1
|
Esmaeilnejad-Ahranjani P, Shahali Y, Dadar M. Detoxification techniques for bacterial toxins: A pathway to effective toxoid vaccines. Toxicon 2025; 260:108365. [PMID: 40246205 DOI: 10.1016/j.toxicon.2025.108365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Bacterial toxins play a critical role in the virulence of many pathogens, leading to serious diseases such as tetanus, diphtheria, botulism, and entrotoxemia. As key virulence factors, these toxins cause significant tissue damage and disease manifestations in infected hosts. Vaccination against these toxins through toxoid vaccines, composed of inactivated forms of the toxins, represents a vital strategy for preventing toxin-mediated diseases. However, creating effective toxoid vaccines necessitates meticulous detoxification processes that ensure the loss of toxicity while retaining the immunogenic properties inherent in the native toxins. This review offers a comprehensive evaluation of the diverse methodologies employed for detoxifying bacterial toxins, highlighting their advantages, limitations, and implications for vaccine development. By detailing comparisons of efficacy, stability, residual toxicity, and clinical applicability, we demonstrate that while traditional methods utilizing chemical reagents (such as formaldehyde) remain widely used, emerging technologies like genetic inactivation and protein engineering present significant advantages. These innovations promise to advance the development of durable and irreversible toxoid vaccines that protect public health and contribute to future vaccine formulation improvements. Ultimately, this knowledge synthesis aims to guide future research efforts and facilitate the design of safer and more effective toxoid vaccines to combat the public health threats posed by toxin-producing bacteria.
Collapse
Affiliation(s)
- Parvaneh Esmaeilnejad-Ahranjani
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Youcef Shahali
- Centre Hospitalier Universitaire de Besançon, Besançon, France
| | | |
Collapse
|
2
|
Panikulam S, Morgan H, Gutknecht M, Villiger TK, Lebesgue N, Karle AC. Host cell protein-mediated adjuvanticity and immunogenicity risks of biotherapeutics. Biotechnol Adv 2025; 81:108575. [PMID: 40180137 DOI: 10.1016/j.biotechadv.2025.108575] [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: 10/18/2024] [Revised: 02/18/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Host cell proteins (HCPs) are process-related impurities of biotherapeutic production that might affect product quality and/or patient safety. In a few cases, adverse events were attributed to HCPs present in the administered biotherapeutic. HCP-associated immune risks include adjuvanticity and immunogenicity with potential cross-reactivity. Based on the published data, some HCPs can act as adjuvants increasing the immunogenicity of the biotherapeutic as a bystander effect. HCPs may also induce immunogenicity against themselves, resulting in anti-HCP T cell responses and anti-HCP antibody formation. Depending on sequence similarities, these anti-HCP immune responses might theoretically be cross-reactive to the biotherapeutic or human endogenous proteins. In this review, we examine HCP-associated immune-related risks reported from non-clinical and clinical studies. We also discuss the potential and limitations of in vitro and in silico methods to evaluate the adjuvanticity and immunogenicity potential of HCPs. A risk-based assessment of the safety impact of HCPs may include the identity of the HCP and similarity to the biotherapeutic and human proteins, as well as product, treatment-, and patient-related factors.
Collapse
Affiliation(s)
- Sherin Panikulam
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Hannah Morgan
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Thomas K Villiger
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
| | - Nicolas Lebesgue
- Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Anette C Karle
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
3
|
Abdeladhim M, Teixeira C, Ressner R, Hummer K, Dey R, Gomes R, de Castro W, de Araujo FF, Turiansky GW, Iniguez E, Meneses C, Oliveira F, Aronson N, Lacsina JR, Valenzuela JG, Kamhawi S. Lutzomyia longipalpis salivary proteins elicit human innate and adaptive immune responses detrimental to Leishmania parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640210. [PMID: 40196468 PMCID: PMC11974753 DOI: 10.1101/2025.02.25.640210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Leishmania parasites are transmitted via the bite of infected sand flies, whose saliva modulates host immune responses to promote Leishmania infection, especially in unexposed individuals. For humans in endemic areas, the immune consequences of chronic exposure to sand fly saliva remain poorly understood. We performed a human challenge study with Lutzomyia longipalpis, the primary vector of visceral leishmaniasis in the Americas. Fifteen healthy volunteers were exposed multiple times to uninfected Lu. longipalpis bites over the course of a year. PBMCs collected after several exposures were stimulated ex vivo by recombinant Lu. longipalpis salivary proteins to measure cytokine responses. Two salivary proteins, LJM19 and LJL143, elicited TH1-polarized cytokine responses, but with high co-expression of the TH2 cytokine IL-13. LJM19 also induced higher levels of IL-6 and IL-7, while both LJM19 and LJL143 induced the innate cytokines IL-1β and IFN-α. Importantly, TH1 polarization induced by LJM19 or LJL143 in PBMCs correlated with enhanced killing of Leishmania in co-cultured macrophages. Skin biopsies from two volunteers revealed bite site infiltration with CD4-CD8- T cells. Our data suggest that sand fly exposed individuals demonstrate robust innate and adaptive cellular immune responses to vector salivary proteins that can be co-opted to protect humans against Leishmania infection.
Collapse
Affiliation(s)
- Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Clarissa Teixeira
- Laboratory of Immunoparasitology, Department of Biotechnology, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Roseanne Ressner
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States
| | - Kelly Hummer
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Regis Gomes
- Laboratory of Immunoparasitology, Department of Biotechnology, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Waldionê de Castro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Fernanda Fortes de Araujo
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - George W. Turiansky
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Naomi Aronson
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - Joshua R. Lacsina
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| |
Collapse
|
4
|
Zhang Y, Lian C, Lai W, Jiang L, Xing Y, Liang H, Li J, Zhang X, Gan J, Li Z, Yin F. Programmable Stapling Peptide Based on Sulfonium as Universal Vaccine Adjuvants for Multiple Types of Vaccines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409567. [PMID: 39878394 PMCID: PMC11923873 DOI: 10.1002/advs.202409567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/22/2024] [Indexed: 01/31/2025]
Abstract
Adjuvants are non-specific immune enhancers commonly used to improve the responsiveness and persistence of the immune system toward antigens. However, due to the undefined chemical structure, toxicity, non-biodegradability, and lack of design technology in many existing adjuvants, it remains difficult to achieve substantive breakthroughs in the adjuvant research field. Here, a novel adjuvant development strategy based on stapling peptides is reported to overcome this challenge. The nano-vaccine incorporating peptide adjuvant and recombinant HBsAg protein not only induced strong antibody titers that are equivalent to aluminum adjuvanted vaccines but also simultaneously activated T-cell immune response. Similar results are also observed in herpes zoster vaccine and more complex influenza vaccine. The mechanism analysis demonstrates that antigen is efficiently carried into antigen-presenting cells (APCs) by peptide, further promoting the secretion of cytokines and activation of APCs. In addition, by redesigning the adjuvant, it is found that the sulfonium centers, rather than the sequence of peptide played an important role in immune activation. This discovery may provide a new paradigm for the rational design of peptide-based adjuvants. In brief, this study demonstrates that stapling peptides with sulfonium centers can provide a well-defined, programmable, biocompatible, and effective adjuvant for multiple types of vaccines.
