1
|
Guan P, Qi C, Xu G, Sheng C, Sun S, Zhou Z, Jia S. Designing a T cell multi-epitope vaccine against hRSV with reverse vaccinology: An immunoinformatics approach. Colloids Surf B Biointerfaces 2025; 251:114599. [PMID: 40031111 DOI: 10.1016/j.colsurfb.2025.114599] [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: 12/14/2024] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
As an infectious viral pathogen, human respiratory syncytial virus (hRSV) can cause severe respiratory infections and is recognized as one of the highest priority pathogens by the World Health Organization (WHO). Although vaccines play an important role in disease prevention and transmission, the wild-type virus is usually prone to immune escape due to the relatively high mutation rate of biological proteins. Therefore, designing a broad-spectrum hRSV vaccine is essential to provide extensive protection against multiple viral variants. Using a consensus sequence approach, we designed a broad-spectrum T-cell epitope vaccine composed of 385 amino acids, consisting of 12 CTLs and 5 HTLs from the fusion protein and glycoprotein. The designed multi-epitope vaccine was expected to have non-allergenicity, high population coverage, strong antigenicity and immunogenicity, appropriate physical and chemical properties, and high solubility. Meanwhile, the structure of the vaccine had a high similarity to that of the natural virus. In addition, through structural biology analysis, the constructed vaccine achieved robust structural compactness and binding stability. Computer-generated immunological simulations indicated that the vaccine could elicit realistic immune responses in humans. The designed vaccine showed good binding affinity and molecular and immune simulation. In conclusion, the broad-spectrum hRSV vaccine could be an excellent candidate for preventing hRSV infection. The employed prediction pipeline was proved to be an efficient method for screening immunogenic epitopes of additional pathogens.
Collapse
Affiliation(s)
- Peibin Guan
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Congyan Qi
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Guojin Xu
- National Institute of Biological Science (NIBS),Beijing 102206, China
| | - Can Sheng
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining 272004, China
| | - Siqi Sun
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China
| | - Zhicheng Zhou
- Hubei Provincial Key Laboratory of Yeast Function, Angel Yeast Co., Ltd, Yichang 443003, China
| | - Shulei Jia
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
2
|
Nguyen KA, Liu Z, Davies JS, McIntosh CP, Draper LM, Norberg SM, Rae Z, Achar SR, Altan-Bonnet G, Zhang L, Wu X, Meyer TJ, Kelly MC, Taylor N, Hinrichs CS, Ishii K. CD22 TCR-engineered T cells exert antileukemia cytotoxicity without causing inflammatory responses. SCIENCE ADVANCES 2025; 11:eadq4297. [PMID: 40203088 PMCID: PMC11980841 DOI: 10.1126/sciadv.adq4297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Chimeric antigen receptor (CAR) T cells effectively treat B cell malignancies. However, CAR-T cells cause inflammatory toxicities such as cytokine release syndrome (CRS), which is in contrast to T cell receptor (TCR)-engineered T cells against various antigens that historically have rarely been associated with CRS. To study whether and how differences in receptor types affect the propensity for eliciting inflammatory responses in a model system wherein TCR and CAR target equalized sources of clinically relevant antigen, we discovered a CD22-specific TCR and compared it to CD22 CAR. Both CD22 TCR-T and CD22 CAR-T cells eradicated leukemia in xenografts, but only CD22 CAR-T cells induced dose-dependent systemic inflammation. Compared to TCR-T cells, CAR-T cells disproportionately upregulated inflammatory pathways without concordant augmentation in pathways involved in direct cytotoxicity upon antigen engagement. These differences in antileukemia responses comparing TCR-T and CAR-T cells highlight the potential opportunity to improve therapeutic safety by using TCRs.
Collapse
MESH Headings
- Humans
- Animals
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Sialic Acid Binding Ig-like Lectin 2/genetics
- Sialic Acid Binding Ig-like Lectin 2/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Mice
- Inflammation/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Leukemia/therapy
- Leukemia/immunology
- Leukemia/pathology
- Cytotoxicity, Immunologic
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kilyna A. Nguyen
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zhihui Liu
- Pediatric Oncology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | - John S. Davies
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA, USA
| | - Crystal P. McIntosh
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lindsey M. Draper
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Scott M. Norberg
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zachary Rae
- Single Cell Analysis Facility, CCR, NCI, NIH, Bethesda, MD, USA
| | - Sooraj R. Achar
- Laboratory of Integrative Cancer Immunology, CCR, NCI, NIH, Bethesda, MD, USA
| | | | - Ling Zhang
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xiaolin Wu
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, NCI, NIH, Frederick, MD, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, CCR, NCI, NIH, Bethesda, MD, USA
| | | | - Naomi Taylor
- Pediatric Oncology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | - Christian S. Hinrichs
- Duncan and Nancy MacMillan Center of Excellence in Cancer Immunotherapy and Metabolism, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Kazusa Ishii
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
3
|
Nasiri MJ, Rogowski L, Venketaraman V. In Silico Targeting and Immunological Profiling of PpiA in Mycobacterium tuberculosis: A Computational Approach. Pathogens 2025; 14:370. [PMID: 40333132 PMCID: PMC12030337 DOI: 10.3390/pathogens14040370] [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: 03/14/2025] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of mortality, with drug resistance highlighting the need for new vaccine targets. Peptidyl-prolyl isomerase A (PpiA), a conserved Mycobacterium tuberculosis (Mtb) protein, plays a role in bacterial stress adaptation and immune evasion, making it a potential target for immunotherapy. This study uses computational methods to assess PpiA's antigenicity, structural integrity, and immunogenic potential. The PpiA sequence was retrieved from NCBI and analyzed for antigenicity and allergenicity using VaxiJen, AllerTOP, and AllergenFP. Physicochemical properties were evaluated using ProtParam, and structural models were generated through PSIPRED and SWISS-MODEL. Structural validation was performed with MolProbity, QMEANDisCo, and ProSA-Web. B-cell epitopes were predicted using BepiPred 2.0 and IEDB, while T-cell epitopes were mapped via IEDB's MHC-I and MHC-II tools. Epitope conservation across Mtb strains was confirmed using ConSurf. Results indicate PpiA is highly antigenic, non-allergenic, and stable, with several immunogenic epitopes identified for both B- and T-cells. This study supports PpiA as a promising immunogenic target for TB vaccine development.
Collapse
Affiliation(s)
- Mohammad J. Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-69411, Iran;
| | - Lily Rogowski
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA;
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA;
| |
Collapse
|
4
|
Teh-Poot CF, Alfaro-Chacón A, Pech-Pisté LM, Rosado-Vallado ME, Asojo OA, Villanueva-Lizama LE, Dumonteil E, Cruz-Chan JV. Immunogenicity of Trypanosoma cruzi Multi-Epitope Recombinant Protein as an Antigen Candidate for Chagas Disease Vaccine in Humans. Pathogens 2025; 14:342. [PMID: 40333154 PMCID: PMC12030589 DOI: 10.3390/pathogens14040342] [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: 02/24/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi (T. cruzi), is the most significant neglected tropical disease affecting individuals in the Americas. Currently, available drugs, such as nifurtimox and benznidazole (BZN), are both toxic and ineffective in the chronic phase of the disease. A promising alternative is the development of a Chagas disease vaccine, although this effort is hampered by the complexity of the parasite and HLA polymorphisms. In addition, the activation of epitope-specific CD8+ T cells is critical to conferring a robust cell-mediated immune response and protection by producing IFN-γ and perforin. Thus, the antigen (s) for the development of a Chagas vaccine or immunotherapy must include CD8+ T cell epitopes. In this study, we aimed to develop a multi-epitope recombinant protein as a novel human vaccine for Chagas disease. Sixteen database programs were used to predict de novo 40 potential epitopes for the HLA-A*02:01 allele. Nine out of the 40 predicted epitopes were able to elicit IFN-γ production in Peripheral Blood Mononuclear Cells (PBMCs) from Chagas patients. Molecular docking revealed a good binding affinity among the epitopes with diverse HLA molecules. Therefore, a recombinant multi-epitope protein including these nine T. cruzi CD8+ epitopes was expressed and demonstrated to recall an antigen-specific immune response in ex-vivo assays using PBMCs from Chagas patients with the HLA-A*02 allele. These findings support the development of this multi-epitope protein as a promising candidate human vaccine against Chagas disease.
Collapse
Affiliation(s)
- Christian F. Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Mérida 97000, Mexico
| | - Andrea Alfaro-Chacón
- Laboratorio de Parasitología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Mérida 97000, Mexico
| | - Landy M. Pech-Pisté
- Laboratorio de Parasitología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Mérida 97000, Mexico
| | - Miguel E. Rosado-Vallado
- Laboratorio de Parasitología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Mérida 97000, Mexico
| | | | - Liliana E. Villanueva-Lizama
- Laboratorio de Parasitología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Mérida 97000, Mexico
| | - Eric Dumonteil
- Department of Tropical Medicine and Infectious Disease, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Mérida 97000, Mexico
| |
Collapse
|
5
|
Masum MHU, Mahdeen AA, Barua A. Revolutionizing Chikungunya Vaccines: mRNA Breakthroughs With Molecular and Immune Simulations. Bioinform Biol Insights 2025; 19:11779322251324859. [PMID: 40182080 PMCID: PMC11967231 DOI: 10.1177/11779322251324859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/14/2025] [Indexed: 04/05/2025] Open
Abstract
With the ability to cause massive epidemics that have consequences on millions of individuals globally, the Chikungunya virus (CHIKV) emerges as a severe menace. Developing an effective vaccine is urgent as no effective therapeutics are available for such viral infections. Therefore, we designed a novel mRNA vaccine against CHIKV with a combination of highly antigenic and potential MHC-I, MHC-II, and B-cell epitopes from the structural polyprotein. The vaccine demonstrated well-characterized physicochemical properties, indicating its solubility and potential functional stability within the body (GRAVY score of -0.639). Structural analyses of the vaccine revealed a well-stabilized secondary and tertiary structure (Ramachandran score of 82.8% and a Z-score of -4.17). Docking studies of the vaccine with TLR-2 (-1027.7 KJ/mol) and TLR-4 (-1212.4 KJ/mol) exhibited significant affinity with detailed hydrogen bond interactions. Molecular dynamics simulations highlighted distinct conformational dynamics among the vaccine, "vaccine-TLR-2" and "vaccine-TLR-4" complexes. The vaccine's ability to elicit both innate and adaptive immune responses, including the presence of memory B-cells and T-cells, persistent B-cell immunity for a year, and the activation of TH cells leading to the release of IFN-γ and IL-2, has significant implications for its potential effectiveness. The CHIKV vaccine developed in this study shows promise as a potential candidate for future vaccine production against CHIKV, suggesting its suitability for further clinical advancement, including in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md. Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chattogram, Bangladesh
| | - Ahmad Abdullah Mahdeen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Abanti Barua
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
6
|
Li J, Ju Y, Jiang M, Li S, Yang XY. Epitope-Based Vaccines: The Next Generation of Promising Vaccines Against Bacterial Infection. Vaccines (Basel) 2025; 13:248. [PMID: 40266107 PMCID: PMC11946261 DOI: 10.3390/vaccines13030248] [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: 01/28/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/24/2025] Open
Abstract
The increasing resistance of bacteria to antibiotics has underscored the need for new drugs or vaccines to prevent bacterial infections. Reducing multidrug resistance is a key objective of the WHO's One Health initiative. Epitopes, the key parts of antigen molecules that determine their specificity, directly stimulate the body to produce specific humoral and/or cellular immune responses. Epitope-based vaccines, which combine dominant epitopes in a rational manner, induce a more efficient and specific immune response than the original antigen. While these vaccines face significant challenges, such as epitope escape or low immunogenicity, they offer advantages including minimal adverse reactions, improved efficacy, and optimized protection. As a result, epitope-based vaccines are considered a promising next-generation approach to combating bacterial infections. This review summarizes the latest advancements, challenges, and future prospects of epitope-based vaccines targeting bacteria, with a focus on their development workflow and application in antibiotic-resistant pathogens with high mortality rates, including Staphylococcus aureus, Streptococcus pneumoniae, Streptococcus pyogenes, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa. The goal of this review is to provide insights into next-generation vaccination strategies to combat bacterial infections associated with antibiotic resistance and high mortality rates.
