165194 DOI: 10.1016/j.isci.2023.106631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/14/2023] [Accepted: 04/04/2023] [Indexed: 05/13/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the most common infection among people with HIV (PWH). Mtb disease-associated inflammation could affect HIV-directed immune responses in PWH. We show that HIV antibodies are broader and more potent in PWH in the presence as compared to the absence of Mtb disease. With co-existing Mtb disease, the virus in PWH also encounters unique antibody selection pressure. The Mtb-linked HIV antibody enhancement associates with specific mediators important for B cell and antibody development. This Mtb humoral augmentation does not occur due to cross-reactivity, a generalized increase in all antibodies, or differences in duration or amount of antigen exposure. We speculate that the co-localization of Mtb and HIV in lymphatic tissues leads to the emergence of potent HIV antibodies. PWH's Mtb disease status has implications for the future use of HIV broadly neutralizing antibodies as prophylaxis or treatment and the induction of better humoral immunity.
Collapse
Affiliation(s)
- Bukola Adeoye
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Lydia Nakiyingi
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Yvetane Moreau
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - Ethel Nankya
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Alex J. Olson
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - Mo Zhang
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - Karen R. Jacobson
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - Amita Gupta
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yukari C. Manabe
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Manish Sagar
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - AIDS Clinical Trials Group A5274 (REMEMBER) Study Team
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
- University of Malawi College of Medicine, Blantyre, Malawi
| |
Collapse
|
32
|
Rando HM, Lordan R, Lee AJ, Naik A, Wellhausen N, Sell E, Kolla L, COVID-19 Review Consortium, Gitter A, Greene CS. Application of Traditional Vaccine Development Strategies to SARS-CoV-2. mSystems 2023; 8:e0092722. [PMID: 36861991 PMCID: PMC10134813 DOI: 10.1128/msystems.00927-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Over the past 150 years, vaccines have revolutionized the relationship between people and disease. During the COVID-19 pandemic, technologies such as mRNA vaccines have received attention due to their novelty and successes. However, more traditional vaccine development platforms have also yielded important tools in the worldwide fight against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A variety of approaches have been used to develop COVID-19 vaccines that are now authorized for use in countries around the world. In this review, we highlight strategies that focus on the viral capsid and outwards, rather than on the nucleic acids inside. These approaches fall into two broad categories: whole-virus vaccines and subunit vaccines. Whole-virus vaccines use the virus itself, in either an inactivated or an attenuated state. Subunit vaccines contain instead an isolated, immunogenic component of the virus. Here, we highlight vaccine candidates that apply these approaches against SARS-CoV-2 in different ways. In a companion article (H. M. Rando, R. Lordan, L. Kolla, E. Sell, et al., mSystems 8:e00928-22, 2023, https://doi.org/10.1128/mSystems.00928-22), we review the more recent and novel development of nucleic acid-based vaccine technologies. We further consider the role that these COVID-19 vaccine development programs have played in prophylaxis at the global scale. Well-established vaccine technologies have proved especially important to making vaccines accessible in low- and middle-income countries. Vaccine development programs that use established platforms have been undertaken in a much wider range of countries than those using nucleic acid-based technologies, which have been led by wealthy Western countries. Therefore, these vaccine platforms, though less novel from a biotechnological standpoint, have proven to be extremely important to the management of SARS-CoV-2. IMPORTANCE The development, production, and distribution of vaccines is imperative to saving lives, preventing illness, and reducing the economic and social burdens caused by the COVID-19 pandemic. Vaccines that use cutting-edge biotechnology have played an important role in mitigating the effects of SARS-CoV-2. However, more traditional methods of vaccine development that were refined throughout the 20th century have been especially critical to increasing vaccine access worldwide. Effective deployment is necessary to reducing the susceptibility of the world's population, which is especially important in light of emerging variants. In this review, we discuss the safety, immunogenicity, and distribution of vaccines developed using established technologies. In a separate review, we describe the vaccines developed using nucleic acid-based vaccine platforms. From the current literature, it is clear that the well-established vaccine technologies are also highly effective against SARS-CoV-2 and are being used to address the challenges of COVID-19 globally, including in low- and middle-income countries. This worldwide approach is critical for reducing the devastating impact of SARS-CoV-2.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ronan Lordan
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amruta Naik
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Sell
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Likhitha Kolla
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - COVID-19 Review Consortium
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Ozer I, Slezak A, Sirohi P, Li X, Zakharov N, Yao Y, Everitt JI, Spasojevic I, Craig SL, Collier JH, Campbell JE, D'Alessio DA, Chilkoti A. An injectable PEG-like conjugate forms a subcutaneous depot and enables sustained delivery of a peptide drug. Biomaterials 2023; 294:121985. [PMID: 36630826 PMCID: PMC10918641 DOI: 10.1016/j.biomaterials.2022.121985] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023]
Abstract
Many biologics have a short plasma half-life, and their conjugation to polyethylene glycol (PEG) is commonly used to solve this problem. However, the improvement in the plasma half-life of PEGylated drugs' is at an asymptote because the development of branched PEG has only had a modest impact on pharmacokinetics and pharmacodynamics. Here, we developed an injectable PEG-like conjugate that forms a subcutaneous depot for the sustained delivery of biologics. The PEG-like conjugate consists of poly[oligo(ethylene glycol) methyl ether methacrylate] (POEGMA) conjugated to exendin, a peptide drug used in the clinic to treat type 2 diabetes. The depot-forming exendin-POEGMA conjugate showed greater efficacy than a PEG conjugate of exendin as well as Bydureon, a clinically approved sustained-release formulation of exendin. The injectable depot-forming exendin-POEGMA conjugate did not elicit an immune response against the polymer, so that it remained effective and safe for long-term management of type 2 diabetes upon chronic administration. In contrast, the PEG conjugate induced an anti-PEG immune response, leading to early clearance and loss of efficacy upon repeat dosing. The exendin-POEGMA depot also showed superior long-term efficacy compared to Bydureon. Collectively, these results suggest that an injectable POEGMA conjugate of biologic drugs that forms a drug depot under the skin, providing favorable pharmacokinetic properties and sustained efficacy while remaining non-immunogenic, offers significant advantages over other commonly used drug delivery technologies.