Collapse
Affiliation(s)
- Yaping Zhang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, 518118, P. R. China
| | - Chenshan Lian
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Wenlong Lai
- Shenzhen Kangtai Biological Products Co. Ltd., Shenzhen, 518057, P. R. China
| | - Leying Jiang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Yun Xing
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
| | - Huiting Liang
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, 518118, P. R. China
| | - Jin Li
- Shenzhen Kangtai Biological Products Co. Ltd., Shenzhen, 518057, P. R. China
| | - Xinming Zhang
- Beijing Minhai Biotechnology Co. Ltd., Beijing, 102609, P. R. China
| | - Jianhui Gan
- Shenzhen Kangtai Biological Products Co. Ltd., Shenzhen, 518057, P. R. China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, 518118, P. R. China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, P. R. China
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, 518118, P. R. China
| |
Collapse
|
5
|
Ranjbar KJ, Sarkoohi P, Shahbazi B, Babaei M, Ahmadi K. Bioinformatics analysis of the in silico engineered protein vaccine with and without Escherichia coli heat labile enterotoxin adjuvant on the model of Klebsiella pneumoniae. Sci Rep 2025; 15:7321. [PMID: 40025224 PMCID: PMC11873140 DOI: 10.1038/s41598-025-91602-y] [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: 11/20/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae) has been identified as a major cause of nosocomial infections with multidrug-resistant phenotypes. Vaccination is one of the most effective methods to prevent infectious diseases. We aim to design a vaccine candidate based on the epitope-rich domains of the OmpA, OMPK17, and fimb proteins of K. pneumoniae that could protect against this infection. A vaccine structure was constructed by selecting five epitope-rich domains from three proteins. We decided to add the heat-labile toxin (LT) of Escherichia coli as an adjuvant to the designed protein structure. The evaluation of the vaccine candidates' interaction with the immune system's receptors showed an appropriate interaction of the specially adjuvated protein with TLR2 and TLR4. The stability of the interactions was also studied by molecular dynamics (MD) for to 100 ns. All parameters showed that the structure of the candidate proteins alone and in complex with TLR2 and TLR4 are stable, especially the adjuvanted protein. Immune response simulations showed that both candidates induce acceptable protective immune responses. Overall, the LT-adjuvanted design protein may have the potential to induce more favorable protective immune responses. However, further in vitro and in vivo studies are required to obtain more definitive results.
Collapse
Affiliation(s)
- Kimia Jafari Ranjbar
- Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parisa Sarkoohi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Behzad Shahbazi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Babaei
- Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Khadijeh Ahmadi
- Department of Medical Biotechnology, School of Paramedicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| |
Collapse
|
6
|
Hickey JM, Kumru OS, Dalençon F, Arvis F, Lutsch C, Joshi SB, Volkin DB. Two recombinant cytomegalovirus antigens formulated with the SPA14 adjuvant system: Impact of temperature, pH and excipients on the stability of each antigen and adjuvant component. J Pharm Sci 2025; 114:1224-1236. [PMID: 39864550 DOI: 10.1016/j.xphs.2024.12.030] [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: 09/26/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/28/2025]
Abstract
By evaluating the stability profiles of each component of a vaccine candidate (antigens, adjuvants), formulation conditions to mitigate vaccine instability can be identified. In this work, two recombinant Cytomegalovirus (CMV) glycoprotein antigens (gB, Pentamer) were formulated with SPA14, a novel liposome-based adjuvant system containing a synthetic TLR4 agonist (E6020) and a saponin (QS21). Analytical characterization and accelerated stability studies were performed with the two CMV antigens, formulated with and without SPA14, under various conditions (temperature, pH, excipients). For the antigens, the Pentamer was less stable than gB, and the addition of SPA14 adjuvant had negligible impact. For the SPA14 components, minor pH shifts (caused by the buffer's temperature dependent pKa shifts) destabilized the liposome (particle size by DLS) and QS21, but E6020 was unaffected (integrity by RP-UHPLC and LC-MS, respectively). The addition of chelators and free radical scavengers stabilized both the QS21 and E6020 components, consistent with oxidative degradation catalyzed by trace metal-ions. Interestingly, QS21 and E6020 also displayed improved storage stability in the presence of the protein antigens. These results are discussed in terms of developing key stability-indicating assays to optimize formulation conditions to stabilize the two CMV antigens and the three components of the SPA14 adjuvant system.
Collapse
Affiliation(s)
- John M Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Ozan S Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | | | - Florence Arvis
- Sanofi Vaccines R&D Department, 69280 Marcy-l'Étoile, France
| | - Charles Lutsch
- Sanofi Vaccines R&D Department, 69280 Marcy-l'Étoile, France
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
7
|
Ambari AM, Qhabibi FR, Desandri DR, Dwiputra B, Baravia PA, Makes IK, Radi B. Unveiling the Group A Streptococcus Vaccine-Based L-Rhamnose from Backbone of Group A Carbohydrate: Current Insight Against Acute Rheumatic Fever to Reduce the Global Burden of Rheumatic Heart Disease. F1000Res 2025; 13:132. [PMID: 39959434 PMCID: PMC11829149 DOI: 10.12688/f1000research.144903.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Group A Streptococcus (GAS) is a widely distributed bacterium that is Gram-positive and serves as the primary cause of acute rheumatic fever (ARF) episodes. Rheumatic heart disease (RHD) is a sequela resulting from repeated ARF attacks which are also caused by repeated GAS infections. ARF/RHD morbidity and mortality rates are incredibly high in low- and middle-income countries. This is closely related to poor levels of sanitation which causes the enhanced incidence of GAS infections. Management of carditis in RHD cases is quite challenging, particularly in developing countries, considering that medical treatment is only palliative, while definitive treatment often requires more invasive procedures with high costs. Preventive action through vaccination against GAS infection is one of the most effective steps as a solution in reducing RHD morbidity and mortality due to curative treatments are expensive. Various developments of M-protein-based GAS vaccines have been carried out over the last few decades and have recently begun to enter the clinical stage. Nevertheless, this vaccination generates cross-reactive antibodies that might trigger ARF assaults as a result of the resemblance between the M-protein structure and proteins found in many human tissues. Consequently, the development of a vaccine utilizing L-Rhamnose derived from the poly-rhamnose backbone of Group A Carbohydrate (GAC) commenced. The L-Rhamnose-based vaccine was chosen due to the absence of the Rhamnose biosynthesis pathway in mammalian cells including humans thus this molecule is not found in any body tissue. Recent pre-clinical studies reveal that L-Rhamnose-based vaccines provide a protective effect by increasing IgG antibody titers without causing cross-reactive antibodies in test animal tissue. These findings demonstrate that the L-Rhamnose-based vaccine possesses strong immunogenicity, which effectively protects against GAS infection while maintaining a significantly higher degree of safety.
Collapse
Affiliation(s)
- Ade Meidian Ambari
- Cardiovascular Prevention and Rehabilitation Department, National Cardiovascular Center Hospital Harapan Kita, Jakarta, Jakarta, 11420, Indonesia
- Cardiology and Vascular Department, Faculty of Medicine, University of Indonesia, Jakarta, Jakarta, 10430, Indonesia
| | - Faqrizal Ria Qhabibi
- Research Assistant, National Cardiovascular Center Hospital Harapan Kita, Jakarta, Jakarta, 11420, Indonesia
| | - Dwita Rian Desandri
- Cardiovascular Prevention and Rehabilitation Department, National Cardiovascular Center Hospital Harapan Kita, Jakarta, Jakarta, 11420, Indonesia
- Cardiology and Vascular Department, Faculty of Medicine, University of Indonesia, Jakarta, Jakarta, 10430, Indonesia
| | - Bambang Dwiputra
- Cardiovascular Prevention and Rehabilitation Department, National Cardiovascular Center Hospital Harapan Kita, Jakarta, Jakarta, 11420, Indonesia
- Cardiology and Vascular Department, Faculty of Medicine, University of Indonesia, Jakarta, Jakarta, 10430, Indonesia
| | - Pirel Aulia Baravia
- Cardiovascular Prevention and Rehabilitation Department, Dr. Saiful Anwar General Hospital, Malang, East Java, 65122, Indonesia
| | - Indira Kalyana Makes
- Research Assistant, National Cardiovascular Center Hospital Harapan Kita, Jakarta, Jakarta, 11420, Indonesia
| | - Basuni Radi
- Cardiovascular Prevention and Rehabilitation Department, National Cardiovascular Center Hospital Harapan Kita, Jakarta, Jakarta, 11420, Indonesia
- Cardiology and Vascular Department, Faculty of Medicine, University of Indonesia, Jakarta, Jakarta, 10430, Indonesia
| |
Collapse
|
8
|
da
Cruz Freire JE, Cardeal dos Santos AN, Coelho de Souza AN, de Oliveira A, Nicolete R, de Sousa BL, Martins da Silva JH, Gomes Vasconcelos YDA, da Silva ING, Soares PM, Guedes MIF, Ceccatto VM. Molecular and Immunological Properties of a Chimeric Glycosyl Hydrolase 18 Based on Immunoinformatics Approaches: A Design of a New Anti- Leishmania Vaccine. ACS Pharmacol Transl Sci 2025; 8:78-96. [PMID: 39816796 PMCID: PMC11729430 DOI: 10.1021/acsptsci.4c00341] [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: 06/04/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Leishmaniasis is a chronic inflammatory zoonotic illness caused by protozoan flagellates belonging to the Leishmania genus. Current data suggest that over 1 billion people worldwide are susceptible to infection, primarily in tropical and subtropical countries, where up to 2 million new cases are reported annually. Therefore, the development of a vaccine is crucial to combating this disease. This study employed immunoinformatics approaches to design a multiepitope anti-Leishmania vaccine, GH18-cpLeish, based on a cluster of six glycosyl hydrolases 18. We identified six helper T lymphocyte (HTL) epitopes and twenty-six cytotoxic T lymphocyte (CTL) epitopes with IC50 values <50 nM, indicating high affinity. Additionally, we also identified 20 continuous and twenty-six discontinuous B-cell epitopes. Analysis for allergenicity and toxicity showed no potential to induce these phenomena. All data obtained from in silico tools suggest that physicochemical and biological studies indicate that the GH18-cpLeish chimeric protein is a promising candidate for an anti-Leishmania vaccine. Docking analysis showed that the Pep1-cpLeish::TLR1, Pep1-cpLeish::TLR2, Pep1-cpLeish::/TLR3, and Pep1-cpLeish::/TLR4 complexes maintained a stable form. The best interaction cluster score was observed in the complex Pep1-cpLeish::TLR2 (center = -622.6 and lowest energy = -841.7 kcal.mol-1) followed by the complexes Pep1-cpLeish::TLR4 (center = -590.3 and lowest energy = -590.3 kcal.mol-1), Pep1-cpLeish::TLR3 (center = -589.1 and lowest energy = -657.0 kcal.mol-1), and Pep1-cpLeish::TLR1 (center = -504.1 and lowest energy = -602.9 kcal.mol-1), respectively. This study suggests that GH18-cpLeish may be suitable for constructing second-generation anti-Leishmania and even third-generation vaccines, given that its gene sequence is optimized for this purpose.