Collapse
Affiliation(s)
| | | | | | | | - Xiao-Yan Yang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China; (J.L.)
| |
Collapse
|
7
|
Sarfraz A, Chaudhary I, Arshad F, Shehroz M, Perveen A, Nishan U, Ali A, Ullah R, Shahat AA, Zaman A, Shah M. Peptide-based vaccine design against Hendra virus through immunoinformatics approach. Vet Immunol Immunopathol 2025; 280:110869. [PMID: 39752846 DOI: 10.1016/j.vetimm.2024.110869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025]
Abstract
The Hendra virus (HeV) has resulted in epidemics of respiratory and neurological illnesses in animals. Humans have contracted diseases with high fatality rates as a result of infected domestic animals, but effective vaccinations and therapies are currently not available against HeV. Herein, we analyzed the proteome of HeV and constructed an effective and innovative multi-epitope vaccine using immunoinformatics techniques. The vaccine construct was generated, targeting one matrix protein, with the help of the five selected B and T cell epitopes, linkers, and adjuvants and evaluated for their immunogenic properties. In-silico analysis revealed that the epitopes were able to interact with immune receptors and had high antigenic qualities. The post-translational modifications (PTMs), globular, disordered regions, and the active site of the vaccine were predicted, and the strong interactions between the vaccine and Toll-like receptor 5 were observed in molecular docking, indicating their potential significance in the immune response to the designed vaccine. The structural and dynamic stability of the vaccine were ensured by the molecular dynamic simulations. The results of the immune simulations indicated that the designed vaccine might activate B and T cells, which produce high levels of antibodies and cytokines to fight HeV infection. The developed vaccine is useful due to its non-toxicity, non-sensitization, good immunogenicity, non-allergic, and antigenic properties, accessed by various tools; however, experimental verification is needed to confirm the findings of the current study.
Collapse
Affiliation(s)
- Asifa Sarfraz
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Irfa Chaudhary
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Fizza Arshad
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Muhammad Shehroz
- Department of Bioinformatics, Kohsar University Murree, Murree 47150, Pakistan
| | - Asia Perveen
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Umar Nishan
- Department of Chemistry, Kohat University of Science & Technology, Kohat, Pakistan
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaaty A Shahat
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aqal Zaman
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan; Department of Microbiology and Molecular Genetics, Bahauddin Zakariya University, Multan 66000, Pakistan.
| | - Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan.
| |
Collapse
|
8
|
Imani S, Li X, Chen K, Maghsoudloo M, Jabbarzadeh Kaboli P, Hashemi M, Khoushab S, Li X. Computational biology and artificial intelligence in mRNA vaccine design for cancer immunotherapy. Front Cell Infect Microbiol 2025; 14:1501010. [PMID: 39902185 PMCID: PMC11788159 DOI: 10.3389/fcimb.2024.1501010] [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: 09/24/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Messenger RNA (mRNA) vaccines offer an adaptable and scalable platform for cancer immunotherapy, requiring optimal design to elicit a robust and targeted immune response. Recent advancements in bioinformatics and artificial intelligence (AI) have significantly enhanced the design, prediction, and optimization of mRNA vaccines. This paper reviews technologies that streamline mRNA vaccine development, from genomic sequencing to lipid nanoparticle (LNP) formulation. We discuss how accurate predictions of neoantigen structures guide the design of mRNA sequences that effectively target immune and cancer cells. Furthermore, we examine AI-driven approaches that optimize mRNA-LNP formulations, enhancing delivery and stability. These technological innovations not only improve vaccine design but also enhance pharmacokinetics and pharmacodynamics, offering promising avenues for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Xiaoyan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Keyi Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Xiaoping Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Rahman MM, Masum MHU, Parvin R, Das SC, Talukder A. Designing of an mRNA vaccine against high-risk human papillomavirus targeting the E6 and E7 oncoproteins exploiting immunoinformatics and dynamic simulation. PLoS One 2025; 20:e0313559. [PMID: 39761277 PMCID: PMC11703113 DOI: 10.1371/journal.pone.0313559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/25/2024] [Indexed: 05/01/2025] Open
Abstract
Human papillomavirus 16 and human papillomavirus 18 have been associated with different life-threatening cancers, including cervical, lung, penal, vulval, vaginal, anal, and oropharyngeal cancers, while cervical cancer is the most prominent one. Several research studies have suggested that the oncoproteins E6 and E7 are the leading cause of cancers associated with the human papillomavirus infection. Therefore, we developed two mRNA vaccines (V1 and V2) targeting these oncoproteins. We used several bioinformatics tools to predict helper T lymphocyte, cytotoxic T lymphocyte, and B-cell epitopes derived from the proteins and assessed their antigenicity, allergenicity, and toxicity. Both vaccines were constructed using selected epitopes, linkers, and adjuvants. After that, the vaccines were applied for physicochemical properties, secondary and tertiary structure predictions, and subsequent docking and simulation analyses. Accordingly, vaccine 1 (V1) and vaccine 2 (V2) showed better hydrophilicity with the grand average hydropathicity score of -0.811 and -0.648, respectively. The secondary and tertiary structures of the vaccines were also deemed satisfactory, with high stability indicated by the Ramachandran plot (V1:94.5% and V2:87.1%) and Z scores (V1: -5.15 and V2: -4.1). Docking analysis revealed substantial affinity of the vaccines towards the toll-like receptor-2 (V1: -1159.3, V2: -1246.3) and toll-like receptor-4 (V1: -1109.3, V2: -1244.8) receptors. Molecular dynamic simulation validated structural integrity and indicated varying stability throughout the simulation. Codon optimization showed significant expression of the vaccines (V1:51.88% and V2:51.63%) in E. coli vectors. Furthermore, regarding immune stimulation, the vaccines elicited significant B-cell and T-cell responses, including sustained adaptive and innate immune responses. Finally, thermodynamic predictions indicated stable mRNA structures of the vaccines (V1: -502.60 kcal/mol and V2: -450.90 kcal/mol). The proposed vaccines designed effectively targeting human papillomavirus oncoproteins have demonstrated promising results via robust immune responses, suggesting their suitability for further clinical advancement, including in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, Australia
| | - Md. Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chittagong, Bangladesh
| | - Rehana Parvin
- Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Khulshi, Chittagong, Bangladesh
| | - Shuvo Chandra Das
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Asma Talukder
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, Australia
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
10
|
Sattari Sarvari S, Rezaei Adriani R, Nazarian S, Fotouhi A, Mousavi Gargari SL. Protective Immunity of a Novel Multi-Epitope Vaccine Encoding OMP31, TF, BLS, SOD, BP26, and L9 Against Brucella spp. Infection. IRANIAN BIOMEDICAL JOURNAL 2025; 29:36-48. [PMID: 40223393 PMCID: PMC12040634 DOI: 10.61186/ibj.4933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/08/2024] [Indexed: 04/17/2025]
Abstract
Background Brucella is a type of bacteria that causes a disease known as brucellosis in both humans and animals. Many different vaccine formulations are available for this disease; however, vaccines based on epitopes have shown to be effective, especially in combating this pathogen. In the present study, we designed a multi-epitope vaccine against brucellosis using a chimeric protein that combines segments from various Brucella proteins known to contain both B- and T-cell epitopes. Methods In this study, a vaccine candidate was developed using multiple epitopes derived from various proteins, including OMP31, TF, BLS, SOD, BP26, and L9. These epitopes were selected based on their high density of both B-cell and T-cell epitopes. The construct of the vaccine candidate was inserted into a pEGFP-N1 vector and introduced into HEK-293T cells. Subsequently, the vaccine was tested on different groups of mice; some received the expressed protein in E. coli, while others received the DNA vaccine candidate. An ELISA assay was employed to evaluate the humoral immune response. Results Both the MEB protein (Pro/Pro) and pCI-MEB plasmid/MEB protein (DNA/Pro) groups showed a specific humoral response. The anti-DNA vaccine antibody titer did not rise as high as that of the protein groups; however, the observed protection indicated the efficiency of the DNA vaccine in activating the immune system. Conclusion While the chimeric DNA vaccine candidate induced a weaker humoral response, it remained effective in protecting against virulent strains of B. abortus and B. melitensis in the challenge route.
Collapse
Affiliation(s)
| | | | - Shahram Nazarian
- Department of Biology, Faculty of Science, Imam Hussein University, Tehran, Iran
| | | | | |
Collapse
|
11
|
Sarabi PA, Rismani E, Shabanpouremam M, Talehahmad S, Vosough M. Developing a multi-epitope vaccine against Helicobacter Pylori. Hum Immunol 2025; 86:111212. [PMID: 39642777 DOI: 10.1016/j.humimm.2024.111212] [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: 09/16/2024] [Revised: 11/17/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Helicobacter pylori, a significant factor in the development of gastric cancer and peptic ulcers, poses challenges for drug development due to its resilience. Computational approaches offer potential solutions for effective vaccine development targeting its antigens while ensuring stability and safety. The four critical antigenic proteins included in this study's innovative vaccine design are neuraminyllactose-binding hemagglutinin (HpaA), catalase (KatA), urease (UreB), and vacuolating toxin (VacA). Advanced immunoinformatics methods identified the possibility of triggering an immunological reaction. An adjuvant (50S ribosomal protein L7/L12) was fused to the vaccine sequence's N-terminus to improve immunogenicity. GROMACS molecular dynamics simulations with the OPLS-AA force field further improved the structure. The vaccine design and human Toll-like receptor 5 (TLR5) demonstrated a strong binding in docking tests. A model of simulating immune response confirmed the vaccine's efficacy and predicted how it would affect the immune system. Using the optimal restriction sites of the pET28b (+) expression vector, the vaccine candidate was cloned in silico. To validate the findings, this vaccine design will be synthesized in a bacterial system, and in experimental studies will be conducted in the following phase.
Collapse
Affiliation(s)
- Pedram Asadi Sarabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elham Rismani
- Molecular Medicine Department, Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mahshid Shabanpouremam
- Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran
| | - Sara Talehahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran..
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, and Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
12
|
Sami MRS, Rani NA, Elahi MME, Hossain MS, Al Mueid MA, Rahim Z, Patil RB, Moin AT, Bithi IJ, Nahar S, Konika IJ, Roy S, Preya JA, Ahmed J. An immunoinformatics and extensive molecular dynamics study to develop a polyvalent multi-epitope vaccine against cryptococcosis. PLoS One 2024; 19:e0315105. [PMID: 39739919 PMCID: PMC11687922 DOI: 10.1371/journal.pone.0315105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
Cryptococcosis is a lethal mycosis instigated by the pathogenic species Cryptococcus neoformans and Cryptococcus gattii, primarily affects the lungs, manifesting as pneumonia, and the brain, where it presents as meningitis. Mortality rate could reach 100% if infections remain untreated in cryptococcal meningitis. Treatment options for cryptococcosis are limited and and there are no licensed vaccines clinically available to treat or prevent cryptococcosis. Our study utilizes an integrated bioinformatics approaches to develop a polyvalent multiepitope subunit vaccine focusing on the key virulent proteins Heat shock transcription factor and Chaperone DnaK of both C. neoformans and C. gatti. Then in silico analysis was done to predict highly antigenic epitopes by assessing antigenicity, transmembrane topology screening, allergenecity, toxicity, and molecular docking approaches. Following this analysis, we designed two vaccine constructs integrating a compatible adjuvant and suitable linkers. These constructs exhibited notable characteristics including high antigenicity, non-toxicity, solubility, stability, and compatibility with Toll-like receptors (TLRs). The interaction between both vaccine constructs and TLR2, TLR3, and TLR9 was assessed through molecular docking analysis. Molecular dynamics simulations and MM-PBSA calculations suggest the substantial stabilizing property and binding affinity of Vaccine Construct V1 against TLR9. Both the vaccines revealed to have a higher number of interchain hydrogen bond with TLR9. These findings serve as a crucial stepping stone towards a comprehensive solution for combating cryptococcus infections induced by both C. neoformans and C. gattii. Further validation through in vivo studies is crucial to confirm the effectiveness and potential of the vaccine to curb the spread of cryptococcosis. Subsequent validation through in vivo studies is paramount to confirm the effectiveness and potential of the vaccine in reducing the spread of cryptococcosis.