Collapse
Affiliation(s)
- Imran Ozer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Anna Slezak
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Parul Sirohi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nikita Zakharov
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yunxin Yao
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Ivan Spasojevic
- Duke School of Medicine, Department of Medicine-Oncology, Durham, NC, USA; Duke Cancer Institute, PK/PD Core Laboratory, Durham, NC, USA
| | | | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Division of Endocrinology, Duke University Medical Center, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - David A D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
34
|
Use of adjuvant ISA VG 71 to produce neutralizing egg yolk antibodies against bothropic venom. Appl Microbiol Biotechnol 2023; 107:1947-1957. [PMID: 36723703 DOI: 10.1007/s00253-023-12409-3] [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: 06/27/2022] [Revised: 01/17/2023] [Accepted: 01/25/2023] [Indexed: 02/02/2023]
Abstract
The use of egg yolk antibodies-IgY technology-represents an alternative to the production of mammalian immunoglobulins and has several advantages regarding animal welfare and lower costs of production. The use of adjuvants to achieve the hyperimmunization of laying hens plays a key role in the success of the production of high levels of the antibodies. In the present work, two different adjuvant systems (Freund's adjuvants and MontanideTM ISA 71 VG) were compared to produce IgY anti-Bothrops alternatus. For the first immunization, formalin-inactivated Salmonella was added to MontanideTM ISA 71 VG to emulate Freund's complete adjuvant which includes a mycobacteria antigen. After eight immunizations, IgY produced by using either adjuvant was able to neutralize the lethal activity of the venom in a mouse model, but differences were found regarding the recognition of components of the venom between the two adjuvants tested. Overall, MontanideTM adjuvant used in this work could be a good alternative choice to produce antibodies capable of neutralizing the lethality of complex antigens. This adjuvant is commercially available and used in the formulation of several poultry vaccines and could be used for the IgY technology instead of traditional immunomodulators such as Freund's adjuvants. Key points • IgY extracts recognized major components of the venom.• Avidity indexes of the IgY extracts increased after the successive immunizations.• IgY obtained by two adjuvant systems neutralized the lethal activity of the venom.
Collapse
|
35
|
Luo Z, Wang T, Zhang Z, Zeng H, Yi M, Li P, Pan J, Zhu C, Lin N, Liang S, Verkhratsky A, Nie H. Polyphyllin VI screened from Chonglou by cell membrane immobilized chromatography relieves inflammatory pain by inhibiting inflammation and normalizing the expression of P2X 3 purinoceptor. Front Pharmacol 2023; 14:1117762. [PMID: 36865911 PMCID: PMC9971013 DOI: 10.3389/fphar.2023.1117762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023] Open
Abstract
Objective: Inflammatory pain is one of the most common diseases in daily life and clinic. In this work, we analysed bioactive components of the traditional Chinese medicine Chonglou and studied mechanisms of their analgesic effects. Material and methods: Molecular docking technology and U373 cells overexpressing P2X3 receptors combined with the cell membrane immobilized chromatography were used to screen possible CL bioactive molecules interacting with the P2X3 receptor. Moreover, we investigated the analgesic and anti-inflammatory effects of Polyphyllin VI (PPIV), in mice with chronic neuroinflammatory pain induced by CFA (complete Freund's adjuvant). Results: The results of cell membrane immobilized chromatography and molecular docking showed that PPVI was one of the effective compounds of Chonglou. In mice with CFA-induced chronic neuroinflammatory pain, PPVI decreased the thermal paw withdrawal latency and mechanical paw withdrawal threshold and diminished foot edema. Additionally, in mice with CFA-induced chronic neuroinflammatory pain, PPIV reduced the expression of the pro-inflammatory factors IL-1, IL-6, TNF-α, and downregulated the expression of P2X3 receptors in the dorsal root ganglion and spinal cord. Conclusion: Our work identifies PPVI as a potential analgesic component in the Chonglou extract. We demonstrated that PPVI reduces pain by inhibiting inflammation and normalizing P2X3 receptor expression in the dorsal root ganglion and spinal cord.