Collapse
Affiliation(s)
| | | | | | | | - Roberto Nicolete
- Oswaldo
Cruz Foundation (Fiocruz Ceará), Eusébio, Ceará 61773-270, Brazil
| | - Bruno Lopes de Sousa
- Superior
Institute of Biomedical Sciences, State
University of Ceará, Fortaleza, Ceará 60714-903, Brazil
| | | | | | - Isaac Neto Goes da Silva
- Department
of Veterinary Sciences, State University
of Ceará, Fortaleza, Ceará 60714-903, Brazil
| | - Paula Matias Soares
- Superior
Institute of Biomedical Sciences, State
University of Ceará, Fortaleza, Ceará 60714-903, Brazil
| | | | - Vânia Marilande Ceccatto
- Superior
Institute of Biomedical Sciences, State
University of Ceará, Fortaleza, Ceará 60714-903, Brazil
| |
Collapse
|
9
|
Chiumiento IR, Tricerri MA, Cortéz MF, Ituarte S, Tau J, Mariño KV, Smaldini PL, Heras H, Dreon MS. Pomacea canaliculata hemocyanin as a novel natural immunostimulant in mammals. Front Immunol 2025; 15:1490260. [PMID: 39845952 PMCID: PMC11750813 DOI: 10.3389/fimmu.2024.1490260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Gastropod hemocyanins are potent immunostimulants in mammals, a trait associated with their large molecular size and unusual glycosylation patterns. While the hemocyanin from the marine snail keyhole limpet (KLH), has been widely studied and successfully employed as a carrier/adjuvant in several immunological applications, as well as a non-specific immunostimulant for bladder cancer treatment, few other gastropod hemocyanins have been biochemically and immunologically characterized. In this work, we investigated the immunogenic properties of the hemocyanin from Pomacea canaliculata (PcH), an invasive south American freshwater snail. This species, known for its high reproductive rate and easy rearing, represents a promising source of potential biomedical compounds, including hemocyanin. Methods Employing flow cytometry, fluorescence microscopy, immunoassays, and quantitative PCR, we analysed the effects of PcH on THP-1 monocytes and their derived macrophages, as well as its ability to induce humoral response on C57BL/6 mice. Additionally, we evaluated the structural stability of PcH across a wide range of temperature and pH values. Results and discussion Our findings demonstrate that PcH is a structurally stable protein that not only triggers a pro-inflammatory effect on THP-1 derived-macrophages by increasing IL1-β and TNF-α levels, but also promotes phenotypic changes associated with the monocyte-to-macrophage differentiation. Moreover, the humoral response induced by PcH in mice was indistinguishable from that of KLH, highlighting the promising immunostimulatory properties of this freshwater snail hemocyanin.
Collapse
Affiliation(s)
- Ignacio Rafael Chiumiento
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
- Cátedra de Química Biológica, Facultad de Ciencias Naturales y Museo, UNLP, La Plata, Argentina
| | - María Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
- Cátedra de Bioquímica Clínica I, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| | - María Fernanda Cortéz
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
| | - Santiago Ituarte
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
- Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| | - Julia Tau
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
| | - Karina Valeria Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Paola Lorena Smaldini
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP - CONICET, La Plata, Argentina
| | - Horacio Heras
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
- Cátedra de Química Biológica, Facultad de Ciencias Naturales y Museo, UNLP, La Plata, Argentina
| | - Marcos Sebastián Dreon
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, (INIBIOLP), Universidad Nacional de La Plata (UNLP) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina
- Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| |
Collapse
|
10
|
Rai P, Mehrotra S, Prajapati VK. Exploring immunotherapy to control human infectious diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:389-429. [PMID: 39978973 DOI: 10.1016/bs.apcsb.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Infectious diseases continue to pose significant challenges to global health, especially with the rise of antibiotic resistance and emerging pathogens. Traditional treatments, while effective, are often limited in the face of rapidly evolving pathogens. Immunotherapy, which harnesses and enhances the body's immune response, offers a promising alternative to conventional approaches for the treatment of infectious diseases. By employing use of monoclonal antibodies, vaccines, cytokine therapies, and immune checkpoint inhibitors, immunotherapy has demonstrated considerable potential in overcoming treatment resistance and improving patient outcomes. Key innovations, including the development of mRNA vaccines, use of immune modulators, adoptive cell transfer, and chimeric antigen receptor (CAR)-T cell therapy are paving the way for more targeted pathogen clearance. Further, combining immunotherapy with conventional antibiotic treatment has demonstrated effectiveness against drug-resistant strains, but this chapter explores the evolving field of immunotherapy for the treatment of bacterial, viral, fungal, and parasitic infections. The chapter also explores the recent breakthroughs and ongoing clinical trials in infectious disease immunotherapy.
Collapse
Affiliation(s)
- Praveen Rai
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
11
|
Muraleedharan A, Acharya S, Kumar R. Recent Updates on Diverse Nanoparticles and Nanostructures in Therapeutic and Diagnostic Applications with Special Focus on Smart Protein Nanoparticles: A Review. ACS OMEGA 2024; 9:42613-42629. [PMID: 39464472 PMCID: PMC11500139 DOI: 10.1021/acsomega.4c05037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024]
Abstract
Nanomedicine enables advanced therapeutics, diagnostics, and predictive analysis, enhancing treatment outcomes and patient care. The choices and development of high-quality organic nanoparticles with relatively lower toxicity are important for achieving advanced medical goals. Among organic molecules, proteins have been prospected as smart candidates to revolutionize nanomedicine due to their inherent fascinating features. The advent of protein nanoarchitectures, which explore the biomolecular corona, offers new insights into their efficient tissue penetration and therapeutic potential. This review examines various animal- and plant-based protein nanoparticles, highlighting their source, activity, products, and unique biomedical applications in regenerative medicine, targeted therapies, gene and drug delivery, antimicrobial activity, bioimaging, immunological adjuvants, etc. It provides an extensive discussion on recent applications of protein nanoparticles across diverse biomedical fields as well as the evolving landscape of other nanoproducts and nanodevices for sensitive medical procedures. Furthermore, this review introduces different preparation technologies of protein nanoparticles, emphasizing how their design and construction significantly influence loading capacity, stability, and targeting effects. Additionally, we delve into the construction of different user-friendly multifunctional modular bioarchitectures by the assembly of protein nanoparticles (PNPs), marking a significant breakthrough in therapies. This review also considers the challenges of synthetic nanomaterials and the emergence of natural alternatives, which provides insights into protein nanoparticle research.