Collapse
Affiliation(s)
- Md. Razwan Sardar Sami
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Nurul Amin Rani
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | | | - Mohammad Sajjad Hossain
- Department of Theoretical and Computational Chemistry, Faculty of Science, University of Dhaka, Dhaka, Bangladesh
| | - Minhaz Abdullah Al Mueid
- Department of Pharmacy, Faculty of Biological Science, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Zahidur Rahim
- Department of Zoology, Jahangirnagar University, Dhaka, Bangladesh
| | - Rajesh B. Patil
- Sinhgad Technical Education Societys, Sinhgad College of Pharmacy, Department of Pharmaceutical Chemistry, Pune, Maharashtra, India
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science, University of Chittagong, Chattogram, Bangladesh
| | - Israt Jahan Bithi
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Sabekun Nahar
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Israt Jahan Konika
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Sneha Roy
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Jannatul Aleya Preya
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Jamil Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Biochemistry and Chemistry, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| |
Collapse
|
13
|
Khan J, Sadiq A, Alrashed MM, Basharat N, Hassan Mohani SNU, Shah TA, Attia KA, Shah AA, Khan H, Ali I, Mohammed AA. Designing multi-epitope vaccines against Echinococcus granulosus: an in-silico study using immuno-informatics. BMC Mol Cell Biol 2024; 25:29. [PMID: 39736511 DOI: 10.1186/s12860-024-00524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/09/2024] [Indexed: 01/01/2025] Open
Abstract
Cystic echinococcosis (CE) is a worldwide zoonotic public health issue. The reasons for this include a lack of specific therapy options, increasing antiparasitic drug resistance, a lack of control strategies, and the absence of an approved vaccine. The aim of the current study is to develop a multiepitope vaccine against CE by in-silico identification and using different Antigen B subunits. The five Echinococcus granulosus antigen B (EgAgB) subunits were examined for eminent antigenic epitopes, and then the best B-cell and Major Histocompatibility Complex MHC-binding epitopes were predicted. Most significant epitopes were combined to create an effective multi-epitope vaccine, which was then validated by testing its secondary and tertiary structures, physicochemical properties, and molecular dynamics (MD) modelling. A multi-epitope vaccine construct of 483 amino acid sequences was designed. It contains B-cell, Helper T Lymphocyte (HTL), and Cytotoxic T Lymphocyte (CTL) epitopes as well as the appropriate adjuvant and linker molecules. The resultant vaccinal construct had a GDT-HA value of 0.9725, RMSD of 0.299, MolProbity of 1.891, Clash score of 13.1, Poor rotamers of 0.9, and qualifying features with Rama favoured of 89.9. It was also highly immunogenic and less allergic. The majority of the amino acids were positioned in the Ramachandran plot's favourable area, and during the molecular dynamic simulation at 100 ns, no notable structural abnormalities were noticed. The resultant construct was significantly expressed and received good endorsement in the pIB2-SEC13-mEGFP expressional vector. In conclusion, the current in-silico multi-epitope vaccine may be evaluated in-vitro, in-vivo, and in clinical trials as an immunogenic vaccine model. It can also play a vital role in preventing this zoonotic parasite infection.
Collapse
Affiliation(s)
- Jadoon Khan
- Faculty of Biological Sciences, Department of Microbiology, Quaid I Azam University Islamabad, Islamabad, Pakistan.
- Molecular Virology Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan.
- Department of Allied Health Sciences, Sarhad University of Science and Information Technology, Islamabad Campus, Islamabad, Pakistan.
| | - Asma Sadiq
- Department of Microbiology, University of Jhang, Jhang, Pakistan
| | - May M Alrashed
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Nosheen Basharat
- Molecular Virology Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Syed Nadeem Ul Hassan Mohani
- Department of Pharmacy, Sarhad University of Science and Information Technology, Islamabad Campus, Islamabad, Pakistan
| | - Tawaf Ali Shah
- College of Agriculture Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China.
| | - Kotb A Attia
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Aamer Ali Shah
- Faculty of Biological Sciences, Department of Microbiology, Quaid I Azam University Islamabad, Islamabad, Pakistan
| | - Hayat Khan
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Khyber Pukhtoonkhwa, Pakistan
| | - Ijaz Ali
- Molecular Virology Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan.
- Center for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally, Kuwait.
| | - Arif Ahmed Mohammed
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
14
|
Liu Y, Liu Z, Zheng Z. Rational Design of an Epidermal Growth Factor Receptor Vaccine: Immunogenicity and Antitumor Research. Biomolecules 2024; 14:1620. [PMID: 39766327 PMCID: PMC11726940 DOI: 10.3390/biom14121620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/15/2025] Open
Abstract
The epidermal growth factor receptor (EGFR) is frequently overexpressed in a variety of human epithelial tumors, and its aberrant activation plays a pivotal role in promoting tumor growth, invasion, and metastasis. The clinically approved passive EGFR-related therapies have numerous limitations. Seven EGFR-ECD epitope peptides (EG1-7) were selected through bioinformatics epitope prediction tools including NetMHCpan-4.1, NetMHCIIpan-3.2, and IEDB Consensus (v2.18 and v2.22) and fused to the translocation domain of diphtheria toxin (DTT). The A549 tumor model was successfully established in a murine mouse model. The vaccine was formulated by combining the adjuvants Alum and CpG and subsequently assessed for its immunogenicity and anti-tumor efficacy. DTT-EG (3;5;6;7) vaccines elicited specific humoral and cellular immune responses and effectively suppressed tumor growth in both prophylactic and therapeutic mouse tumor models. The selected epitopes EG3 (HGAVRFSNNPALCNV145-159), EG5 (KDSLSINATNIKHFK346-360), EG6 (VKEITGFLLIQAWPE398-412), and EG7 (LCYANTINWKKLFGT469-483) were incorporated into vaccines for active immunization, representing a promising strategy for the treatment of tumors with overexpressed epidermal growth factor receptor (EGFR). The vaccine design and fusion method employed in this study demonstrate a viable approach toward the development of cancer vaccines.
Collapse
Affiliation(s)
| | | | - Zhongliang Zheng
- College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (Z.L.)
| |
Collapse
|
15
|
Fadaie M, Shahmoradi Z, Khanahmad H. Immunoinformatic approach to the design of a novel multi-epitope vaccine against Leishmania major fused to human IgG-Fc. Res Pharm Sci 2024; 19:729-745. [PMID: 39911897 PMCID: PMC11792711 DOI: 10.4103/rps.rps_145_24] [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: 08/08/2024] [Revised: 10/14/2024] [Accepted: 11/18/2024] [Indexed: 02/07/2025] Open
Abstract
Background and purpose Cutaneous leishmaniasis poses significant health and socioeconomic challenges, making vaccine development a top priority, especially in endemic regions. Cysteine proteases, KMP-11, and HASPB proteins are promising candidates for leishmaniasis vaccine development owing to their immunogenic properties and capacity to provoke robust immune responses, as evidenced by different investigations. This study aimed to design a recombinant chimeric protein (MEV-Fc) vaccine using multi-epitopes from these Leishmania major proteins. Experimental approach The antigens were subjected to immunoinformatic prediction and screening of HTL, CTL, and B-cell epitopes. The multi-epitope protein was designed with significantly high-scoring epitopes and suitable linkers. Natural adjuvants were then added to enhance immunogenicity. Vaccine potency was innovatively improved by covalently fusing human IgG1 Fc with multi-epitope protein. To investigate how the MEV-Fc vaccine interacts with Toll-like receptors, molecular docking, multi-scale normal mode analysis simulation, and computational immune simulation were employed to study humoral and cellular immune responses. Findings/Results The results demonstrated the vaccine's antigenicity, stability, and nontoxicity. The structural validation confirmed the accuracy of the 3D models, indicating robust interactions with TLR2 and TLR4, with binding free energies of -1269.9 and -1128.7 (kcal/mol), respectively. Immune simulation results showed significant increases in IgM and IgG antibody levels following three vaccinations, along with enhanced activation of B cells, helper T cells, and cytotoxic T lymphocytes. Conclusion and implications These findings provide novel insights for developing effective candidates for cutaneous leishmaniasis vaccines. However, laboratory experiments are necessary to evaluate its protective effects.
Collapse
Affiliation(s)
- Mahmood Fadaie
- Skin Diseases and Leishmaniasis Research Center, Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zabihollah Shahmoradi
- Skin Diseases and Leishmaniasis Research Center, Department of Dermatology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Skin Diseases and Leishmaniasis Research Center, Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Barazesh M, Abbasi M, Mohammadi M, Nasiri MN, Rezaei F, Mohammadi S, Kavousipour S. Bioinformatics analysis to design a multi-epitope mRNA vaccine against S. agalactiae exploiting pathogenic proteins. Sci Rep 2024; 14:28294. [PMID: 39550419 PMCID: PMC11569170 DOI: 10.1038/s41598-024-79503-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: 05/03/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
Antibiotic resistance in bacterial pathogen infections is a growing global issue that occurs due to their adaptation to changing environmental conditions. Therefore, producing an efficient vaccine as an alternative approach can improve the immune system, eradicate related pathogens, and overcome this growing problem. Streptococcus agalactiae belongs to group B Streptococcus (GBS). Colonization of GBS during pregnancy is a significant risk factor for infants and young children. S. agalactiae infected population exhibits resistance to beta-lactams, including penicillin and the second-line antibiotics erythromycin and clindamycin. On the other hand, there are currently no commercial vaccines against this pathogen. Vaccination of pregnant women is a highly effective method to protect newborns and infants from S. agalactiae infection, and it has been identified as an urgent demand by the World Health Organization. This study employed various immunoinformatic tools to develop an effective vaccine that could trigger both humoral and cell-mediated immunity and prevent disease. For this purpose, three conserved antigenic proteins of the main pathogenic strains of S. agalactiae were utilized to predict CTL, HTL, and B-cell epitopes for producing an mRNA vaccine against different strains of S. agalactiae. The selected epitopes were fused using proper linkers. The Resuscitation promoting factor E (RpfE) sequence was incorporated in the designed vaccine construct as an adjuvant to boost its immune response. Different physicochemical characteristics of the final designed vaccine, modeling of the three-dimensional structure, molecular docking, molecular dynamics simulation, and immunological response simulation were screened following vaccine administration in an in vivo model. Computational immune simulation data identified that IgG1, IgM, INF γ, IL-2, T helper, and B-cell populations increased significantly after vaccination. These findings suggested that the vaccine candidate may provide good protection against S. agalactiae infection. However, experimental and animal model studies are required for additional validation and implementation in human vaccination programs.
Collapse
Affiliation(s)
- Mahdi Barazesh
- Department of Medical Biotechnology, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Maryam Abbasi
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohsen Mohammadi
- Hepatitis Research Center and Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Naser Nasiri
- Department of Clinical pharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Faranak Rezaei
- Razi Herbal Medicines Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shiva Mohammadi
- Hepatitis Research Center, Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Soudabeh Kavousipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, BandarAbbas, Iran
| |
Collapse
|
17
|
Chowdhury S, Sadhukhan P, Mahata N. Immunoinformatics investigation on pathogenic Escherichia coli proteome to develop an epitope-based peptide vaccine candidate. Mol Divers 2024:10.1007/s11030-024-11034-0. [PMID: 39516450 DOI: 10.1007/s11030-024-11034-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Escherichia coli (E. coli), a gram-negative bacterium, quickly colonizes in the human gastrointestinal tract after birth and typically sustains a long-term, symbiotic relationship with the host. However, certain virulent strains of E. coli can cause diseases such as urinary tract infections, meningitis, and enteric disorders. The rising antibiotic resistance among these strains has heightened the urgency for an effective vaccine. This study employs immunoinformatics and a reverse vaccinology technique to identify prospective antigens and create an efficient vaccine construct. In this study, we reported the "Attaching and Effacing Protein" a novel outer-membrane protein conserved in all pathogenic E. coli strains, based on proteome screening. We developed an in silico multi-epitope vaccine that includes helper T lymphocyte (HTL), cytotoxic T lymphocyte (CTL), B cell lymphocyte (BCL), and pan HLA DR-binding reactive epitope (PADRE) sequences, along with appropriate linkers and adjuvants. Machine Learning algorithms were used to evaluate antigenicity, solubility, stability, and non-allergenicity of the vaccine construct. Additionally, molecular docking analysis revealed that vaccine construct has a strong predicted binding affinity for human toll-like receptors on the cell surface. In this context, laboratory validations are necessary to demonstrate the effectiveness of the possible vaccine design that showed encouraging findings through computational validation.