Collapse
Affiliation(s)
- Zhenhui Luo
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Tingting Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhenglang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Hekun Zeng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Mengqin Yi
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Peiyang Li
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiaqin Pan
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Chunyan Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Na Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shangdong Liang
- Neuropharmacology Laboratory of Physiology Department, Basic Medical School, Nanchang University, Nanchang, Jiangxi, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom,*Correspondence: Alexei Verkhratsky, ; Hong Nie,
| | - Hong Nie
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China,*Correspondence: Alexei Verkhratsky, ; Hong Nie,
| |
Collapse
|
36
|
Khadka S, Omura S, Sato F, Tsunoda I. Adjuvant Injections Altered the Ileal and Fecal Microbiota Differently with Changes in Immunoglobulin Isotypes and Antimycobacterial Antibody Responses. Int J Mol Sci 2023; 24:2818. [PMID: 36769136 PMCID: PMC9917480 DOI: 10.3390/ijms24032818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Alterations in the gut microbiota, "dysbiosis," have been reported in autoimmune diseases, including multiple sclerosis (MS), and their animal models. Although the animal models were induced by injections of autoantigens with adjuvants, including complete Freund's adjuvant (CFA) and pertussis toxin (PT), the effects of adjuvant injections on the microbiota are largely unknown. We aimed to clarify whether adjuvant injections could affect the microbiota in the ileum and feces. Using 16S rRNA sequencing, we found decreased alpha diversities of the gut microbiota in mice injected with CFA and PT, compared with naïve mice. Overall, microbial profiles visualized by principal component analysis demonstrated dysbiosis in feces, but not in the ileum, of adjuvant-injected mice, where the genera Lachnospiraceae NK4A136 group and Alistipes contributed to dysbiosis. When we compared the relative abundances of individual bacteria, we found changes in 16 bacterial genera in feces and seven genera in the ileum of adjuvant-injected mice, in which increased serum levels of antibody against mycobacteria (a component of CFA) and total IgG2c were correlated with the genus Facklamia. On the other hand, increased IgG1 and IgA concentrations were correlated with the genus Atopostipes. Therefore, adjuvant injections alone could alter the overall microbial profiles (i.e., microbiota) and individual bacterial abundances with altered antibody responses; dysbiosis in animal models could be partly due to adjuvant injections.
Collapse
Affiliation(s)
| | | | | | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| |
Collapse
|
37
|
Yang N, Jin X, Zhu C, Gao F, Weng Z, Du X, Feng G. Subunit vaccines for Acinetobacter baumannii. Front Immunol 2023; 13:1088130. [PMID: 36713441 PMCID: PMC9878323 DOI: 10.3389/fimmu.2022.1088130] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Acinetobacter baumannii is a gram-negative bacterium and a crucial opportunistic pathogen in hospitals. A. baumannii infection has become a challenging problem in clinical practice due to the increasing number of multidrug-resistant strains and their prevalence worldwide. Vaccines are effective tools to prevent and control A. baumannii infection. Many researchers are studying subunit vaccines against A. baumannii. Subunit vaccines have the advantages of high purity, safety, and stability, ease of production, and highly targeted induced immune responses. To date, no A. baumannii subunit vaccine candidate has entered clinical trials. This may be related to the easy degradation of subunit vaccines in vivo and weak immunogenicity. Using adjuvants or delivery vehicles to prepare subunit vaccines can slow down degradation and improve immunogenicity. The common immunization routes include intramuscular injection, subcutaneous injection, intraperitoneal injection and mucosal vaccination. The appropriate immunization method can also enhance the immune effect of subunit vaccines. Therefore, selecting an appropriate adjuvant and immunization method is essential for subunit vaccine research. This review summarizes the past exploration of A. baumannii subunit vaccines, hoping to guide current and future research on these vaccines.