Collapse
Affiliation(s)
- Anju Muraleedharan
- Department
of Bioscience and Engineering, National
Institute of Technology Calicut, Kozhikode, Kerala, India, 673601
| | - Sarbari Acharya
- Department
of Life Science, School of Applied Sciences, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, India, 751024
| | - Ravindra Kumar
- Department
of Bioscience and Engineering, National
Institute of Technology Calicut, Kozhikode, Kerala, India, 673601
| |
Collapse
|
12
|
Banico EC, Sira EMJS, Fajardo LE, Dulay ANG, Odchimar NMO, Simbulan AM, Orosco FL. Advancing one health vaccination: In silico design and evaluation of a multi-epitope subunit vaccine against Nipah virus for cross-species immunization using immunoinformatics and molecular modeling. PLoS One 2024; 19:e0310703. [PMID: 39325755 PMCID: PMC11426463 DOI: 10.1371/journal.pone.0310703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
The resurgence of the Nipah virus (NiV) in 2023 has raised concerns for another potentially severe pandemic, given its history of high mortality from previous outbreaks. Unfortunately, no therapeutics and vaccines have been available for the virus. This study used immunoinformatics and molecular modeling to design and evaluate a multi-epitope subunit vaccine targeting NiV. The designed vaccine construct aims to stimulate immune responses in humans and two other intermediate animal hosts of the virus-swine and equine. Using several epitope prediction tools, ten peptides that induced B-lymphocyte responses, 17 peptides that induced cytotoxic T-lymphocyte (CTL) responses, and 12 peptides that induced helper T-lymphocyte (HTL) responses were mapped from nine NiV protein sequences. However, the CTL and HTL-inducing peptides were reduced to ten and eight, respectively, following molecular docking and dynamics. These screened peptides exhibited stability with 30 common major histocompatibility complex (MHC) receptors found in humans, swine, and equine. All peptides were linked using peptide linkers to form the multi-epitope construct and various adjuvants were tested to enhance its immunogenicity. The vaccine construct with resuscitation-promoting factor E (RpfE) adjuvant was selected as the final design based on its favorable physicochemical properties and superior immune response profile. Molecular docking was used to visualize the interaction of the vaccine to toll-like receptor 4 (TLR4), while molecular dynamics confirmed the structural stability of this interaction. Physicochemical property evaluation and computational simulations showed that the designed vaccine construct exhibited favorable properties and elicited higher antibody titers than the six multi-epitope NiV vaccine designs available in the literature. Further in vivo and in vitro experiments are necessary to validate the immunogenicity conferred by the designed vaccine construct and its epitope components. This study demonstrates the capability of computational methodologies in rational vaccine design and highlights the potential of cross-species vaccination strategies for mitigating potential NiV threats.
Collapse
Affiliation(s)
- Edward Coralde Banico
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Ella Mae Joy Sinco Sira
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Lauren Emily Fajardo
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Albert Neil Gura Dulay
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Nyzar Mabeth Obenio Odchimar
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Alea Maurice Simbulan
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Fredmoore Legaspi Orosco
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
- Department of Science and Technology, S&T Fellows Program, Taguig City, Metro Manila, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila City, Metro Manila, Philippines
| |
Collapse
|
13
|
Zhao H, Li W, Liu J, Li X, Ji H, Hu M, Li M. Label-Free Quantitative Proteomics Analysis of COVID-19 Vaccines by Nano LC-HRMS. Vaccines (Basel) 2024; 12:1055. [PMID: 39340085 PMCID: PMC11436057 DOI: 10.3390/vaccines12091055] [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: 07/03/2024] [Revised: 08/26/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
A nanoliter liquid chromatography-high resolution mass spectrometry-based method was developed for quantitative proteomics analysis of COVID-19 vaccines. It can be used for simultaneous qualitative and quantitative analysis of target proteins and host cell proteins (HCPs) in vaccine samples. This approach can directly provide protein information at the molecular level. Based on this, the proteomes of 15 batches of COVID-19 inactivated vaccine samples from two companies and 12 batches of COVID-19 recombinant protein vaccine samples from one company were successfully analyzed, which provided a significant amount of valuable information. Samples produced in different batches or by different companies can be systematically contrasted in this way, offering powerful supplements for existing quality standards. This strategy paves the way for profiling proteomics in complex samples and provides a novel perspective on the quality evaluation of bio-macromolecular drugs.
Collapse
Affiliation(s)
- Hengzhi Zhao
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drugs, Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing 102206, China
| | - Wendong Li
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drugs, Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing 102206, China
| | - Jingjing Liu
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drugs, Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing 102206, China
| | - Xiao Li
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drugs, Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing 102206, China
| | - Hong Ji
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drugs, Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing 102206, China
| | - Mo Hu
- Changping Laboratory, Beijing 102206, China
| | - Min Li
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drugs, Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing 102206, China
| |
Collapse
|
14
|
Sarkar R, Adhikary K, Banerjee A, Ganguly K, Sarkar R, Mohanty S, Dhua R, Bhattacharya K, Ahuja D, Pal S, Maiti R. Novel targets and improved immunotherapeutic techniques with an emphasis on antimycosal drug resistance for the treatment and management of mycosis. Heliyon 2024; 10:e35835. [PMID: 39224344 PMCID: PMC11367498 DOI: 10.1016/j.heliyon.2024.e35835] [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: 06/19/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Infections due to pathogenic fungi are endemic in particular area with increased morbidity and mortality. More than a thousand people are infected per year and the way of treatment is of high demand having a significant impact on the population health. Medical practitioners confront various troublesome analytic and therapeutical challenges in the administration of immunosuppressed sufferer at high danger of expanding fungal infections. An upgraded antimycosal treatment is fundamental for a fruitful result while treating intrusive mycoses. A collection of antimycosal drugs keeps on developing with their specific antifungal targets including cell membrane, mitochondria, cell wall, and deoxyribonucleic acid (DNA)/ribonucleic acid (RNA) or protein biosynthesis. Some fundamental classes of ordinarily directed medications are the polyenes, amphotericin B, syringomycin, allylamines, honokiol, azoles, flucytosine, echinocandins etc. However, few immunotherapy processes and vaccinations are being developed to mark this need, although one presently can't seem to arrive at the conclusion. In this review article, there has been a trial to give details upgradation about the current immune therapeutic techniques and vaccination strategies against prevention or treatment of mycosis as well as the difficulties related with their turn of events. There has been also a visualization in the mentioned review paper about the various assorted drugs and their specific target analysis along with therapeutic interventions.
Collapse
Affiliation(s)
- Riya Sarkar
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Adhikary
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Arundhati Banerjee
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Ganguly
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Riya Sarkar
- Department of Medical Laboratory Technology, Dr. B. C. Roy Academy of Professional Courses, Durgapur, West Bengal, 713206, India
| | - Satyajit Mohanty
- Department of Advanced Pharmacology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rumpa Dhua
- Department of Nutrition, Bankura Sammilani College, Kenduadihi, Bankura, West Bengal, 722102, India
| | - Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Deepika Ahuja
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Suchandra Pal
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal, 713209, India
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, 722101, India
| |
Collapse
|
15
|
Khalid K, Lim HX, Hwang JS, Poh CL. The Development of Epitope-Based Recombinant Protein Vaccines against SARS-CoV-2. AAPS J 2024; 26:93. [PMID: 39138686 DOI: 10.1208/s12248-024-00963-1] [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: 05/22/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024] Open
Abstract
The COVID-19 pandemic continues to cause infections and deaths, which are attributable to the SARS-CoV-2 Omicron variant of concern (VOC). Moderna's response to the declining protective efficacies of current SARS-CoV-2 vaccines against Omicron was to develop a bivalent booster vaccine based on the Spike (S) protein from the Wuhan and Omicron BA.4/BA.5 strains. This approach, while commendable, is unfeasible in light of rapidly emerging mutated viral strains. PubMed and Google Scholar were systematically reviewed for peer-reviewed papers up to January 2024. Articles included focused on specific themes such as the clinical history of recombinant protein vaccine development against different diseases, including COVID-19, the production of recombinant protein vaccines using different host expression systems, aspects to consider in recombinant protein vaccine development, and overcoming problems associated with large-scale recombinant protein vaccine production. In silico approaches to identify conserved and immunogenic epitopes could provide broad protection against SARS-CoV-2 VOCs but require validation in animal models. The recombinant protein vaccine development platform has shown a successful history in clinical development. Recombinant protein vaccines incorporating conserved epitopes may utilize a number of expression systems, such as yeast (Saccharomyces cerevisiae), baculovirus-insect cells (Sf9 cells), and Escherichia coli (E. coli). Current multi-epitope subunit vaccines against SARS-CoV-2 utilizing synthetic peptides are unfeasible for large-scale immunizations. Recombinant protein vaccines based on conserved and immunogenic proteins produced using E. coli offer high production yields, convenient purification, and cost-effective production of large-scale vaccine quantities capable of protecting against the SARS-CoV-2 D614G strain and its VOCs.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Petaling Jaya, Selangor, 47500, Malaysia
| | - Hui Xuan Lim
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Petaling Jaya, Selangor, 47500, Malaysia
| | - Jung Shan Hwang
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Petaling Jaya, Selangor, 47500, Malaysia
| | - Chit Laa Poh
- ALPS Global Holding Berhad, 1 Jalan 1/68F, Off Jalan Tun Razak, Kuala Lumpur, 50400, Malaysia.