Collapse
Affiliation(s)
- Soham Chowdhury
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India
| | - Pinkan Sadhukhan
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| |
Collapse
|
18
|
Masum MHU, Mahdeen AA, Barua L, Parvin R, Heema HP, Ferdous J. Developing a chimeric multiepitope vaccine against Nipah virus (NiV) through immunoinformatics, molecular docking and dynamic simulation approaches. Microb Pathog 2024; 197:107098. [PMID: 39521154 DOI: 10.1016/j.micpath.2024.107098] [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: 07/06/2024] [Revised: 10/09/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Nipah virus (NiV) is a highly lethal zoonotic pathogen that poses a significant threat to human and animal health. Unfortunately, no effective treatments have been developed for this deadly zoonotic disease. Therefore, we designed a chimeric multiepitope vaccine targeting the Nipah virus (NiV) glycoprotein and fusion protein through immunoinformatic approaches. Therefore, the vaccine was developed by combining promising and potential antigenic MHC-I, MHC-II, and B-cell epitopes obtained from the selected proteins. When combined, the MHC-I and MHC-II epitopes offered 100 % global population coverage. The physicochemical characterization also exhibited favorable properties, including solubility and potential functional stability of the vaccine within the body (GRAVY score of -0.308). Structural analyses unveiled a well-stabilized secondary and tertiary structure with a Ramachandran score of 84.4 % and a Z score of -5.02. Findings from docking experiments with TLR-2 (-1089.3 kJ/mol) and TLR-4 (-1016.7 kJ/mol) showed a strong affinity of the vaccine towards the receptor. Molecular dynamics simulations revealed unique conformational dynamics among the "vaccine-apo," "vaccine-TLR-2," and "vaccine-TLR-4″ complexes. Consequently, the complexes exhibited significant compactness, flexibility, and exposure to solvents. The results of the codon optimization were remarkable, as the vaccine showed a significant amount of expression in the E. coli vector (GC content of 45.36 % and a CAI score of 1.0). The results of immune simulations, however, showed evidence of both adaptive and innate immune responses induced by the vaccine. Therefore, we highly recommend further research on this chimeric multiepitope vaccine to establish its efficacy and safety against the Nipah virus (NiV).
Collapse
Affiliation(s)
- Md Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University (CVASU), Khulshi, 4225, Chattogram, Bangladesh.
| | - Ahmad Abdullah Mahdeen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Logon Barua
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Rehana Parvin
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Khulshi, 4225, Chattogram, Bangladesh
| | - Homaira Pervin Heema
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Khulshi, 4225, Chattogram, Bangladesh
| | - Jannatul Ferdous
- Department of Obstetrics and Gynecology, Chittagong Medical College Hospital, Chattogram, 4203, Bangladesh
| |
Collapse
|
19
|
Moin AT, Rani NA, Sharker YA, Ahammed T, Rahman US, Yasmin S, Ratul IH, Joyoti SA, Musa MS, Rahaman MU, Biswas D, Ali MH, Alam SMMU, Patil RB, Nabi RU, Uddin MH. Computational design and evaluation of a polyvalent vaccine for viral nervous necrosis (VNN) in fish to combat Betanodavirus infection. Sci Rep 2024; 14:27020. [PMID: 39505874 PMCID: PMC11542017 DOI: 10.1038/s41598-024-72116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/03/2024] [Indexed: 11/08/2024] Open
Abstract
Viral nervous necrosis (VNN) poses a significant threat to the aquaculture industry, causing substantial losses and economic burdens. The disease, attributed to nervous necrosis viruses within the Betanodavirus genus, is particularly pervasive in the Mediterranean region, affecting various fish species across all production stages with mortality rates reaching 100%. Developing effective preventive measures against VNN is imperative. In this study, we employed rigorous immunoinformatics techniques to design a novel multi-epitope vaccine targeting VNN. Five RNA-directed RNA polymerases, crucial to the lifecycle of Betanodavirus, were selected as vaccine targets. The antigenicity and favorable physicochemical properties of these proteins were confirmed, and epitope mapping identified cytotoxic T lymphocyte, helper T lymphocyte, and linear B lymphocyte epitopes essential for eliciting a robust immune response. The selected epitopes, characterized by high antigenicity, non-allergenicity, and non-toxicity, were further enhanced by adding PADRE sequences and hBD adjuvants to increase immunogenicity. Two vaccine constructs were developed by linking epitopes using appropriate linkers, demonstrating high antigenicity, solubility, and stability. Molecular dynamics simulations revealed stable interactions between the vaccine constructs and Toll-like receptors (TLRs), essential for pathogen recognition and immune response activation in fish. Notably, vaccine construct V2 exhibited superior stability and binding affinity with TLR8, suggesting its potential as a promising candidate for VNN prevention. Overall, our study presents a comprehensive approach to VNN vaccine design utilizing immunoinformatics, offering safe, immunogenic, and effective solutions across multiple Betanodavirus species. Further experimental validation in model animals is recommended to fully assess the vaccine's efficacy. This research contributes to improved vaccine development against diverse fish pathogens by addressing emerging challenges and individualized immunization requirements in aquaculture.
Collapse
Affiliation(s)
- Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh.
| | - Nurul Amin Rani
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | | | - Tanbir Ahammed
- Department of Biotechnology and Bioinformatics, School of Environment and Life Science, Independent University Bangladesh, Dhaka, 1229, Bangladesh
| | - Umme Sadea Rahman
- Department of Pharmacy, School of Pharmacy and Public Health, Independent University Bangladesh, Dhaka, 1229, Bangladesh
| | - Sadia Yasmin
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong, Chattogram, 4202, Bangladesh
| | | | - Shanjida Akter Joyoti
- Department of Fisheries, Faculty of Marine Sciences and Fisheries, University of Chittagong, Chattogram, 4331, Bangladesh
| | - Muhammad Sakib Musa
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, 4331, Bangladesh
| | - Mizan Ur Rahaman
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Dipta Biswas
- Department of Fisheries, Faculty of Marine Sciences and Fisheries, University of Chittagong, Chattogram, 4331, Bangladesh
| | - Md Hazrat Ali
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - S M Murshid Ul Alam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh.
| | - Rajesh B Patil
- Department of Pharmaceutical Chemistry, Sinhgad Technical Education Society's, Sinhgad College of Pharmacy, Pune, 411041, Maharashtra, India.
| | - Rashed Un Nabi
- Department of Fisheries, Faculty of Marine Sciences and Fisheries, University of Chittagong, Chattogram, 4331, Bangladesh.
| | - Mohammad Helal Uddin
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, 4331, Bangladesh.
| |
Collapse
|
20
|
Douradinha B. Computational strategies in Klebsiella pneumoniae vaccine design: navigating the landscape of in silico insights. Biotechnol Adv 2024; 76:108437. [PMID: 39216613 DOI: 10.1016/j.biotechadv.2024.108437] [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: 05/28/2024] [Revised: 07/07/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
The emergence of multidrug-resistant Klebsiella pneumoniae poses a grave threat to global public health, necessitating urgent strategies for vaccine development. In this context, computational tools have emerged as indispensable assets, offering unprecedented insights into klebsiellal biology and facilitating the design of effective vaccines. Here, a review of the application of computational methods in the development of K. pneumoniae vaccines is presented, elucidating the transformative impact of in silico approaches. Through a systematic exploration of bioinformatics, structural biology, and immunoinformatics techniques, the complex landscape of K. pneumoniae pathogenesis and antigenicity was unravelled. Key insights into virulence factors, antigen discovery, and immune response mechanisms are discussed, highlighting the pivotal role of computational tools in accelerating vaccine development efforts. Advancements in epitope prediction, antigen selection, and vaccine design optimisation are examined, highlighting the potential of in silico approaches to update vaccine development pipelines. Furthermore, challenges and future directions in leveraging computational tools to combat K. pneumoniae are discussed, emphasizing the importance of multidisciplinary collaboration and data integration. This review provides a comprehensive overview of the current state of computational contributions to K. pneumoniae vaccine development, offering insights into innovative strategies for addressing this urgent global health challenge.
Collapse
|
21
|
Jiang D, Ma Z, Zhang J, Sun Y, Bai T, Liu R, Wang Y, Guan L, Fu S, Sun Y, Li Y, Zhou B, Yang Y, Yang S, Chang Y, Sun B, Yang K. Immunoreactivity Analysis of MHC-I Epitopes Derived from the Nucleocapsid Protein of SARS-CoV-2 via Computation and Vaccination. Vaccines (Basel) 2024; 12:1214. [PMID: 39591116 PMCID: PMC11598499 DOI: 10.3390/vaccines12111214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Since 2019, the SARS-CoV-2 virus has been responsible for the global spread of respiratory illness. As of 1 September 2024, the cumulative number of infections worldwide exceeded 776 million. There are many structural proteins of the virus, among which the SARS-CoV-2 nucleocapsid (N) protein plays a pivotal role in the viral life cycle, participating in a multitude of essential activities following viral invasion. An important antiviral immune response is the major histocompatibility complex (MHC)-restricted differentiation cluster 8 (CD8+) T cell cytotoxicity. Therefore, understanding the immunogenicity of SARS-CoV-2 NP-specific MHC-I-restricted epitopes is highly important. Methods: MHC-I molecules from 11 human leukocyte antigen I (HLA-I) superfamilies with 98% population coverage and 6 mouse H2 alleles were selected. The affinity were screened by IEDB, NetMHCpan, SYFPEITHI, SMMPMBEC and Rankpep. Further immunogenicity and conservative analyses were performed using VaxiJen and BLASTp, respectively. EpiDock was used to simulate molecular docking. Cluster analysis was performed. Selective epitopes were validated by enzyme-linked immunospot (ELISpot) assay and flow cytometry in the mice with pVAX-NPSARS-CoV-2 immunization. Enzyme-Linked Immunosorbent Assay (ELISA) was used to detect whether the preferred epitope induced humoral immunity. Results: There were 64 dominant epitopes for the H-2 haplotype and 238 dominant epitopes for the HLA-I haplotype. Further analysis of immunogenicity and conservation yielded 8 preferred epitopes, and docking simulations were conducted with corresponding MHC-I alleles. The relationships between the NP peptides and MHC-I haplotypes were then determined via two-way hierarchical clustering. ELISA, ELISpot assay, and flow cytometry revealed that the preferred epitope stimulated both humoral and cellular immunity and enhanced cytokine secretion in mice. Conclusions: our study revealed the general patterns among multiple haplotypes within the humans and mice superfamily, providing a comprehensive assessment of the pan-MHC-I immunoreactivity of SARS-CoV-2 NP. Our findings would render prospects for the development and application of epitope-based immunotherapy in lasting viral epidemics.
Collapse
Affiliation(s)
- Dongbo Jiang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Zilu Ma
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Junqi Zhang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yubo Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Tianyuan Bai
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Ruibo Liu
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yongkai Wang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Liang Guan
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Shuaishuai Fu
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yuanjie Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yuanzhe Li
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Bingquan Zhou
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yulin Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Shuya Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yuanhang Chang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Baozeng Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
- Yingtan Detachment, Jiangxi General Hospital, Chinese People’s Armed Police Force, Nanchang 330001, China
- General Practice Medicine Base of Shanghai Changzheng Hospital, Shanghai 200041, China
| | - Kun Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| |
Collapse
|
22
|
Vietzen H, Simonitsch C, Friedel B, Berger SM, Kühner LM, Furlano PL, Florian DM, Görzer I, Koblischke M, Aberle JH, Puchhammer-Stöckl E. Torque teno viruses exhaust and imprint the human immune system via the HLA-E/NKG2A axis. Front Immunol 2024; 15:1447980. [PMID: 39295866 PMCID: PMC11408220 DOI: 10.3389/fimmu.2024.1447980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/09/2024] [Indexed: 09/21/2024] Open
Abstract
The ubiquitous Torque teno virus (TTV) establishes a chronically persistent infection in the human host. TTV has not been associated with any apparent disease, but, as part of the human virome, it may confer a regulatory imprint on the human immune system with as yet unclear consequences. However, so far, only few studies have characterized the TTV-specific immune responses or the overall immunological imprints by TTV. Here, we reveal that TTV infection leads to a highly exhausted TTV-specific CD8+ T-cell response, hallmarked by decreased IFN-γ production and the expression of the inhibitory NKG2A-receptor. On a functional level, we identified a panel of highly polymorphic TTV-encoded peptides that lead to an expansion of regulatory NKG2A+ natural killer, NKG2A+CD4+, and NKG2A+CD8+ T cells via the stabilization of the non-classical HLA-E molecule. Our results thus demonstrate that TTV leads to a distinct imprint on the human immune system that may further regulate overall human immune responses in infectious, autoimmune, and malignant diseases.