Collapse
Affiliation(s)
- Ning Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Jin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenghua Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fenglin Gao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheqi Weng
- The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingran Du
- Department of Infectious Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xingran Du, ; Ganzhu Feng,
| | - Ganzhu Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xingran Du, ; Ganzhu Feng,
| |
Collapse
|
38
|
Aboul-Ella H, Sayed RH, Abo-Elyazeed HS. Development, preparation, and evaluation of a novel dotted lateral flow immunochromatographic kit for rapid diagnosis of dermatophytosis. Sci Rep 2023; 13:248. [PMID: 36604481 PMCID: PMC9816107 DOI: 10.1038/s41598-023-27443-4] [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: 06/07/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023] Open
Abstract
Dermatophytosis is a widely spread contagious zoonotic disease, affecting both man (tinea) and animals (ringworm). This disease is caused by a group of closely related keratinophilic fungi known collectively as the dermatophytes group. Although the wide distribution of dermatophytosis cases throughout the whole world and its adverse clinical effect on human health, economical effect on productive animals, and pet animal welfare, there is no rapid accurate diagnostic tool for such disease. The current conducted study tries to accomplish the difficult equation by achieving an accurate, sensitive, specific, user-friendly, rapid, robust, device-less, deliverable to end-users, and economic cost for the development and production of diagnostic kits. Through the development of a rapid diagnostic kit based on immunochromatographic assay with three major affordable reproducible production stages; preliminary stage, developmental and standardization stage, and evaluation stage. Obtaining dermatophytes-specific polyclonal antibodies against criteria-based selected dermatophytes strains associating proper gold nanoparticle preparation, characterization, and conjugation, with proper loading of the different bio-reactants on the efficiently laminated and fabricated lateral flow strips were the main challenge and control points through the whole process. Also, as a result of examining 100 animal samples using the new kit, the κ coefficients of the kit with the direct microscopy while the kit with the culture were 0.44 and 0.76, respectively. Therefore, the newly designated and developed kit showed a very promising competitive diagnostic result within 5-7 min through easy-to-be-performed three steps.
Collapse
Affiliation(s)
- Hassan Aboul-Ella
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Rafik Hamed Sayed
- grid.418376.f0000 0004 1800 7673Department of Microbiology, Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Heidy Shawky Abo-Elyazeed
- grid.7776.10000 0004 0639 9286Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
39
|
Lukehart SA, Molini B, Gomez A, Godornes C, Hof R, Fernandez MC, Pitner RA, Gray SA, Carter D, Giacani L, Cameron CE. Immunization with a tri-antigen syphilis vaccine significantly attenuates chancre development, reduces bacterial load, and inhibits dissemination of Treponema pallidum. Vaccine 2022; 40:7676-7692. [PMID: 36376214 PMCID: PMC10318934 DOI: 10.1016/j.vaccine.2022.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022]
Abstract
Syphilis continues to be a significant public health concern worldwide. The disease is endemic in many low- and middle-income countries, and rates have risen sharply in high-income countries over the last decade. The continued prevalence of infectious and congenital syphilis worldwide highlights the need for the development of an effective syphilis vaccine to complement public health measures for syphilis control. The complex, multi-stage course of syphilis infection necessitates a holistic approach to the development of an effective vaccine, in which immunization prevents both the localized stage of infection (typified by the highly infectious chancre) and the disseminated stages of infection (typified by the secondary rash, neurosyphilis, and destructive tertiary lesions, as well as congenital syphilis). Inhibiting development of the infectious chancre would reduce transmission thus providing community- level protection, while preventing dissemination would provide individual-level protection by reducing serious sequelae and may also provide community level protection by reducing shedding during secondary syphilis. In the current study we build upon prior investigations which demonstrated that immunizations with individual, well characterized T. pallidum TprK, TprC, and Tp0751 peptides elicits partial protection against infection in the animal model. Specifically, we show here that immunization with a TprC/TprK/Tp0751 tri-antigen cocktail protects animals from progressive syphilis lesions and substantially inhibits dissemination of the infection.
Collapse
Affiliation(s)
- Sheila A Lukehart
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Barbara Molini
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Alloysius Gomez
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Charmie Godornes
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Rebecca Hof
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Mark C Fernandez
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA
| | | | | | | | - Lorenzo Giacani
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Caroline E Cameron
- Department of Medicine, Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA; Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
40
|
Romero-Giraldo LE, Pulido S, Berrío MA, Flórez MF, Rey-Suárez P, Nuñez V, Pereañez JA. Heterologous Expression and Immunogenic Potential of the Most Abundant Phospholipase A 2 from Coral Snake Micrurus dumerilii to Develop Antivenoms. Toxins (Basel) 2022; 14:toxins14120825. [PMID: 36548722 PMCID: PMC9788014 DOI: 10.3390/toxins14120825] [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: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Micrurus dumerilii is a coral snake of clinic interest in Colombia. Its venom is mainly composed of phospholipases A2 being MdumPLA2 the most abundant protein. Nevertheless, Micrurus species produce a low quantity of venom, which makes it difficult to produce anticoral antivenoms. Therefore, in this work, we present the recombinant expression of MdumPLA2 to evaluate its biological activities and its immunogenic potential to produce antivenoms. For this, a genetic construct rMdumPLA2 was cloned into the pET28a vector and expressed heterologously in bacteria. His-rMdumPLA2 was extracted from inclusion bodies, refolded in vitro, and isolated using affinity and RP-HPLC chromatography. His-rMdumPLA2 was shown to have phospholipase A2 activity, a weak anticoagulant effect, and induced myonecrosis and edema. The anti-His-rMdumPLA2 antibodies produced in rabbits recognized native PLA2, the complete venom of M. dumerilii, and a phospholipase from another species of the Micrurus genus. Antibodies neutralized 100% of the in vitro phospholipase activity of the recombinant toxin and a moderate percentage of the myotoxic activity of M. dumerilii venom in mice. These results indicate that His-rMdumPLA2 could be used as an immunogen to improve anticoral antivenoms development. This work is the first report of an M. dumerilii functional recombinant PLA2.