| |
Collapse
|
16
|
Biswas R, Swetha RG, Basu S, Roy A, Ramaiah S, Anbarasu A. Designing multi-epitope vaccine against human cytomegalovirus integrating pan-genome and reverse vaccinology pipelines. Biologicals 2024; 87:101782. [PMID: 39003966 DOI: 10.1016/j.biologicals.2024.101782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/13/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) is accountable for high morbidity in neonates and immunosuppressed individuals. Due to the high genetic variability of HCMV, current prophylactic measures are insufficient. In this study, we employed a pan-genome and reverse vaccinology approach to screen the target for efficient vaccine candidates. Four proteins, envelope glycoprotein M, UL41A, US23, and US28, were shortlisted based on cellular localization, high solubility, antigenicity, and immunogenicity. A total of 29 B-cell and 44 T-cell highly immunogenic and antigenic epitopes with high global population coverage were finalized using immunoinformatics tools and algorithms. Further, the epitopes that were overlapping among the finalized B-cell and T-cell epitopes were linked with suitable linkers to form various combinations of multi-epitopic vaccine constructs. Among 16 vaccine constructs, Vc12 was selected based on physicochemical and structural properties. The docking and molecular simulations of VC12 were performed, which showed its high binding affinity (-23.35 kcal/mol) towards TLR4 due to intermolecular hydrogen bonds, salt bridges, and hydrophobic interactions, and there were only minimal fluctuations. Furthermore, Vc12 eliciting a good response was checked for its expression in Escherichia coli through in silico cloning and codon optimization, suggesting it to be a potent vaccine candidate.
Collapse
Affiliation(s)
- Rhitam Biswas
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Rayapadi G Swetha
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biosciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Soumya Basu
- Department of Biotechnology, NIST University, Berhampur, 761008, Odisha, India
| | - Aditi Roy
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biosciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
17
|
Muñoz SM, Vallejos-Baccelliere G, Manubens A, Salazar ML, Nascimento AFZ, Tapia-Reyes P, Meneses C, Ambrosio ALB, Becker MI, Guixé V, Castro-Fernandez V. Structural insights into a functional unit from an immunogenic mollusk hemocyanin. Structure 2024; 32:812-823.e4. [PMID: 38513659 DOI: 10.1016/j.str.2024.02.018] [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: 11/10/2023] [Revised: 01/30/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Mollusk hemocyanins, among the largest known proteins, are used as immunostimulants in biomedical and clinical applications. The hemocyanin of the Chilean gastropod Concholepas concholepas (CCH) exhibits unique properties, which makes it safe and effective for human immunotherapy, as observed in animal models of bladder cancer and melanoma, and dendritical cell vaccine trials. Despite its potential, the structure and amino acid sequence of CCH remain unknown. This study reports two sequence fragments of CCH, representing three complete functional units (FUs). We also determined the high-resolution (1.5 Å) X-ray crystal structure of an "FU-g type" from the CCHB subunit. This structure enables in-depth analysis of chemical interactions at the copper-binding center and unveils an unusual, truncated N-glycosylation pattern. These features are linked to eliciting more robust immunological responses in animals, offering insights into CCH's enhanced immunostimulatory properties and opening new avenues for its potential applications in biomedical research and therapies.
Collapse
Affiliation(s)
- Sebastián M Muñoz
- Laboratorio de Bioquímica y Biología Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 780003, Chile
| | - Gabriel Vallejos-Baccelliere
- Laboratorio de Bioquímica y Biología Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 780003, Chile
| | - Augusto Manubens
- Departamento de Investigación y Desarrollo, Biosonda Corp., Santiago 7750629, Chile; Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750629, Chile
| | - Michelle L Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750629, Chile
| | - Andrey F Z Nascimento
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo 13083-970, Brazil
| | - Patricio Tapia-Reyes
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomás, Santiago 8370003, Chile; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Claudio Meneses
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; Departamento de Fruticultura y Enología, Facultad de Agronomía y Sistemas Naturales, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; Millennium Nucleus Development of Super Adaptable Plants (MN-SAP), Santiago 8331150, Chile; Millennium Institute Center for Genome Regulation (CRG), Santiago 8331150, Chile
| | - Andre L B Ambrosio
- Sao Carlos Institute of Physics (IFSC), University of Sao Paulo (USP), Sao Carlos, Sao Paulo 13563-120, Brazil
| | - María Inés Becker
- Departamento de Investigación y Desarrollo, Biosonda Corp., Santiago 7750629, Chile; Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750629, Chile
| | - Victoria Guixé
- Laboratorio de Bioquímica y Biología Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 780003, Chile.
| | - Victor Castro-Fernandez
- Laboratorio de Bioquímica y Biología Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 780003, Chile.
| |
Collapse
|
18
|
Ji R, Guan L, Hu Z, Cheng Y, Cai M, Zhao G, Zang J. A comprehensive review on hemocyanin from marine products: Structure, functions, its implications for the food industry and beyond. Int J Biol Macromol 2024; 269:132041. [PMID: 38705315 DOI: 10.1016/j.ijbiomac.2024.132041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Hemocyanin, an oxygen-transport protein, is widely distributed in the hemolymph of marine arthropods and mollusks, playing an important role in their physiological processes. Recently, hemocyanin has been recognized as a multifunctional glycoprotein involved in the immunological responses of aquatic invertebrates. Consequently, the link between hemocyanin functions and their potential applications has garnered increased attention. This review offers an integrated overview of hemocyanin's structure, physicochemical characteristics, and bioactivities to further promote the utilization of hemocyanin derived from marine products. Specifically, we review its implication in two aspects of food and aquaculture industries: quality and health. Hemocyanin's inducible phenoloxidase activity is thought to be an inducer of melanosis in crustaceans. New anti-melanosis agents targeted to hemocyanin need to be explored. The red-color change observed in shrimp shells is related to hemocyanin, affecting consumer preferences. Hemocyanin's adaptive modification in response to the aquatic environment is available as a biomarker. Additionally, hemocyanin is endowed with bioactivities encompassing anti-microbial, antiviral, and therapeutic activities. Hemocyanin is also a novel allergen and its allergenic features remain incompletely characterized.