Collapse
Affiliation(s)
- Hannes Vietzen
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Cara Simonitsch
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - Sarah M Berger
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Laura M Kühner
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - David M Florian
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Irene Görzer
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - Judith H Aberle
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | |
Collapse
|
23
|
Ritaparna P, Ray M, Dhal AK, Mahapatra RK. An immunoinformatics approach for design and validation of multi-subunit vaccine against Plasmodium falciparum from essential hypothetical proteins. J Parasit Dis 2024; 48:593-609. [PMID: 39145352 PMCID: PMC11319695 DOI: 10.1007/s12639-024-01696-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/11/2024] [Indexed: 08/16/2024] Open
Abstract
Malaria, caused by Plasmodium falciparum, remains a pressing global health concern. Advancements in combating this parasite involve the development of a protein vaccine. This study employs immunoinformatics to identify potential vaccine candidates within the repertoire of 218 P. falciparum exported essential proteins identified through saturaturation mutagenesis study. Our screening approach narrows down to 65 Plasmodium-exported proteins with uncharacterized functions while exhibiting non-mutability in CDS (coding sequences). The transmembrane helix, antigenicity, allergenicity of the shortlisted proteins was assessed through diverse prediction algorithm, culminating in the identification of five promising vaccination contenders, based on probability scores. We discerned B-cell, helper T-lymphocyte, and cytotoxic T-lymphocyte epitopes. Two proteins with the most favorable epitope were harnessed to construct a multi-subunit vaccine, through judicious linker integration. Employing the I-TASSER software, three-dimensional models of the constituent proteins was obtained and was validated using diverse tools like ProSA, VERIFY3D, and ERRAT. The modelled proteins underwent Molecular Dynamics (MD) simulation in a solvent environment to evaluate the stability of the multi-subunit vaccine. Furthermore, we conducted molecular docking through the ClusPro web server to elucidate potential interactions with Toll-like receptors (TLR2 and TLR4). Docking scores revealed a pronounced affinity of the multi-subunit vaccine for TLR2. Significantly, 100 ns MD simulation of the protein-receptor complex unveiled a persistent hydrogen bond linkage between the ARG63 residue of the sub-unit vaccine and the GLU32 residue of the TLR2 receptor. These findings collectively advocate the potential efficacy of the first multi-subunit vaccine from the potential hypothetical proteins of P. falciparum. Supplementary Information The online version contains supplementary material available at 10.1007/s12639-024-01696-w.
Collapse
Affiliation(s)
- Prajna Ritaparna
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
- National Innovation Foundation, India, KIIT-TBI, Bhubaneswar, Odisha 751024 India
| | - Muskan Ray
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Ajit Kumar Dhal
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | | |
Collapse
|
24
|
Kaur A, Kumar A, Kumari G, Muduli R, Das M, Kundu R, Mukherjee S, Majumdar T. Rational design and computational evaluation of a multi-epitope vaccine for monkeypox virus: Insights into binding stability and immunological memory. Heliyon 2024; 10:e36154. [PMID: 39247273 PMCID: PMC11380015 DOI: 10.1016/j.heliyon.2024.e36154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Multi-epitope vaccines strategically tackle rapidly mutating viruses by targeting diverse epitopes from different proteins, providing a comprehensive and adaptable immune protection approach for enhanced coverage against various viral variants. This research employs a comprehensive approach that includes the mapping of immune cells activating epitopes derived from the six structural glycoproteins (A29L, A30L, A35R, L1R, M1R, and E8L) of Monkeypox virus (Mpox). A total of 7 T-cells-specific epitopes, 13 B-cells-specific epitopes, and 5 IFN-γ activating epitopes were forecasted within these glycoproteins. The selection process focused on epitopes indicating high immunogenicity and favorable binding affinity with multiple MHC alleles. Following this, a vaccine has been formulated by incorporating the chosen epitopes, alongside adjuvants (PADRE peptide) and various linkers (EAAAK, GPGPG, and AAY). The physicochemical properties and 3D structure of the multi-epitope hybrid vaccine were analysed for characterization. MD simulations were employed to predict the binding stability between the vaccine and various pathogen recognition receptors such as TLRs (TLR1, TLR2, TLR4, and TLR6), as well as both class I and II MHC, achieved through hydrogen bonding and hydrophobic interactions. Through in silico cloning and immune simulation, it was observed that the multi-epitopes vaccine induced a robust memory immune response upon booster doses, forecasting protective immunity upon viral challenge. This protective immunity was characterized by the production of IgM + IgG antibodies, along with release of inflammatory cytokines like IFN-γ, and IL12, and the activation of various immune cells. This study offers valuable insights into the potential of a multi-epitope vaccine targeting the Mpox virus.
Collapse
Affiliation(s)
| | - Amit Kumar
- National Institute of Immunology, New Delhi, India
| | | | | | - Mayami Das
- National Institute of Immunology, New Delhi, India
| | - Rakesh Kundu
- Department of Zoology, Visva-Bharati University, Santiniketan, West Bengal, India
| | - Suprabhat Mukherjee
- Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | | |
Collapse
|
25
|
Cui M, Ji X, Guan F, Su G, Du L. Design of a Helicobacter pylori multi-epitope vaccine based on immunoinformatics. Front Immunol 2024; 15:1432968. [PMID: 39247202 PMCID: PMC11377293 DOI: 10.3389/fimmu.2024.1432968] [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: 05/15/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Helicobacter pylori (H. pylori) is an infectious bacterium that colonizes the stomach of approximately half of the global population. It has been classified as a Group I carcinogen by the World Health Organization due to its strong association with an increased incidence of gastric cancer and exacerbation of stomach diseases. The primary treatment for H. pylori infection currently involves triple or quadruple therapy, primarily consisting of antibiotics and proton pump inhibitors. However, the increasing prevalence of antibiotic resistance poses significant challenges to this approach, underscoring the urgent need for an effective vaccine. In this study, a novel multi-epitope H. pylori vaccine was designed using immunoinformatics. The vaccine contains epitopes derived from nine essential proteins. Software tools and online servers were utilized to predict, evaluate, and analyze the physiochemical properties, secondary and tertiary structures, and immunogenicity of the candidate vaccine. These comprehensive assessments ultimately led to the formulation of an optimal design scheme for the vaccine. Through constructing a novel multi-epitope vaccine based on immunoinformatics, this study offers promising prospects and great potential for the prevention of H. pylori infection. This study also provides a reference strategy to develop multi-epitope vaccines for other pathogens.
Collapse
Affiliation(s)
- Man Cui
- Research and Development Centre, Beijing Zhifei Lvzhu Biopharmaceutical Co., Ltd., Beijing, China
- Beijing Bacterial Vaccine Engineering Research Centre, Beijing, China
| | - Xiaohui Ji
- Research and Development Centre, Beijing Zhifei Lvzhu Biopharmaceutical Co., Ltd., Beijing, China
- Beijing Bacterial Vaccine Engineering Research Centre, Beijing, China
| | - Fengtao Guan
- Research and Development Centre, Beijing Zhifei Lvzhu Biopharmaceutical Co., Ltd., Beijing, China
- Beijing Bacterial Vaccine Engineering Research Centre, Beijing, China
| | - Guimin Su
- Research and Development Centre, Beijing Zhifei Lvzhu Biopharmaceutical Co., Ltd., Beijing, China
- Beijing Bacterial Vaccine Engineering Research Centre, Beijing, China
| | - Lin Du
- Research and Development Centre, Beijing Zhifei Lvzhu Biopharmaceutical Co., Ltd., Beijing, China
- Beijing Bacterial Vaccine Engineering Research Centre, Beijing, China
| |
Collapse
|
26
|
Asad M, Hassan A, Wang W, Alonazi WB, Khan MS, Ogunyemi SO, Ibrahim M, Bin L. An integrated in silico approach for the identification of novel potential drug target and chimeric vaccine against Neisseria meningitides strain 331401 serogroup X by subtractive genomics and reverse vaccinology. Comput Biol Med 2024; 178:108738. [PMID: 38870724 DOI: 10.1016/j.compbiomed.2024.108738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/15/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024]
Abstract
Neisseria meningitidis, commonly known as the meningococcus, leads to substantial illness and death among children and young adults globally, revealing as either epidemic or sporadic meningitis and/or septicemia. In this study, we have designed a novel peptide-based chimeric vaccine candidate against the N. meningitidis strain 331,401 serogroup X. Through rigorous analysis of subtractive genomics, two essential cytoplasmic proteins, namely UPI000012E8E0(UDP-3-O-acyl-GlcNAc deacetylase) and UPI0000ECF4A9(UDP-N-acetylglucosamine acyltransferase) emerged as potential drug targets. Additionally, using reverse vaccinology, the outer membrane protein UPI0001F4D537 (Membrane fusion protein MtrC) identified by subcellular localization and recognized for its known indispensable role in bacterial survival was identified as a novel chimeric vaccine target. Following a careful comparison of MHC-I, MHC-II, T-cell, and B-cell epitopes, three epitopes derived from UPI0001F4D537 were linked with three types of linkers-GGGS, EAAAK, and the essential PADRE-for vaccine construction. This resulted in eight distinct vaccine models (V1-V8). Among them V1 model was selected as the final vaccine construct. It exhibits exceptional immunogenicity, safety, and enhanced antigenicity, with 97.7 % of its residues in the Ramachandran plot's most favored region. Subsequently, the vaccine structure was docked with the TLR4/MD2 complex and six different HLA allele receptors using the HADDOCK server. The docking resulted in the lowest HADDOCK score of 39.3 ± 9.0 for TLR/MD2. Immune stimulation showed a strong immune response, including antibodies creation and the activation of B-cells, T Cytotoxic cells, T Helper cells, Natural Killer cells, and interleukins. Furthermore, the vaccine construct was successfully expressed in the Escherichia coli system by reverse transcription, optimization, and ligation in the pET-28a (+) vector for the expression study. The current study proposes V1 construct has the potential to elicit both cellular and humoral responses, crucial for the developing an epitope-based vaccine against N. meningitidis strain 331,401 serogroup X.
Collapse
Affiliation(s)
- Muhammad Asad
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Pakistan
| | - Ahmad Hassan
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Pakistan
| | - Weiyu Wang
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Wadi B Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | | | - Solabomi Olaitan Ogunyemi
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Muhammad Ibrahim
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Pakistan.
| | - Li Bin
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
27
|
Li X, Zhou Q, Lu Z, Huang R, Lin D, Xu J, Yu X, Li X. Association of HLA-DRB1 alleles with status of antibodies to hepatitis B surface and e antigen. J Med Virol 2024; 96:e29867. [PMID: 39169719 DOI: 10.1002/jmv.29867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/11/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Antigen presentation by HLA class II molecules to CD4+ T cells is an essential step for generating antibodies to hepatitis B antigens. In this study, we investigated the association between the HLA-DRB1 gene and the status of antibodies to hepatitis B surface and e antigens. Our results revealed a significant association between the status of anti-HBsAg and HLA-DRB1*04:03 (OR = 4.11, 95% CI = 1.50-10.84, p = 0.005, Padj. = 0.05) as well as HLA-DRB1*15:01 (OR = 1.74, 95% CI = 1.20-2.50, p = 0.002, Padj. = 0.045). MHC II binding predictions and in silico docking demonstrated strong binding affinity of HBsAg peptides to these two HLA-DRB1 molecules. Conversely, the status of anti-HBeAg was inversely associated with HLA-DRB1*14:54 (OR = 0.34, 95% CI = 0.18-0.64, p = 0.001, Padj. = 0.011), and in silico analysis revealed weak binding affinity of HBeAg peptides to HLA-DRB1*14:54. In conclusion, these findings support the involvement of HLA-DRB1 in humoral immunity against HBV infection.
Collapse
Affiliation(s)
- Xinze Li
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
- NHC Key Laboratory of Tropical Disease Control,School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiaomiao Zhou
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Zhe Lu
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Renliang Huang
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Dan Lin
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Jing Xu
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Xinhua Yu
- Hainan Women and Children's Medical Center, Haikou, Hainan, China
- Priority Area Chronic Lung Diseases, Research Center Borstel, Borstel, Germany
| | - Xuexia Li
- NHC Key Laboratory of Tropical Disease Control,School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
28
|
Roy SK, Biswas MS, Foyzur Raman M, Hasan R, Rahmann Z, Uddin PK MM. A computational approach to developing a multi-epitope vaccine for combating Pseudomonas aeruginosa-induced pneumonia and sepsis. Brief Bioinform 2024; 25:bbae401. [PMID: 39133098 PMCID: PMC11318047 DOI: 10.1093/bib/bbae401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/16/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024] Open
Abstract
Pseudomonas aeruginosa is a complex nosocomial infectious agent responsible for numerous illnesses, with its growing resistance variations complicating treatment development. Studies have emphasized the importance of virulence factors OprE and OprF in pathogenesis, highlighting their potential as vaccine candidates. In this study, B-cell, MHC-I, and MHC-II epitopes were identified, and molecular linkers were active to join these epitopes with an appropriate adjuvant to construct a vaccine. Computational tools were employed to forecast the tertiary framework, characteristics, and also to confirm the vaccine's composition. The potency was weighed through population coverage analysis and immune simulation. This project aims to create a multi-epitope vaccine to reduce P. aeruginosa-related illness and mortality using immunoinformatics resources. The ultimate complex has been determined to be stable, soluble, antigenic, and non-allergenic upon inspection of its physicochemical and immunological properties. Additionally, the protein exhibited acidic and hydrophilic characteristics. The Ramachandran plot, ProSA-web, ERRAT, and Verify3D were employed to ensure the final model's authenticity once the protein's three-dimensional structure had been established and refined. The vaccine model showed a significant binding score and stability when interacting with MHC receptors. Population coverage analysis indicated a global coverage rate of 83.40%, with the USA having the highest coverage rate, exceeding 90%. Moreover, the vaccine sequence underwent codon optimization before being cloned into the Escherichia coli plasmid vector pET-28a (+) at the EcoRI and EcoRV restriction sites. Our research has developed a vaccine against P. aeruginosa that has strong binding affinity and worldwide coverage, offering an acceptable way to mitigate nosocomial infections.