Collapse
Affiliation(s)
- Luz E. Romero-Giraldo
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
| | - Sergio Pulido
- Tropical Disease Study and Control Program—PECET, University of Antioquia, Medellín 50010, Colombia
- LifeFactors Zona Franca SAS, Rionegro 54047, Colombia
| | - Mario A. Berrío
- Tropical Disease Study and Control Program—PECET, University of Antioquia, Medellín 50010, Colombia
| | - María F. Flórez
- Tropical Disease Study and Control Program—PECET, University of Antioquia, Medellín 50010, Colombia
| | - Paola Rey-Suárez
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
- Centro de Investigación en Recursos Naturales y Sustentabilidad, Universidad Bernardo O’Higgins, Santiago 8320000, Chile
| | - Vitelbina Nuñez
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
- Microbiology School, University of Antioquia, Medellín 50010, Colombia
| | - Jaime A. Pereañez
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
- Correspondence:
| |
Collapse
|
41
|
Eka Saputri M, Aisyah Rahmalia Effendi S, Nadila R, Azzam Fajar S, Damajanti Soejoedono R, Handharyani E, Nadia Poetri O. Immunoglobulin yolk targeting spike 1, receptor binding domain of spike glycoprotein and nucleocapsid of SARS-CoV-2 blocking RBD-ACE2 binding interaction. Int Immunopharmacol 2022; 112:109280. [PMID: 36183680 PMCID: PMC9515349 DOI: 10.1016/j.intimp.2022.109280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/13/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022]
Abstract
Coronavirus disease (COVID)-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has become a global pandemic disease that has social and economic chaos. An alternative mitigation strategy may involve the use of specific immunoglobulin (Ig)-Y derived from chicken eggs. Our study aimed to evaluate the neutralizing potential of specific IgY targeting S1, receptor-binding-domain (RBD) of spike glycoprotein and nucleocapsid (N) of SARS-CoV-2 to inhibit RBD and angiotensin-converting-enzyme-2 (ACE2) binding interaction. Hy-Line Brown laying hens were immunized with recombinant S1, RBD spike glycoprotein, and nucleocapsid (N) of SARS-CoV-2. The presence of specific S1,RBD,N-IgY in serum and egg yolk was verified by indirect enzyme-linked immunosorbent assay (ELISA). Specific S1,RBD,N-IgY was purified and characterized from egg yolk using sodium-dodecyl-sulfate-polyacrylamide-gel-electrophoresis (SDS-PAGE), and was subsequently evaluated for inhibition of the RBD-ACE2 binding interaction in vitro. Specific IgY was present in serum at 1 week post–initial immunization (p.i.i), whereas its present in egg yolk was confirmed at 4 weeks p.i.i. Specific S1,RBD,N-IgY in serum was able to inhibit RBD-ACE2 binding interaction between 4 and 15 weeks p.i.i. The results of the SDS-PAGE revealed the presence of bands with molecular weights of 180 kDa, indicating the presence of whole IgY. Our results demonstrated that S1,RBD,N-IgY was able to inhibit RBD-ACE2 binding interaction in vitro, suggesting its potential use in blocking virus entry. Our study also demonstrated proof-of-concept that laying hens were able to produce this specific IgY, which could block the viral binding and large production of this specific IgY is feasible.
Collapse
Affiliation(s)
- Meliana Eka Saputri
- Study Programme of Medical Microbiology, IPB Postgraduate School, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia
| | - Siti Aisyah Rahmalia Effendi
- Study Programme of Veterinary Medicine, School of Veterinary Medicine and Biomedical Science, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia
| | - Rifa Nadila
- Study Programme of Veterinary Medicine, School of Veterinary Medicine and Biomedical Science, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia
| | - Syauqi Azzam Fajar
- Study Programme of Veterinary Medicine, School of Veterinary Medicine and Biomedical Science, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia
| | - Retno Damajanti Soejoedono
- Division of Medical Microbiology, School of Veterinary Medicine and Biomedical Science, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia
| | - Ekowati Handharyani
- Division of Veterinary Pathology, School of Veterinary Medicine and Biomedical Science, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia
| | - Okti Nadia Poetri
- Division of Medical Microbiology, School of Veterinary Medicine and Biomedical Science, IPB University, Jl Agatis, Kampus IPB Dramaga, Bogor 16680, Indonesia.