Collapse
Affiliation(s)
- Ruiyang Ji
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Leying Guan
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ziyan Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yishen Cheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Meng Cai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Guanghua Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jiachen Zang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
19
|
Liu Y, Lam DMK, Luan M, Zheng W, Ai H. Recent development of oral vaccines (Review). Exp Ther Med 2024; 27:223. [PMID: 38590568 PMCID: PMC11000446 DOI: 10.3892/etm.2024.12511] [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: 08/24/2023] [Accepted: 02/08/2024] [Indexed: 04/10/2024] Open
Abstract
Oral immunization can elicit an effective immune response and immune tolerance to specific antigens. When compared with the traditional injection route, delivering antigens via the gastrointestinal mucosa offers superior immune effects and compliance, as well as simplicity and convenience, making it a more optimal route for immunization. At present, various oral vaccine delivery systems exist. Certain modified bacteria, such as Salmonella, Escherichia coli and particularly Lactobacillus, are considered promising carriers for oral vaccines. These carriers can significantly enhance immunization efficiency by actively replicating in the intestinal tract following oral administration. The present review provided a discussion of the main mechanisms of oral immunity and the research progress made in the field of oral vaccines. Additionally, it introduced the advantages and disadvantages of the currently more commonly administered injectable COVID-19 vaccines, alongside the latest advancements in this area. Furthermore, recent developments in oral vaccines are summarized, and their potential benefits and side effects are discussed.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | | | - Mei Luan
- Department of Geriatric Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Wenfu Zheng
- Chinese Academy of Sciences Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Ai
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
20
|
Simbulan AM, Banico EC, Sira EMJS, Odchimar NMO, Orosco FL. Immunoinformatics-guided approach for designing a pan-proteome multi-epitope subunit vaccine against African swine fever virus. Sci Rep 2024; 14:1354. [PMID: 38228670 DOI: 10.1038/s41598-023-51005-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024] Open
Abstract
Despite being identified over a hundred years ago, there is still no commercially available vaccine for the highly contagious and deadly African swine fever virus (ASFV). This study used immunoinformatics for the rapid and inexpensive designing of a safe and effective multi-epitope subunit vaccine for ASFV. A total of 18,858 proteins from 100 well-annotated ASFV proteomes were screened using various computational tools to identify potential epitopes, or peptides capable of triggering an immune response in swine. Proteins from genotypes I and II were prioritized for their involvement in the recent global ASFV outbreaks. The screened epitopes exhibited promising qualities that positioned them as effective components of the ASFV vaccine. They demonstrated antigenicity, immunogenicity, and cytokine-inducing properties indicating their ability to induce potent immune responses. They have strong binding affinities to multiple swine allele receptors suggesting a high likelihood of yielding more amplified responses. Moreover, they were non-allergenic and non-toxic, a crucial prerequisite for ensuring safety and minimizing any potential adverse effects when the vaccine is processed within the host. Integrated with an immunogenic 50S ribosomal protein adjuvant and linkers, the epitopes formed a 364-amino acid multi-epitope subunit vaccine. The ASFV vaccine construct exhibited notable immunogenicity in immune simulation and molecular docking analyses, and stable profiles in secondary and tertiary structure assessments. Moreover, this study designed an optimized codon for efficient translation of the ASFV vaccine construct into the Escherichia coli K-12 expression system using the pET28a(+) vector. Overall, both sequence and structural evaluations suggested the potential of the ASFV vaccine construct as a candidate for controlling and eradicating outbreaks caused by the pathogen.
Collapse
Affiliation(s)
- Alea Maurice Simbulan
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Edward C Banico
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Ella Mae Joy S Sira
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Nyzar Mabeth O Odchimar
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Fredmoore L Orosco
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines.
- Department of Science and Technology, S&T Fellows Program, Bicutan, 1634, Taguig, Metro Manila, Philippines.
- Department of Biology, University of the Philippines Manila, 1000, Manila, Philippines.
| |
Collapse
|
21
|
Naveed M, Ali U, Aziz T, Jabeen K, Arif MH, Alharbi M, Alasmari AF, Albekairi TH. Development and immunological evaluation of an mRNA-based vaccine targeting Naegleria fowleri for the treatment of primary amoebic meningoencephalitis. Sci Rep 2024; 14:767. [PMID: 38191579 PMCID: PMC10774437 DOI: 10.1038/s41598-023-51127-8] [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: 11/21/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Abstract
More than 95% of patients fall victim to primary amoebic meningoencephalitis (PAM), a fatal disease attacking the central nervous system. Naegleria fowleri, a brain-eating microorganism, is PAM's most well-known pathogenic ameboflagellate. Despite the use of antibiotics, the fatality rate continues to rise as no clinical trials have been conducted against this disease. To address this, we mined the UniProt database for pathogenic proteins and selected assumed epitopes to create an mRNA-based vaccine. We identified thirty B-cell and T-cell epitopes for the vaccine candidate. These epitopes, secretion boosters, subcellular trafficking structures, and linkers were used to construct the vaccine candidate. Through predictive modeling and confirmation via the Ramachandran plot (with a quality factor of 92.22), we assessed secondary and 3D structures. The adjuvant RpfE was incorporated to enhance the vaccine construct's immunogenicity (GRAVY index: 0.394, instability index: 38.99, antigenicity: 0.8). The theoretical model of immunological simulations indicated favorable responses from both innate and adaptive immune cells, with memory cells expected to remain active for up to 350 days post-vaccination, while the antigen was eliminated from the body within 24 h. Notably, strong interactions were observed between the vaccine construct and TLR-4 (- 11.9 kcal/mol) and TLR-3 (- 18.2 kcal/mol).
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan.
| | - Urooj Ali
- Department of Biotechnology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47132, Arta, Greece.
| | - Khizra Jabeen
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan
| | - Muhammad Hammad Arif
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, 11451, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, 11451, Riyadh, Saudi Arabia
| | - Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Pfeifer BA, Beitelshees M, Hill A, Bassett J, Jones CH. Harnessing synthetic biology for advancing RNA therapeutics and vaccine design. NPJ Syst Biol Appl 2023; 9:60. [PMID: 38036580 PMCID: PMC10689799 DOI: 10.1038/s41540-023-00323-3] [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: 06/01/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023] Open
Abstract
Recent global events have drawn into focus the diversity of options for combatting disease across a spectrum of prophylactic and therapeutic approaches. The recent success of the mRNA-based COVID-19 vaccines has paved the way for RNA-based treatments to revolutionize the pharmaceutical industry. However, historical treatment options are continuously updated and reimagined in the context of novel technical developments, such as those facilitated through the application of synthetic biology. When it comes to the development of genetic forms of therapies and vaccines, synthetic biology offers diverse tools and approaches to influence the content, dosage, and breadth of treatment with the prospect of economic advantage provided in time and cost benefits. This can be achieved by utilizing the broad tools within this discipline to enhance the functionality and efficacy of pharmaceutical agent sequences. This review will describe how synthetic biology principles can augment RNA-based treatments through optimizing not only the vaccine antigen, therapeutic construct, therapeutic activity, and delivery vector. The enhancement of RNA vaccine technology through implementing synthetic biology has the potential to shape the next generation of vaccines and therapeutics.