Collapse
Affiliation(s)
- Suronjit Kumar Roy
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Mohammad Shahangir Biswas
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
- Department of Public Health, Daffodil International University, Dhaka 1216, Bangladesh
| | - Md Foyzur Raman
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Rubait Hasan
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Zahidur Rahmann
- Institute of Biological Science, Rajshahi University, Motihar, Rajshahi 6205, Bangladesh
| | - Md Moyen Uddin PK
- Riceland Healthcare, 538 Broadway Ave, Winnie, TX 77665, United States
| |
Collapse
|
29
|
Masum MHU, Wajed S, Hossain MI, Moumi NR, Talukder A, Rahman MM. An mRNA vaccine for pancreatic cancer designed by applying in silico immunoinformatics and reverse vaccinology approaches. PLoS One 2024; 19:e0305413. [PMID: 38976715 PMCID: PMC11230540 DOI: 10.1371/journal.pone.0305413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma is the most prevalent pancreatic cancer, which is considered a significant global health concern. Chemotherapy and surgery are the mainstays of current pancreatic cancer treatments; however, a few cases are suitable for surgery, and most of the cases will experience recurrent episodes. Compared to DNA or peptide vaccines, mRNA vaccines for pancreatic cancer have more promise because of their delivery, enhanced immune responses, and lower proneness to mutation. We constructed an mRNA vaccine by analyzing S100 family proteins, which are all major activators of receptors for advanced glycation end products. We applied immunoinformatic approaches, including physicochemical properties analysis, structural prediction and validation, molecular docking study, in silico cloning, and immune simulations. The designed mRNA vaccine was estimated to have a molecular weight of 165023.50 Da and was highly soluble (grand average of hydropathicity of -0.440). In the structural assessment, the vaccine seemed to be a well-stable and functioning protein (Z score of -8.94). Also, the docking analysis suggested that the vaccine had a high affinity for TLR-2 and TLR-4 receptors. Additionally, the molecular mechanics with generalized Born and surface area solvation analysis of the "Vaccine-TLR-2" (-141.07 kcal/mol) and "Vaccine-TLR-4" (-271.72 kcal/mol) complexes also suggests a strong binding affinity for the receptors. Codon optimization also provided a high expression level with a GC content of 47.04% and a codon adaptation index score 1.0. The appearance of memory B-cells and T-cells was also observed over a while, with an increased level of helper T-cells and immunoglobulins (IgM and IgG). Moreover, the minimum free energy of the mRNA vaccine was predicted at -1760.00 kcal/mol, indicating the stability of the vaccine following its entry, transcription, and expression. This hypothetical vaccine offers a groundbreaking tool for future research and therapeutic development of pancreatic cancer.
Collapse
Affiliation(s)
- Md Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Shah Wajed
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Infectiology: Biology of Infectious Diseases, Universite Paris-Saclay, Gif-sur-Yvette, France
| | - Md Imam Hossain
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nusrat Rahman Moumi
- Medical Sciences, University of Central Lancashire, Preston, Lancashire, United Kingdom
| | - Asma Talukder
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| | - Md Mijanur Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
30
|
Rasool D, Jan SA, Khan SU, Nahid N, Ashfaq UA, Umar A, Qasim M, Noor F, Rehman A, Shahzadi K, Alshammari A, Alharbi M, Nisar MA. Subtractive proteomics-based vaccine targets annotation and reverse vaccinology approaches to identify multiepitope vaccine against Plesiomonas shigelloides. Heliyon 2024; 10:e31304. [PMID: 38845922 PMCID: PMC11153098 DOI: 10.1016/j.heliyon.2024.e31304] [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: 03/30/2023] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024] Open
Abstract
Plesiomonas shigelloides, an aquatic bacterium belonging to the Enterobacteriaceae family, is a frequent cause of gastroenteritis with diarrhea and gastrointestinal severe disease. Despite decades of research, discovering a licensed and globally accessible vaccine is still years away. Developing a putative vaccine that can combat the Plesiomonas shigelloides infection by boosting population immunity against P. shigelloides is direly needed. In the framework of the current study, the entire proteome of P. shigelloides was explored using subtractive genomics integrated with the immunoinformatics approach for designing an effective vaccine construct against P. shigelloides. The overall stability of the vaccine construct was evaluated using molecular docking, which demonstrated that MEV showed higher binding affinities with toll-like receptors (TLR4: 51.5 ± 10.3, TLR2: 60.5 ± 9.2) and MHC receptors(MHCI: 79.7 ± 11.2 kcal/mol, MHCII: 70.4 ± 23.7). Further, the therapeutic efficacy of the vaccine construct for generating an efficient immune response was evaluated by computational immunological simulation. Finally, computer-based cloning and improvement in codon composition without altering amino acid sequence led to the development of a proposed vaccine. In a nutshell, the findings of this study add to the existing knowledge about the pathogenesis of this infection. The schemed MEV can be a possible prophylactic agent for individuals infected with P. shigelloides. Nevertheless, further authentication is required to guarantee its safeness and immunogenic potential.
Collapse
Affiliation(s)
- Danish Rasool
- Department of Bioinformatics and Biosciences, Capital University of Science and Technology, Islamabad, 44000, Pakistan
| | - Sohail Ahmad Jan
- Department of Bioinformatics and Biosciences, Capital University of Science and Technology, Islamabad, 44000, Pakistan
| | | | - Nazia Nahid
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Ahitsham Umar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Fatima Noor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A & F Uiversity, yangling, 712100, Shaanxi, China
| | - Kiran Shahzadi
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Muhammad Atif Nisar
- College of Science and Engineering, Flinders University, Adelaide, 5042, Australia
| |
Collapse
|
31
|
Benyamini P. Phylogenetic Tracing of Evolutionarily Conserved Zonula Occludens Toxin Reveals a "High Value" Vaccine Candidate Specific for Treating Multi-Strain Pseudomonas aeruginosa Infections. Toxins (Basel) 2024; 16:271. [PMID: 38922165 PMCID: PMC11209546 DOI: 10.3390/toxins16060271] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Extensively drug-resistant Pseudomonas aeruginosa infections are emerging as a significant threat associated with adverse patient outcomes. Due to this organism's inherent properties of developing antibiotic resistance, we sought to investigate alternative strategies such as identifying "high value" antigens for immunotherapy-based purposes. Through extensive database mining, we discovered that numerous Gram-negative bacterial (GNB) genomes, many of which are known multidrug-resistant (MDR) pathogens, including P. aeruginosa, horizontally acquired the evolutionarily conserved gene encoding Zonula occludens toxin (Zot) with a substantial degree of homology. The toxin's genomic footprint among so many different GNB stresses its evolutionary importance. By employing in silico techniques such as proteomic-based phylogenetic tracing, in conjunction with comparative structural modeling, we discovered a highly conserved intermembrane associated stretch of 70 amino acids shared among all the GNB strains analyzed. The characterization of our newly identified antigen reveals it to be a "high value" vaccine candidate specific for P. aeruginosa. This newly identified antigen harbors multiple non-overlapping B- and T-cell epitopes exhibiting very high binding affinities and can adopt identical tertiary structures among the least genetically homologous P. aeruginosa strains. Taken together, using proteomic-driven reverse vaccinology techniques, we identified multiple "high value" vaccine candidates capable of eliciting a polarized immune response against all the P. aeruginosa genetic variants tested.
Collapse
Affiliation(s)
- Payam Benyamini
- Department of Health Sciences at Extension, University of California Los Angeles, 1145 Gayley Ave., Los Angeles, CA 90024, USA
| |
Collapse
|
32
|
Kashif M, Waseem M, Subbarao N. In silico prediction of CD8 + and CD4 + T cell epitopes in Leishmania major proteome: Using immunoinformatics. J Mol Graph Model 2024; 129:108759. [PMID: 38492406 DOI: 10.1016/j.jmgm.2024.108759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/12/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
The leishmaniases are NDTs (neglected tropical diseases) that affect people all over the world. They are brought on by protozoans from the genus Leishmania and disseminated by phlebotomine flies that are afflicted with the disease. The best option to manage and lower the incidence of these diseases has been thought by the creation of a safe and effective vaccination. This research used an in silico based mining approach to look for high potential epitopes that might bind to MHC Class I and MHC Class II molecules (mainly; HLA-A*02:01 & HLA-DRB1*03:01) from human population in order to promote vaccine development. Based on the presence of signal peptides, GPI anchors, antigenicity predictions, and a subtractive proteomic technique, we have screened 17 putative antigenic proteins from the 8083 total proteins of L. major. After that thorough immunogenic epitope prediction were done using IEDB-AR tools. We isolated five immunogenic epitopes (three 9-mer & two 15-mer) from five antigenic proteins through docking and MD simulation analysis. Finally, these five anticipated epitopes, viz., TLPEIPVNV, ELMAPVFGL, TLAAAVALL, NSINIRLDGVTSAGF and NVPLVVDASSLFRVA have considerably stronger binding potential with their respective alleles and may trigger immunological responses. The goal of this work was to identify MHC restricted epitopes for CD8+ and CD4+ T cells activation using immunoinformatics in order to identify potential vaccine candidates against L. major parasites.
Collapse
Affiliation(s)
- Mohammad Kashif
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Mohd Waseem
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Naidu Subbarao
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
33
|
Mubarak AS, Ameen ZS, Hassan AS, Ozsahin DU. Enhancing tuberculosis vaccine development: a deconvolution neural network approach for multi-epitope prediction. Sci Rep 2024; 14:10375. [PMID: 38710737 DOI: 10.1038/s41598-024-59291-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024] Open
Abstract
Tuberculosis (TB) a disease caused by Mycobacterium tuberculosis (Mtb) poses a significant threat to human life, and current BCG vaccinations only provide sporadic protection, therefore there is a need for developing efficient vaccines. Numerous immunoinformatic methods have been utilized previously, here for the first time a deep learning framework based on Deconvolutional Neural Networks (DCNN) and Bidirectional Long Short-Term Memory (DCNN-BiLSTM) was used to predict Mtb Multiepitope vaccine (MtbMEV) subunits against six Mtb H37Rv proteins. The trained model was used to design MEV within a few minutes against TB better than other machine learning models with 99.5% accuracy. The MEV has good antigenicity, and physiochemical properties, and is thermostable, soluble, and hydrophilic. The vaccine's BLAST search ruled out the possibility of autoimmune reactions. The secondary structure analysis revealed 87% coil, 10% beta, and 2% alpha helix, while the tertiary structure was highly upgraded after refinement. Molecular docking with TLR3 and TLR4 receptors showed good binding, indicating high immune reactions. Immune response simulation confirmed the generation of innate and adaptive responses. In-silico cloning revealed the vaccine is highly expressed in E. coli. The results can be further experimentally verified using various analyses to establish a candidate vaccine for future clinical trials.
Collapse
Affiliation(s)
- Auwalu Saleh Mubarak
- Operational Research Centre in Healthcare, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey
- Department of Electrical Engineering, Aliko Dangote University of Science and Technology, Wudil, Kano, Nigeria
| | - Zubaida Said Ameen
- Operational Research Centre in Healthcare, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey
- Department of Biochemistry, Yusuf Maitama Sule University, Kano, Nigeria
| | - Abdurrahman Shuaibu Hassan
- Department of Electrical Electronics and Automation Systems Engineering, Kampala International University, Kampala, Uganda.
| | - Dilber Uzun Ozsahin
- Operational Research Centre in Healthcare, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey.
- Department of Medical Diagnostic Imaging, College of Health Science, University of Sharjah, Sharjah, UAE.