| |
Collapse
|
42
|
Kwak HW, Hong SH, Park HJ, Park HJ, Bang YJ, Kim JY, Lee YS, Bae SH, Yoon H, Nam JH. Adjuvant effect of IRES-based single-stranded RNA on melanoma immunotherapy. BMC Cancer 2022; 22:1041. [PMID: 36199130 PMCID: PMC9533600 DOI: 10.1186/s12885-022-10140-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Background Adjuvant therapies such as radiation therapy, chemotherapy, and immunotherapy are usually given after cancer surgery to improve the survival of cancer patients. However, despite advances in several adjuvant therapies, they are still limited in the prevention of recurrences. Methods We evaluated the immunological effects of RNA-based adjuvants in a murine melanoma model. Single-stranded RNA (ssRNA) were constructed based on the cricket paralysis virus (CrPV) internal ribosome entry site (IRES). Populations of immune cells in bone marrow cells and lymph node cells following immunization with CrPVIRES-ssRNA were determined using flow cytometry. Activated cytokine levels were measured using ELISA and ELISpot. The tumor protection efficacy of CrPVIRES-ssRNA was analyzed based on any reduction in tumor size or weight, and overall survival. Results CrPVIRES-ssRNA treatment stimulated antigen-presenting cells in the drain lymph nodes associated with activated antigen-specific dendritic cells. Next, we evaluated the expression of CD40, CD86, and XCR1, showing that immunization with CrPVIRES-ssRNA enhanced antigen presentation by CD8a+ conventional dendritic cell 1 (cDC1), as well as activated antigen-specific CD8 T cells. In addition, CrPVIRES-ssRNA treatment markedly increased the frequency of antigen-specific CD8 T cells and interferon-gamma (IFN-γ) producing cells, which promoted immune responses and reduced tumor burden in melanoma-bearing mice. Conclusions This study provides evidence that the CrPVIRES-ssRNA adjuvant has potential for use in therapeutic cancer vaccines. Moreover, CrPVIRES-ssRNA possesses protective effects on various cancer cell models. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10140-2.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyeong-Jun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - Yoo-Jin Bang
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - Jae-Yong Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - Yu-Sun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Seo-Hyeon Bae
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea. .,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea. .,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea. .,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea.
| |
Collapse
|
43
|
Triller G, Garyfallos DA, Papavasiliou FN, Sklaviadis T, Stavropoulos P, Xanthopoulos K. Immunization with Genetically Modified Trypanosomes Provides Protection against Transmissible Spongiform Encephalopathies. Int J Mol Sci 2022; 23:ijms231810629. [PMID: 36142526 PMCID: PMC9503410 DOI: 10.3390/ijms231810629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Transmissible spongiform encephalopathies are incurable neurodegenerative diseases, associated with the conversion of the physiological prion protein to its disease-associated counterpart. Even though immunization against transmissible spongiform encephalopathies has shown great potential, immune tolerance effects impede the use of active immunization protocols for successful prophylaxis. In this study, we evaluate the use of trypanosomes as biological platforms for the presentation of a prion antigenic peptide to the host immune system. Using the engineered trypanosomes in an immunization protocol without the use of adjuvants led to the development of a humoral immune response against the prion protein in wild type mice, without the appearance of adverse reactions. The immune reaction elicited with this protocol displayed in vitro therapeutic potential and was further evaluated in a bioassay where immunized mice were partially protected in a representative murine model of prion diseases. Further studies are underway to better characterize the immune reaction and optimize the immunization protocol.
Collapse
Affiliation(s)
- Gianna Triller
- Laboratory of Lymphocyte Biology, The Rockefeller University, New York, NY 10065, USA
| | - Dimitrios A. Garyfallos
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - F. Nina Papavasiliou
- Division of Immune Diversity, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Theodoros Sklaviadis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Pete Stavropoulos
- Laboratory of Lymphocyte Biology, The Rockefeller University, New York, NY 10065, USA
- Correspondence: (P.S.); (K.X.); Tel.: +30-2310-997-654 (Κ.Χ.)
| | - Konstantinos Xanthopoulos
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, 57001 Thermi, Greece
- Correspondence: (P.S.); (K.X.); Tel.: +30-2310-997-654 (Κ.Χ.)