Collapse
Affiliation(s)
- Blaine A Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | | | - Andrew Hill
- Pfizer, 66 Hudson Boulevard, New York, NY, 10001, USA
| | - Justin Bassett
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | | |
Collapse
|
23
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
24
|
Díaz-Dinamarca DA, Salazar ML, Escobar DF, Castillo BN, Valdebenito B, Díaz P, Manubens A, Salazar F, Troncoso MF, Lavandero S, Díaz J, Becker MI, Vásquez AE. Surface immunogenic protein from Streptococcus agalactiae and Fissurella latimarginata hemocyanin are TLR4 ligands and activate MyD88- and TRIF dependent signaling pathways. Front Immunol 2023; 14:1186188. [PMID: 37790926 PMCID: PMC10544979 DOI: 10.3389/fimmu.2023.1186188] [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: 03/14/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
The development of vaccine adjuvants is of interest for the management of chronic diseases, cancer, and future pandemics. Therefore, the role of Toll-like receptors (TLRs) in the effects of vaccine adjuvants has been investigated. TLR4 ligand-based adjuvants are the most frequently used adjuvants for human vaccines. Among TLR family members, TLR4 has unique dual signaling capabilities due to the recruitment of two adapter proteins, myeloid differentiation marker 88 (MyD88) and interferon-β adapter inducer containing the toll-interleukin-1 receptor (TIR) domain (TRIF). MyD88-mediated signaling triggers a proinflammatory innate immune response, while TRIF-mediated signaling leads to an adaptive immune response. Most studies have used lipopolysaccharide-based ligands as TLR4 ligand-based adjuvants; however, although protein-based ligands have been proven advantageous as adjuvants, their mechanisms of action, including their ability to undergo structural modifications to achieve optimal immunogenicity, have been explored less thoroughly. In this work, we characterized the effects of two protein-based adjuvants (PBAs) on TLR4 signaling via the recruitment of MyD88 and TRIF. As models of TLR4-PBAs, we used hemocyanin from Fissurella latimarginata (FLH) and a recombinant surface immunogenic protein (rSIP) from Streptococcus agalactiae. We determined that rSIP and FLH are partial TLR4 agonists, and depending on the protein agonist used, TLR4 has a unique bias toward the TRIF or MyD88 pathway. Furthermore, when characterizing gene products with MyD88 and TRIF pathway-dependent expression, differences in TLR4-associated signaling were observed. rSIP and FLH require MyD88 and TRIF to activate nuclear factor kappa beta (NF-κB) and interferon regulatory factor (IRF). However, rSIP and FLH have a specific pattern of interleukin 6 (IL-6) and interferon gamma-induced protein 10 (IP-10) secretion associated with MyD88 and TRIF recruitment. Functionally, rSIP and FLH promote antigen cross-presentation in a manner dependent on TLR4, MyD88 and TRIF signaling. However, FLH activates a specific TRIF-dependent signaling pathway associated with cytokine expression and a pathway dependent on MyD88 and TRIF recruitment for antigen cross-presentation. Finally, this work supports the use of these TLR4-PBAs as clinically useful vaccine adjuvants that selectively activate TRIF- and MyD88-dependent signaling to drive safe innate immune responses and vigorous Th1 adaptive immune responses.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
- Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Michelle L. Salazar
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Daniel F. Escobar
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
| | - Byron N. Castillo
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Bastián Valdebenito
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
| | - Pablo Díaz
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
| | | | - Fabián Salazar
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
- Investigación y Desarrollo, BIOSONDA S.A., Santiago, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Janepsy Díaz
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - María Inés Becker
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
- Investigación y Desarrollo, BIOSONDA S.A., Santiago, Chile
| | - Abel E. Vásquez
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
- Facultad de Ciencias de la Salud, Escuela de Medicina, Universidad del Alba, Santiago, Chile
| |
Collapse
|
25
|
Gupta S, Pellett S. Recent Developments in Vaccine Design: From Live Vaccines to Recombinant Toxin Vaccines. Toxins (Basel) 2023; 15:563. [PMID: 37755989 PMCID: PMC10536331 DOI: 10.3390/toxins15090563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
Vaccines are one of the most effective strategies to prevent pathogen-induced illness in humans. The earliest vaccines were based on live inoculations with low doses of live or related pathogens, which carried a relatively high risk of developing the disease they were meant to prevent. The introduction of attenuated and killed pathogens as vaccines dramatically reduced these risks; however, attenuated live vaccines still carry a risk of reversion to a pathogenic strain capable of causing disease. This risk is completely eliminated with recombinant protein or subunit vaccines, which are atoxic and non-infectious. However, these vaccines require adjuvants and often significant optimization to induce robust T-cell responses and long-lasting immune memory. Some pathogens produce protein toxins that cause or contribute to disease. To protect against the effects of such toxins, chemically inactivated toxoid vaccines have been found to be effective. Toxoid vaccines are successfully used today at a global scale to protect against tetanus and diphtheria. Recent developments for toxoid vaccines are investigating the possibilities of utilizing recombinant protein toxins mutated to eliminate biologic activity instead of chemically inactivated toxins. Finally, one of the most contemporary approaches toward vaccine design utilizes messenger RNA (mRNA) as a vaccine candidate. This approach was used globally to protect against coronavirus disease during the COVID-19 pandemic that began in 2019, due to its advantages of quick production and scale-up, and effectiveness in eliciting a neutralizing antibody response. Nonetheless, mRNA vaccines require specialized storage and transport conditions, posing challenges for low- and middle-income countries. Among multiple available technologies for vaccine design and formulation, which technology is most appropriate? This review focuses on the considerable developments that have been made in utilizing diverse vaccine technologies with a focus on vaccines targeting bacterial toxins. We describe how advancements in vaccine technology, combined with a deeper understanding of pathogen-host interactions, offer exciting and promising avenues for the development of new and improved vaccines.
Collapse
Affiliation(s)
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| |
Collapse
|
26
|
Al-Hawary SIS, Saleh EAM, Mamajanov NA, S Gilmanova N, Alsaab HO, Alghamdi A, Ansari SA, Alawady AHR, Alsaalamy AH, Ibrahim AJ. Breast cancer vaccines; A comprehensive and updated review. Pathol Res Pract 2023; 249:154735. [PMID: 37611432 DOI: 10.1016/j.prp.2023.154735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023]
Abstract
According to the International Agency for Research on Cancer, breast cancer is more common than lung cancer globally. By 2040, mortality from breast cancer will rise by 50% and 40%, respectively. Despite advances in chemotherapy, endocrine therapy, and HER2-targeted therapy, breast cancer metastases and recurrences remain challenging to treat. Cancer vaccines are an effective treatment option because they stimulate a long-lasting immune response that will eliminate tumor cells. In studies on the breast cancer vaccine, no appreciable advantages were discovered. A recent study claims that immune checkpoint inhibitors or anti-HER2 monoclonal antibodies may be used in vaccinations. This vaccination strengthens the immune system to fight off breast cancer cells. Clinical trials have been conducted on DNA, dendritic cells, and peptide-based breast cancer vaccines. Studies on the breast cancer vaccine have employed subcutaneous, intramuscular, and intradermal injections. Clinical studies have shown that these efforts have not been successful. Several factors might have slowed the development of a breast cancer vaccine. The complexity of the immune system makes it challenging to create cancer vaccines. Given the heterogeneity of breast cancer, there may be a need for different vaccination strategies. Despite these obstacles, research into breast cancer vaccines continues. Effective methods for creating vaccines include immune checkpoint inhibition and anti-HER2 monoclonal antibodies. Research is also being done on specialized tumor vaccinations.