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE.
| |
Collapse
|
34
|
Mortazavi B, Molaei A, Fard NA. Multi-epitopevaccines, from design to expression; an in silico approach. Hum Immunol 2024; 85:110804. [PMID: 38658216 DOI: 10.1016/j.humimm.2024.110804] [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: 03/01/2024] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
The development of vaccines against a wide range of infectious diseases and pathogens often relies on multi-epitope strategies that can effectively stimulate both humoral and cellular immunity. Immunoinformatics tools play a pivotal role in designing such vaccines, enhancing immune response potential, and minimizing the risk of failure. This review presents a comprehensive overview of practical tools for epitope prediction and the associated immune responses. These immunoinformatics tools facilitate the selection of epitopes based on parameters such as antigenicity, absence of toxic and allergenic sequences, secondary and tertiary structures, sequence conservation, and population coverage. The chosen epitopes can be tailored for B-cells or T-cells, both of which require further assessments covered in this study. We offer a range of suitable linkers that effectively separate cytotoxic T lymphocyte and helper T lymphocyte epitopes while preserving their functionality. Additionally, we identify various adjuvants for specific purposes. We delve into the evaluation of MHC-epitope interactions, MHC clusters, and the simulation of final constructs through molecular docking techniques. We provide diverse linkers and adjuvants optimized for epitope functions to bolster immune responses through epitope attachment. By leveraging these comprehensive tools, the development of multi-epitope vaccines holds the promise of robust immunity and a significant reduction in experimental costs.
Collapse
Affiliation(s)
- Behnam Mortazavi
- Department of systems Biotechnology, Faculty of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ali Molaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Najaf Allahyari Fard
- Department of systems Biotechnology, Faculty of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| |
Collapse
|
35
|
Shu Y, Huang R, Li Q, Lu Y, Yin J, Li H, Lan Z, Zheng X, Ye J, Long Y, Wang Z, Xiao L, Zhou Q, Liu X, Fu Y, Chen H, Chen J, Zhou Y, Zhou J, Zhang L, Zhou J, Jiang Y, Peng F, Lu Z, Petersen F, Qiu W, Yu X. Autoimmune Glial Fibrillary Acidic Protein Astrocytopathy Is Associated with HLA-A*3303 and HLA-DPB1*0501. Ann Neurol 2024; 95:901-906. [PMID: 38400794 DOI: 10.1002/ana.26899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
We determined the genetic association between specific human leucocyte antigen (HLA) loci and autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy. Our results showed that autoimmune GFAP astrocytopathy was associated with HLA-A*3303 (odds ratio [OR] = 2.02, 95% confidence interval [CI] = 1.32-3.06, p = 0.00072, padj. = 0.046) and HLA-DBP1*0501 (OR = 0.51, 95% CI = 0.36-0.71, p = 0.000048, padj. = 0.0062). Moreover, HLA-A*3303 carriers with the disease had a longer hospital stay (p = 0.0005) than non-carriers. This study for the first time provides evidence for a role of genetic factor in the development of autoimmune GFAP astrocytopathy. ANN NEUROL 2024;95:901-906.
Collapse
Affiliation(s)
- Yaqing Shu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Renliang Huang
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Qihui Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yi Lu
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Junping Yin
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Huilu Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhike Lan
- Department of Neurology, GuangDong 999 Brain Hospital, Guangzhou, China
| | - Xiujun Zheng
- Department of Neurology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou, China
| | - Jinlong Ye
- Department of Neurology, GuangDong 999 Brain Hospital, Guangzhou, China
| | - Youming Long
- Department of Neurology, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and The Ministry of Education of China, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhanhang Wang
- Department of Neurology, GuangDong 999 Brain Hospital, Guangzhou, China
| | - Li Xiao
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiaomiao Zhou
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Xu Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Fu
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Hao Chen
- Department of Neurology, The First Affiliated Hospital Nanchang University, Nanchang, China
| | - Juanjuan Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yanxia Zhou
- Department of Neurology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Juan Zhou
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou, China
| | - Liting Zhang
- Department of Neurology, Jiangxi Chest Hospital, Jiangxi, China
| | - Jing Zhou
- Department of Neurology, Foshan First People's Hospital, Foshan, China
| | - Ying Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Frank Petersen
- Division of Pulmonary Immune Diseases, Priority Area Chronic Lung Diseases, Research Center Borstel, Borstel, Germany
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xinhua Yu
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, Hainan, China
- Division of Pulmonary Immune Diseases, Priority Area Chronic Lung Diseases, Research Center Borstel, Borstel, Germany
| |
Collapse
|
36
|
Guo Z, Ren H, Chang Q, Liu R, Zhou X, Xue K, Sun T, Luo J, Wang F, Ge J. Lactobacilli-derived adjuvants combined with immunoinformatics-driven multi-epitope antigens based approach protects against Clostridium perfringens in a mouse model. Int J Biol Macromol 2024; 267:131475. [PMID: 38608984 DOI: 10.1016/j.ijbiomac.2024.131475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/14/2024]
Abstract
Clostridium perfringens is ubiquitously distributed and capable of secreting toxins, posing a significant threat to animal health. Infections caused by Clostridium perfringens, such as Necrotic Enteritis (NE), result in substantial economic losses to the livestock industry annually. However, there is no effective commercial vaccine available. Hence, we set out to propose an effective approach for multi-epitope subunit vaccine construction utilizing biomolecules. We utilized immunoinformatics to design a novel multi-epitope antigen against C. perfringens (CPMEA). Furthermore, we innovated novel bacterium-like particles (BLPs) through thermal acid treatment of various Lactobacillus strains and selected BLP23017 among them. Then, we detailed the structure of CPMEA and BLPs and utilized them to prepare a multi-epitope vaccine. Here, we showed that our vaccine provided full protection against C. perfringens infection after a single dose in a mouse model. Additionally, BLP23017 notably augmented the secretion of secretory immunoglobulin A (sIgA) and enhanced antibody production. We conclude that our vaccine possess safety and high efficacy, making it an excellent candidate for preventing C. perfringens infection. Moreover, we demonstrate our approach to vaccine construction and the preparation of BLP23017 with distinct advantages may contribute to the prevention of a wider array of diseases and the novel vaccine development.
Collapse
Affiliation(s)
- Zhiyuan Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Hongkun Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qingru Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Runhang Liu
- State Key Laboratory for Animal Disease control and prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Kun Xue
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Tong Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jilong Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Fang Wang
- State Key Laboratory for Animal Disease control and prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150030, China.
| |
Collapse
|
37
|
Akter A, Ananna NF, Ullah H, Islam S, Al Amin M, Kibria KMK, Mahmud S. Computational approach for identifying immunogenic epitopes and optimizing peptide vaccine through in-silico cloning against Mycoplasma genitalium. Heliyon 2024; 10:e28223. [PMID: 38596014 PMCID: PMC11002066 DOI: 10.1016/j.heliyon.2024.e28223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Mycoplasma genitalium is a pathogenic microorganism linked to a variety of severe health conditions including ovarian cancer, prostate cancer, HIV transmission, and sexually transmitted diseases. A more effective approach to address the challenges posed by this pathogen, given its high antibiotic resistance rates, could be the development of a peptide vaccine. In this study, we used experimentally validated 13 membrane proteins and their immunogenicity to identify suitable vaccine candidates. Thus, based on immunogenic properties and high conservation among other Mycoplasma genitalium sub-strains, the P110 surface protein is considered for further investigation. Later on, we identified T-cell epitopes and B-cell epitopes from the P110 protein to construct a multiepitope-based vaccine. As a result, the 'NIAPISFSFTPFTAA' T-cell epitope and 'KVKYESSGSNNISFDS' B-cell epitope have shown 99.53% and 87.50% population coverage along with 100% conservancy among the subspecies, and both epitopes were found to be non-allergenic. Furthermore, focusing on molecular docking analysis showed the lowest binding energy for MHC-I (-137.5 kcal/mol) and MHC-II (-183.3 kcal/mol), leading to a satisfactory binding strength between the T-cell epitopes and the MHC molecules. However, the constructed multiepitope vaccine (MEV) consisting of 54 amino acids demonstrates favorable characteristics for a vaccine candidate, including a theoretical pI of 4.25 with a scaled solubility of 0.812 and high antigenicity probabilities. Additionally, structural analyses reveal that the MEV displays substantial alpha helices and extended strands, vital for its immunogenicity. Molecular docking with the human Toll-like receptors TLR1/2 heterodimer shows strong binding affinity, reinforcing its potential to elicit an immune response. Our immune simulation analysis demonstrates immune memory development and robust immunity, while codon adaptation suggests optimal expression in E. coli using the pET-28a(+) vector. These findings collectively highlight the MEV's potential as a valuable vaccine candidate against M. genitalium.
Collapse
Affiliation(s)
- Asma Akter
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Natasha Farhin Ananna
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Hedayet Ullah
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Sirajul Islam
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Md Al Amin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - K M Kaderi Kibria
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Shahin Mahmud
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| |
Collapse
|
38
|
Samudrala M, Dhaveji S, Savsani K, Dakshanamurthy S. AutoEpiCollect, a Novel Machine Learning-Based GUI Software for Vaccine Design: Application to Pan-Cancer Vaccine Design Targeting PIK3CA Neoantigens. Bioengineering (Basel) 2024; 11:322. [PMID: 38671743 PMCID: PMC11048108 DOI: 10.3390/bioengineering11040322] [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: 02/27/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Previous epitope-based cancer vaccines have focused on analyzing a limited number of mutated epitopes and clinical variables preliminarily to experimental trials. As a result, relatively few positive clinical outcomes have been observed in epitope-based cancer vaccines. Further efforts are required to diversify the selection of mutated epitopes tailored to cancers with different genetic signatures. To address this, we developed the first version of AutoEpiCollect, a user-friendly GUI software, capable of generating safe and immunogenic epitopes from missense mutations in any oncogene of interest. This software incorporates a novel, machine learning-driven epitope ranking method, leveraging a probabilistic logistic regression model that is trained on experimental T-cell assay data. Users can freely download AutoEpiCollectGUI with its user guide for installing and running the software on GitHub. We used AutoEpiCollect to design a pan-cancer vaccine targeting missense mutations found in the proto-oncogene PIK3CA, which encodes the p110ɑ catalytic subunit of the PI3K kinase protein. We selected PIK3CA as our gene target due to its widespread prevalence as an oncokinase across various cancer types and its lack of presence as a gene target in clinical trials. After entering 49 distinct point mutations into AutoEpiCollect, we acquired 361 MHC Class I epitope/HLA pairs and 219 MHC Class II epitope/HLA pairs. From the 49 input point mutations, we identified MHC Class I epitopes targeting 34 of these mutations and MHC Class II epitopes targeting 11 mutations. Furthermore, to assess the potential impact of our pan-cancer vaccine, we employed PCOptim and PCOptim-CD to streamline our epitope list and attain optimized vaccine population coverage. We achieved a world population coverage of 98.09% for MHC Class I data and 81.81% for MHC Class II data. We used three of our predicted immunogenic epitopes to further construct 3D models of peptide-HLA and peptide-HLA-TCR complexes to analyze the epitope binding potential and TCR interactions. Future studies could aim to validate AutoEpiCollect's vaccine design in murine models affected by PIK3CA-mutated or other mutated tumor cells located in various tissue types. AutoEpiCollect streamlines the preclinical vaccine development process, saving time for thorough testing of vaccinations in experimental trials.
Collapse
Affiliation(s)
- Madhav Samudrala
- College of Arts and Sciences, The University of Virginia, Charlottesville, VA 22903, USA
| | | | - Kush Savsani
- College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA 22043, USA
| | - Sivanesan Dakshanamurthy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| |
Collapse
|
39
|
Naveed M, Ali U, Aziz T, Naveed R, Mahmood S, Khan MM, Alharbi M, Albekairi TH, Alasmari AF. An Aedes-Anopheles Vaccine Candidate Supplemented with BCG Epitopes Against the Aedes and Anopheles Genera to Overcome Hypersensitivity to Mosquito Bites. Acta Parasitol 2024; 69:483-504. [PMID: 38194049 DOI: 10.1007/s11686-023-00771-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Skeeter syndrome is a severe local allergic response to mosquito bites that is accompanied by considerable inflammation and, in some cases, a systemic response like fever. People with the syndrome develop serious allergies, ranging from rashes to anaphylaxis or shock. The few available studies on mosquito venom immunotherapy have utilized whole-body preparations and small sample sizes. Still, owing to their little success, vaccination remains a promising alternative as well as a permanent solution for infections like Skeeter's. METHODS This study, therefore, illustrated the construction of an epitope-based vaccine candidate against Skeeter Syndrome using established immunoinformatic techniques. We selected three species of mosquitoes, Anopheles melas, Anopheles funestus, and Aedes aegypti, to derive salivary antigens usually found in mosquito bites. Our construct was also supplemented with bacterial epitopes known to elicit a strong TH1 response and suppress TH2 stimulation that is predicted to reduce hypersensitivity against the bites. RESULTS A quality factor of 98.9496, instability index of 38.55, aliphatic index of 79.42, solubility of 0.934747, and GRAVY score of -0.02 indicated the structural (tertiary and secondary) stability, thermostability, solubility, and hydrophilicity of the construct, respectively. The designed Aedes-Anopheles vaccine (AAV) candidate was predicted to be flexible and less prone to deformability with an eigenvalue of 1.5911e-9 and perfected the human immune response against Skeeter (hypersensitivity) and many mosquito-associated diseases as we noted the production of 30,000 Th1 cells per mm3 with little (insignificant production of Th2 cells. The designed vaccine also revealed stable interactions with the pattern recognition receptors of the host. The TLR2/vaccine complex interacted with a free energy of - 1069.2 kcal/mol with 26 interactions, whereas the NLRP3/vaccine complex interacted with a free energy of - 1081.2 kcal/mol with 16 molecular interactions. CONCLUSION Although being a pure in-silico study, the in-depth analysis performed herein speaks volumes of the potency of the designed vaccine candidate predicting that the proposition can withstand rigorous in-vitro and in-vivo clinical trials and may proceed to become the first preventative immunotherapy against mosquito bite allergy.