| |
Collapse
|
44
|
Wu Y, Wen H, Bernstein ZJ, Hainline KM, Blakney TS, Congdon KL, Snyder DJ, Sampson JH, Sanchez-Perez L, Collier JH. Multiepitope supramolecular peptide nanofibers eliciting coordinated humoral and cellular antitumor immune responses. SCIENCE ADVANCES 2022; 8:eabm7833. [PMID: 35857833 PMCID: PMC9299545 DOI: 10.1126/sciadv.abm7833] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Abstract
Subunit vaccines inducing antibodies against tumor-specific antigens have yet to be clinically successful. Here, we use a supramolecular α-helical peptide nanofiber approach to design epitope-specific vaccines raising simultaneous B cell, CD8+ T cell, and CD4+ T cell responses against combinations of selected epitopes and show that the concurrent induction of these responses generates strong antitumor effects in mice, with significant improvements over antibody or CD8+ T cell-based vaccines alone, in both prophylactic and therapeutic subcutaneous melanoma models. Nanofiber vaccine-induced antibodies mediated in vitro tumoricidal antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). The addition of immune checkpoint and phagocytosis checkpoint blockade antibodies further improved the therapeutic effect of the nanofiber vaccines against murine melanoma. These findings highlight the potential clinical benefit of vaccine-induced antibody responses for tumor treatments, provided that they are accompanied by simultaneous CD8+ and CD4+ responses, and they illustrate a multiepitope cancer vaccine design approach using supramolecular nanomaterials.
Collapse
Affiliation(s)
- Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Hanning Wen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tykia S Blakney
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - David J Snyder
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - John H Sampson
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | | | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
45
|
Fan J, Jin S, Gilmartin L, Toth I, Hussein WM, Stephenson RJ. Advances in Infectious Disease Vaccine Adjuvants. Vaccines (Basel) 2022; 10:1120. [PMID: 35891284 PMCID: PMC9316175 DOI: 10.3390/vaccines10071120] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Vaccines are one of the most significant medical interventions in the fight against infectious diseases. Since their discovery by Edward Jenner in 1796, vaccines have reduced the worldwide transmission to eradication levels of infectious diseases, including smallpox, diphtheria, hepatitis, malaria, and influenza. However, the complexity of developing safe and effective vaccines remains a barrier for combating many more infectious diseases. Immune stimulants (or adjuvants) are an indispensable factor in vaccine development, especially for inactivated and subunit-based vaccines due to their decreased immunogenicity compared to whole pathogen vaccines. Adjuvants are widely diverse in structure; however, their overall function in vaccine constructs is the same: to enhance and/or prolong an immunological response. The potential for adverse effects as a result of adjuvant use, though, must be acknowledged and carefully managed. Understanding the specific mechanisms of adjuvant efficacy and safety is a key prerequisite for adjuvant use in vaccination. Therefore, rigorous pre-clinical and clinical research into adjuvant development is essential. Overall, the incorporation of adjuvants allows for greater opportunities in advancing vaccine development and the importance of immune stimulants drives the emergence of novel and more effective adjuvants. This article highlights recent advances in vaccine adjuvant development and provides detailed data from pre-clinical and clinical studies specific to infectious diseases. Future perspectives into vaccine adjuvant development are also highlighted.
Collapse
Affiliation(s)
- Jingyi Fan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Shengbin Jin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Lachlan Gilmartin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Rachel J. Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| |
Collapse
|
46
|
Abstract
The traditional method for generating polyclonal and monoclonal antibodies requires the immunization of an animal. Selecting the best species of animal and getting that animal's immune system to respond to a target antigen with an antibody response are essential to obtaining good-quality antibodies and hybridomas. There are only a limited number of opportunities for a researcher to intervene to manipulate and tailor the response to a particular antigen. Here we present advice and methods for designing the way in which the antigen is presented to the immune system (i.e., the immunization protocol), including the choice of animal, the antigen dose, the use of adjuvants, the route and number of injections, and the period between injections.
Collapse
|
47
|
Golestani F, Malekan M, Rasooli I, Jahangiri A, Ramezanalizadeh F, Chaudhuri S, Farshchi Andisi V, Schryvers AB. Immunogenicity of loop 3 of Omp34 from A. Baumannii in loopless C-lobe of TbpB of N. meningitidis. Int Immunopharmacol 2022; 110:109013. [PMID: 35785727 DOI: 10.1016/j.intimp.2022.109013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 12/29/2022]
Abstract
Acinetobacter baumannii is a common causative agent of nosocomial infections, with a mortality rate of 43% in infected patients. Due to the emergence of multidrug-resistant (MDR) strains, vaccine development has become necessary. Since the 34 kDa outer membrane protein Omp34 has been identified as a potential vaccine target, we implemented a hybrid antigen approach to target its extracellular loops. Using bioinformatic and structural analyses, we selected Loop 3 from Omp34 and displayed it on the loopless C-lobe (LCL) of TbpB of Neisseria meningitidis. The hybrid antigen and the LCL were produced and used to immunize mice for passive and active immunization and challenge experiments in which the reactivity of the sera was assessed by ELISAs, the bacterial load in the tissues measured and the survival of immunized mice compared. LCL was ineffective in immunization against A. baumannii thus the resulting immunity was due to the presence of Omp34 loop 3. It resulted in increased survival and a reduced bacterial load in the tissues compared to the control groups. The findings indicate that the immunogenicity of Omp34 loops can induce protection against A. baumannii infection, and it could probably be used as a vaccine candidate to control the pathogenesis of A. baumannii.