Collapse
Affiliation(s)
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, Prince Sattam Bin Abdulaziz University, College of Arts and Science, Wadi Al-Dawasir 11991, Saudi Arabia
| | - Nodirjon Akhmetovich Mamajanov
- Teaching Assistant, MD, Department of Public Health, Healthcare Management and Physical Culture, Tashkent State Dental Institute, Tashkent, Uzbekistan; Research scholar, Department of Scientific Affairs, Samarkand State Medical Institute, Samarkand, Uzbekistan
| | - Nataliya S Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| | - Adel Alghamdi
- Preparatory Year Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Shakeel Ahmed Ansari
- Department of Biochemistry, General Medicine Practice Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Ahmed Hussien Radie Alawady
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Hashiem Alsaalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | | |
Collapse
|
27
|
Díaz-Dinamarca DA, Díaz P, Barra G, Puentes R, Arata L, Grossolli J, Riveros-Rodriguez B, Ardiles L, Santelises J, Vasquez-Saez V, Escobar DF, Soto D, Canales C, Díaz J, Lamperti L, Castillo D, Urra M, Zuñiga F, Ormazabal V, Nova-Lamperti E, Benítez R, Rivera A, Cortes CP, Valenzuela MT, García-Escorza HE, Vasquez AE. Humoral immunity against SARS-CoV-2 evoked by heterologous vaccination groups using the CoronaVac (Sinovac) and BNT162b2 (Pfizer/BioNTech) vaccines in Chile. Front Public Health 2023; 11:1229045. [PMID: 37693706 PMCID: PMC10483147 DOI: 10.3389/fpubh.2023.1229045] [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: 05/25/2023] [Accepted: 07/27/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Severe acute respiratory syndrome virus 2 (SARS-CoV-2) has caused over million deaths worldwide, with more than 61,000 deaths in Chile. The Chilean government has implemented a vaccination program against SARS-CoV-2, with over 17.7 million people receiving a complete vaccination scheme. The final target is 18 million individuals. The most common vaccines used in Chile are CoronaVac (Sinovac) and BNT162b2 (Pfizer-Biotech). Given the global need for vaccine boosters to combat the impact of emerging virus variants, studying the immune response to SARS-CoV-2 is crucial. In this study, we characterize the humoral immune response in inoculated volunteers from Chile who received vaccination schemes consisting of two doses of CoronaVac [CoronaVac (2x)], two doses of CoronaVac plus one dose of BNT162b2 [CoronaVac (2x) + BNT162b2 (1x)], and three doses of BNT162b2 [BNT162b2 (3x)]. Methods We recruited 469 participants from Clínica Dávila in Santiago and the Health Center Víctor Manuel Fernández in the city of Concepción, Chile. Additionally, we included participants who had recovered from COVID-19 but were not vaccinated (RCN). We analyzed antibodies, including anti-N, anti-S1-RBD, and neutralizing antibodies against SARS-CoV-2. Results We found that antibodies against the SARS-CoV-2 nucleoprotein were significantly higher in the CoronaVac (2x) and RCN groups compared to the CoronaVac (2x) + BNT162b2 (1x) or BNT162b2 (3x) groups. However, the CoronaVac (2x) + BNT162b2 (1x) and BNT162b2 (3x) groups exhibited a higher concentration of S1-RBD antibodies than the CoronaVac (2x) group and RCN group. There were no significant differences in S1-RBD antibody titers between the CoronaVac (2x) + BNT162b2 (1x) and BNT162b2 (3x) groups. Finally, the group immunized with BNT162b2 (3x) had higher levels of neutralizing antibodies compared to the RCN group, as well as the CoronaVac (2x) and CoronaVac (2x) + BNT162b2 (1x) groups. Discussion These findings suggest that vaccination induces the secretion of antibodies against SARS-CoV-2, and a booster dose of BNT162b2 is necessary to generate a protective immune response. In the current state of the pandemic, these data support the Ministry of Health of the Government of Chile's decision to promote heterologous vaccination as they indicate that a significant portion of the Chilean population has neutralizing antibodies against SARS-CoV-2.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Pablo Díaz
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Gisselle Barra
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Rodrigo Puentes
- Sección gestión de la información, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Loredana Arata
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jonnathan Grossolli
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Boris Riveros-Rodriguez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Luis Ardiles
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Julio Santelises
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
- Tecnología Medica, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Universidad del Desarrollo, Santiago, Chile
| | - Valeria Vasquez-Saez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Daniel F. Escobar
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Daniel Soto
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Cecilia Canales
- Sección gestión de la información, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Janepsy Díaz
- Sección gestión de la información, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Liliana Lamperti
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Daniela Castillo
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Mychel Urra
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Felipe Zuñiga
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Valeska Ormazabal
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Estefanía Nova-Lamperti
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Rosana Benítez
- Unidad de investigación Clínica, Clínica Dávila, Santiago, Chile
| | - Alejandra Rivera
- Unidad de investigación Clínica, Clínica Dávila, Santiago, Chile
| | - Claudia P. Cortes
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Clínica Santa María, Santiago, Chile
| | | | | | - Abel E. Vasquez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
- Tecnología Medica, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Universidad del Desarrollo, Santiago, Chile
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| |
Collapse
|
28
|
Zeng Z, Geng X, Wen X, Chen Y, Zhu Y, Dong Z, Hao L, Wang T, Yang J, Zhang R, Zheng K, Sun Z, Zhang Y. Novel receptor, mutation, vaccine, and establishment of coping mode for SARS-CoV-2: current status and future. Front Microbiol 2023; 14:1232453. [PMID: 37645223 PMCID: PMC10461067 DOI: 10.3389/fmicb.2023.1232453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Since the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its resultant pneumonia in December 2019, the cumulative number of infected people worldwide has exceeded 670 million, with over 6.8 million deaths. Despite the marketing of multiple series of vaccines and the implementation of strict prevention and control measures in many countries, the spread and prevalence of SARS-CoV-2 have not been completely and effectively controlled. The latest research shows that in addition to angiotensin converting enzyme II (ACE2), dozens of protein molecules, including AXL, can act as host receptors for SARS-CoV-2 infecting human cells, and virus mutation and immune evasion never seem to stop. To sum up, this review summarizes and organizes the latest relevant literature, comprehensively reviews the genome characteristics of SARS-CoV-2 as well as receptor-based pathogenesis (including ACE2 and other new receptors), mutation and immune evasion, vaccine development and other aspects, and proposes a series of prevention and treatment opinions. It is expected to provide a theoretical basis for an in-depth understanding of the pathogenic mechanism of SARS-CoV-2 along with a research basis and new ideas for the diagnosis and classification, of COVID-19-related disease and for drug and vaccine research and development.
Collapse
Affiliation(s)
- Zhaomu Zeng
- Department of Neurosurgery, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital of Central South University, National Regional Medical Center for Nervous System Diseases, Nanchang, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Xiuchao Geng
- Department of Nursing, School of Medicine, Taizhou University, Taizhou, China
| | - Xichao Wen
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Yixi Zhu
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zishu Dong
- Department of Zoology, Advanced Research Institute, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liangchao Hao
- Department of Plastic Surgery, Shaoxing People’s Hospital, Shaoxing, China
| | - Tingting Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Jifeng Yang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Ruobing Zhang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Kebin Zheng
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Yuhao Zhang
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
29
|
Han S, Lee P, Choi HJ. Non-Invasive Vaccines: Challenges in Formulation and Vaccine Adjuvants. Pharmaceutics 2023; 15:2114. [PMID: 37631328 PMCID: PMC10458847 DOI: 10.3390/pharmaceutics15082114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Given the limitations of conventional invasive vaccines, such as the requirement for a cold chain system and trained personnel, needle-based injuries, and limited immunogenicity, non-invasive vaccines have gained significant attention. Although numerous approaches for formulating and administrating non-invasive vaccines have emerged, each of them faces its own challenges associated with vaccine bioavailability, toxicity, and other issues. To overcome such limitations, researchers have created novel supplementary materials and delivery systems. The goal of this review article is to provide vaccine formulation researchers with the most up-to-date information on vaccine formulation and the immunological mechanisms available, to identify the technical challenges associated with the commercialization of non-invasive vaccines, and to guide future research and development efforts.
Collapse
Affiliation(s)
| | | | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada; (S.H.); (P.L.)
| |
Collapse
|
30
|
Rhee JH, Khim K, Puth S, Choi Y, Lee SE. Deimmunization of flagellin adjuvant for clinical application. Curr Opin Virol 2023; 60:101330. [PMID: 37084463 DOI: 10.1016/j.coviro.2023.101330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Flagellin is the cognate ligand for host pattern recognition receptors, toll-like receptor 5 (TLR5) in the cell surface, and NAIP5/NLRC4 inflammasome in the cytosol. TLR5-binding domain is located in D1 domain, where crucial amino acid sequences are conserved among diverse bacteria. The highly conserved C-terminal 35 amino acids of flagellin were proved to be responsible for the inflammasome activation by binding to NAIP5. D2/D3 domains, located in the central region and exposed to the outside surface of flagellar filament, are heterogeneous across bacterial species and highly immunogenic. Taking advantage of TLR5- and NLRC4-stimulating activities, flagellin has been actively developed as a vaccine adjuvant and immunotherapeutic. Because of its immunogenicity, there exist worries concerning diminished efficacy and possible reactogenicity after repeated administration. Deimmunization of flagellin derivatives while preserving the TLR5/NLRC4-mediated immunomodulatory activity should be the most reasonable option for clinical application. This review describes strategies and current achievements in flagellin deimmunization.
Collapse
Affiliation(s)
- Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea.
| | - Koemchhoy Khim
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Sao Puth
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea; Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| |
Collapse
|
31
|
Matsuzaka Y, Yashiro R. Extracellular Vesicle-Based SARS-CoV-2 Vaccine. Vaccines (Basel) 2023; 11:vaccines11030539. [PMID: 36992123 DOI: 10.3390/vaccines11030539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/06/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Messenger ribonucleic acid (RNA) vaccines are mainly used as SARS-CoV-2 vaccines. Despite several issues concerning storage, stability, effective period, and side effects, viral vector vaccines are widely used for the prevention and treatment of various diseases. Recently, viral vector-encapsulated extracellular vesicles (EVs) have been suggested as useful tools, owing to their safety and ability to escape from neutral antibodies. Herein, we summarize the possible cellular mechanisms underlying EV-based SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, The Institute of Medical Science, Center for Gene and Cell Therapy, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8551, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8551, Japan
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| |
Collapse
|