Collapse
Affiliation(s)
| | | | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran; Molecular Microbiology Research Center and Department of Biology, Shahed University, Tehran, Iran.
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems biology and poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Somshukla Chaudhuri
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Canada
| | - Vahid Farshchi Andisi
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Canada
| | - Anthony B Schryvers
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Canada
| |
Collapse
|
48
|
Escalante-Sansores AR, Absalón AE, Cortés-Espinosa DV. Improving immunogenicity of poultry vaccines by use of molecular adjuvants. WORLD POULTRY SCI J 2022. [DOI: 10.1080/00439339.2022.2091502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | - Angel E. Absalón
- Vaxbiotek SC Departamento de Investigación y Desarrollo, Cuautlancingo, Puebla, Mexico
| | - Diana V. Cortés-Espinosa
- Instituto Politécnico Nacional, Centro de Investigación en Biotecnología Aplicadla, Tlaxcala, Mexico
| |
Collapse
|
49
|
Putri DD, Poetri ON, Candra AA, Soejoedono RD. Production of hyperimmune serum against genotype VII Newcastle disease virus in rabbits with several applications. J Adv Vet Anim Res 2022; 9:211-220. [PMID: 35891669 PMCID: PMC9298101 DOI: 10.5455/javar.2022.i586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/03/2022] Open
Abstract
Objective This study aimed to produce hyperimmune serum against genotype VII Newcastle disease virus (NDV) with several applications. Materials and Methods Production of hyperimmune serum against genotype VII NDV was performed on eight New Zealand white rabbits divided into four groups. Rabbits were immunized three times on the 1st day, the 14th day, and the 30th day. Blood sampling was carried out on the 8th day after the third immunization. Results All groups showed the same pattern of hemagglutination inhibition (HI) titer results. HI titers would peak on the 5th or the 9th day after the second immunization, then decrease until the 3rd day after the third immunization, and increase again on the 5th day after the third immunization. Rabbits immunized intravenously showed higher HI titers than the other groups. These results indicate that the intravenous route for hyperimmune serum production against genotype VII Newcastle disease virus greatly affects the immune response result. Conclusions The production of hyperimmune serum by intravenous immunization three times was able to produce the highest titer of 210 at 38 days. The agar gel precipitation test and the Western blot assay showed that the hyperimmune serum was specific for the Newcastle disease antigen.
Collapse
Affiliation(s)
- Dwi Desmiyeni Putri
- Department of Animal Husbandry, Politeknik Negeri Lampung, Lampung, Indonesia
| | - Okti Nadia Poetri
- Department of Clinic Reproduction and Pathology, Faculty of Veterinary Medicine, IPB University, West Java, Indonesia
| | - Agung Adi Candra
- Department of Animal Husbandry, Politeknik Negeri Lampung, Lampung, Indonesia
| | - Retno Damajanti Soejoedono
- Department of Clinic Reproduction and Pathology, Faculty of Veterinary Medicine, IPB University, West Java, Indonesia
| |
Collapse
|
50
|
Facciolà A, Visalli G, Laganà A, Di Pietro A. An Overview of Vaccine Adjuvants: Current Evidence and Future Perspectives. Vaccines (Basel) 2022; 10:vaccines10050819. [PMID: 35632575 PMCID: PMC9147349 DOI: 10.3390/vaccines10050819] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Vaccinations are one of the most important preventive tools against infectious diseases. Over time, many different types of vaccines have been developed concerning the antigen component. Adjuvants are essential elements that increase the efficacy of vaccination practises through many different actions, especially acting as carriers, depots, and stimulators of immune responses. For many years, few adjuvants have been included in vaccines, with aluminium salts being the most commonly used adjuvant. However, recent research has focused its attention on many different new compounds with effective adjuvant properties and improved safety. Modern technologies such as nanotechnologies and molecular biology have forcefully entered the production processes of both antigen and adjuvant components, thereby improving vaccine efficacy. Microparticles, emulsions, and immune stimulators are currently in the spotlight for their huge potential in vaccine production. Although studies have reported some potential side effects of vaccine adjuvants such as the recently recognised ASIA syndrome, the huge worth of vaccines remains unquestionable. Indeed, the recent COVID-19 pandemic has highlighted the importance of vaccines, especially in regard to managing future potential pandemics. In this field, research into adjuvants could play a leading role in the production of increasingly effective vaccines.
Collapse
Affiliation(s)
- Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Correspondence:
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| | - Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Multi-Specialist Clinical Institute for Orthopaedic Trauma Care (COT), 98124 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| |
Collapse
|