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
- Department of Agriculture, University of Ioannina Arta, 47100, Arta, Greece.
| | - Rida Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan
| | - Sarmad Mahmood
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan
| | - Muhammad Mustajab Khan
- Department of Biotechnology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
40
|
Zhou M, Zhao F, Yu L, Liu J, Wang J, Zhang JZH. An Efficient Approach to the Accurate Prediction of Mutational Effects in Antigen Binding to the MHC1. Molecules 2024; 29:881. [PMID: 38398632 PMCID: PMC10892774 DOI: 10.3390/molecules29040881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
The major histocompatibility complex (MHC) can recognize and bind to external peptides to generate effective immune responses by presenting the peptides to T cells. Therefore, understanding the binding modes of peptide-MHC complexes (pMHC) and predicting the binding affinity of pMHCs play a crucial role in the rational design of peptide vaccines. In this study, we employed molecular dynamics (MD) simulations and free energy calculations with an Alanine Scanning with Generalized Born and Interaction Entropy (ASGBIE) method to investigate the protein-peptide interaction between HLA-A*02:01 and the G9209 peptide derived from the melanoma antigen gp100. The energy contribution of individual residue was calculated using alanine scanning, and hotspots on both the MHC and the peptides were identified. Our study shows that the pMHC binding is dominated by the van der Waals interactions. Furthermore, we optimized the ASGBIE method, achieving a Pearson correlation coefficient of 0.91 between predicted and experimental binding affinity for mutated antigens. This represents a significant improvement over the conventional MM/GBSA method, which yields a Pearson correlation coefficient of 0.22. The computational protocol developed in this study can be applied to the computational screening of antigens for the MHC1 as well as other protein-peptide binding systems.
Collapse
Affiliation(s)
- Mengchen Zhou
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Key Laboratory of Green Chemistry & Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China;
| | - Fanyu Zhao
- NYU-ECNU Center for Computational Chemistry and Shanghai Frontiers Science Center of AI and DL, NYU Shanghai, 567 West Yangsi Road, Shanghai 200126, China;
| | - Lan Yu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jinfeng Liu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jian Wang
- Faculty of Synthetic Biology and Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - John Z. H. Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Key Laboratory of Green Chemistry & Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China;
- NYU-ECNU Center for Computational Chemistry and Shanghai Frontiers Science Center of AI and DL, NYU Shanghai, 567 West Yangsi Road, Shanghai 200126, China;
- Faculty of Synthetic Biology and Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Department of Chemistry, New York University, New York, NY 10003, USA
- Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
41
|
Khazaei S, Varela-Calviño R, Rad-Malekshahi M, Quattrini F, Jokar S, Rezaei N, Balalaie S, Haririan I, Csaba N, Garcia-Fuentes M. Self-assembled peptide/polymer hybrid nanoplatform for cancer immunostimulating therapies. Drug Deliv Transl Res 2024; 14:455-473. [PMID: 37721693 PMCID: PMC10761384 DOI: 10.1007/s13346-023-01410-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 09/19/2023]
Abstract
Integrating peptide epitopes in self-assembling materials is a successful strategy to obtain nanovaccines with high antigen density and improved efficacy. In this study, self-assembling peptides containing MAGE-A3/PADRE epitopes were designed to generate functional therapeutic nanovaccines. To achieve higher stability, peptide/polymer hybrid nanoparticles were formulated by controlled self-assembly of the engineered peptides. The nanoparticles showed good biocompatibility to both human red blood- and dendritic cells. Incubation of the nanoparticles with immature dendritic cells triggered immune effects that ultimately activated CD8 + cells. The antigen-specific and IgG antibody responses of healthy C57BL/6 mice vaccinated with the nanoparticles were analyzed. The in vivo results indicate a specific response to the nanovaccines, mainly mediated through a cellular pathway. This research indicates that the immunogenicity of peptide epitope vaccines can be effectively enhanced by developing self-assembled peptide-polymer hybrid nanostructures.
Collapse
Affiliation(s)
- Saeedeh Khazaei
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ruben Varela-Calviño
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Federico Quattrini
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Safura Jokar
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Noemi Csaba
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
42
|
Nguyen TL, Kim H. Designing a Multiepitope Vaccine against Eastern Equine Encephalitis Virus: Immunoinformatics and Computational Approaches. ACS OMEGA 2024; 9:1092-1105. [PMID: 38222668 PMCID: PMC10785064 DOI: 10.1021/acsomega.3c07322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 01/16/2024]
Abstract
Eastern equine encephalitis virus (EEEV) is a significant threat to human and animal populations, causing severe encephalitis, often leading to long-term neurological complications and even mortality. Despite this, no approved antiviral treatments or EEEV human vaccines currently exist. In response, we utilized immunoinformatics and computational approaches to design a multiepitope vaccine candidate for EEEV. By screening the structural polyprotein of EEEV, we predicted both T-cell and linear B-cell epitopes. These epitopes underwent comprehensive evaluations for their antigenicity, toxicity, and allergenicity. From these evaluations, we selected ten epitopes highly suitable for vaccine design, which were connected with adjuvants using a stable linker. The resulting vaccine construct demonstrated exceptional antigenic, nontoxic, nonallergenic, and physicochemical properties. Subsequently, we employed molecular docking and molecular dynamics simulations to reveal a stable interaction pattern between the vaccine candidate and Toll-like receptor 5. Besides, computational immune simulations predicted the vaccine's capability to induce robust immune responses. Our study addresses the urgent need for effective EEEV preventive strategies and offers valuable insights for EEEV vaccine development. As EEEV poses a severe threat with potential spread due to climate change, our research provides a crucial step in enhancing public health defenses against this menacing zoonotic disease.
Collapse
Affiliation(s)
- Truc Ly Nguyen
- Department
of Agricultural Biotechnology and Research Institute of Agriculture
and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Heebal Kim
- Department
of Agricultural Biotechnology and Research Institute of Agriculture
and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary
Program in Bioinformatics, Seoul National
University, Seoul 08826, Republic
of Korea
- eGnome,
Inc., Seoul 05836, Republic of Korea
| |
Collapse
|
43
|
Biernacki MA, Lok J, Black RG, Foster KA, Cummings C, Woodward KB, Monahan T, Oehler VG, Stirewalt DL, Wu D, Rongvaux A, Deeg HJ, Bleakley M. Discovery of U2AF1 neoantigens in myeloid neoplasms. J Immunother Cancer 2023; 11:e007490. [PMID: 38164756 PMCID: PMC10729103 DOI: 10.1136/jitc-2023-007490] [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] [Accepted: 11/19/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Myelodysplastic syndromes (MDS) arise from somatic mutations acquired in hematopoietic stem and progenitor cells, causing cytopenias and predisposing to transformation into secondary acute myeloid leukemia (sAML). Recurrent mutations in spliceosome genes, including U2AF1, are attractive therapeutic targets as they are prevalent in MDS and sAML, arise early in neoplastic cells, and are generally absent from normal cells, including normal hematopoietic cells. MDS and sAML are susceptible to T cell-mediated killing, and thus engineered T-cell immunotherapies hold promise for their treatment. We hypothesized that targeting spliceosome mutation-derived neoantigens with transgenic T-cell receptor (TCR) T cells would selectively eradicate malignant cells in MDS and sAML. METHODS We identified candidate neoantigen epitopes from recurrent protein-coding mutations in the spliceosome genes SRSF2 and U2AF1 using a multistep in silico process. Candidate epitopes predicted to bind human leukocyte antigen (HLA) class I, be processed and presented from the parent protein, and not to be subject to tolerance then underwent in vitro immunogenicity screening. CD8+ T cells recognizing immunogenic neoantigen epitopes were evaluated in in vitro assays to assess functional avidity, confirm the predicted HLA restriction, the potential for recognition of similar peptides, and the ability to kill neoplastic cells in an antigen-specific manner. Neoantigen-specific TCR were sequenced, cloned into lentiviral vectors, and transduced into third-party T cells after knock-out of endogenous TCR, then tested in vitro for specificity and ability to kill neoplastic myeloid cells presenting the neoantigen. The efficacy of neoantigen-specific T cells was evaluated in vivo in a murine cell line-derived xenograft model. RESULTS We identified two neoantigens created from a recurrent mutation in U2AF1, isolated CD8+ T cells specific for the neoantigens, and demonstrated that transferring their TCR to third-party CD8+ T cells is feasible and confers specificity for the U2AF1 neoantigens. Finally, we showed that these neoantigen-specific TCR-T cells do not recognize normal hematopoietic cells but efficiently kill malignant myeloid cells bearing the specific U2AF1 mutation, including primary cells, in vitro and in vivo. CONCLUSIONS These data serve as proof-of-concept for developing precision medicine approaches that use neoantigen-directed T-cell receptor-transduced T cells to treat MDS and sAML.
Collapse
MESH Headings
- Humans
- Mice
- Animals
- CD8-Positive T-Lymphocytes
- Splicing Factor U2AF/genetics
- Splicing Factor U2AF/metabolism
- Antigens, Neoplasm
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/metabolism
- Epitopes/metabolism
Collapse
Affiliation(s)
- Melinda Ann Biernacki
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jessica Lok
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Ralph Graeme Black
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kimberly A Foster
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Carrie Cummings
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kyle B Woodward
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tim Monahan
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Vivian G Oehler
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Derek L Stirewalt
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - David Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Anthony Rongvaux
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Hans Joachim Deeg
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Marie Bleakley
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
44
|
Mao Y, Xiao X, Zhang J, Mou X, Zhao W. Designing a multi-epitope vaccine against Peptostreptococcus anaerobius based on an immunoinformatics approach. Synth Syst Biotechnol 2023; 8:757-770. [PMID: 38099061 PMCID: PMC10720267 DOI: 10.1016/j.synbio.2023.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/15/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023] Open
Abstract
Peptostreptococcus anaerobius is an anaerobic bacterium, which has been found selectively en-riched in the fecal and mucosal microbiota of colorectal cancer (CRC) patients. Emerging evidence suggest P. anaerobius may contribute to the development of CRC in human. In this study, we designed a multi-epitope chimeric vaccine against P. anaerobius PCWBR2, a recently identified adhesin that interacts directly with colon cell lines by binding α2/β1 integrin frequently overexpressed in human CRC tumors and cell lines. Immunoinformatics tools predicted six cytotoxic T lymphocyte epitopes, five helper T lymphocyte epitopes, and six linear B lymphocyte epitopes. The predicted epitopes were joined with AAY or GPGPG linkers and a previously reported TLR4 agonist was added to the vaccine construct's N terminal as an adjuvant using EAAAK linkers and the order of epitopes was optimized. Further in silico analysis revealed that the vaccine construct possesses satisfactory antigenicity, allergenicity, solubility, physicochemical properties, adjuvant-TLR4 molecular docking, and immune profile characteristics. Our study provided a promising design for vaccines against P. anaerobius.
Collapse
Affiliation(s)
- Yudan Mao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Xianzun Xiao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Jie Zhang
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Xiangyu Mou
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Wenjing Zhao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
45
|
Amin Rani N, Moin AT, Patil R, Barketullah Robin T, Zubair T, Nawal N, Sami MRS, Morshed MM, Zhai J, Xue M, Hossain M, Zheng C, Abul Manchur M, Islam NN. Designing a polyvalent vaccine targeting multiple strains of varicella zoster virus using integrated bioinformatics approaches. Front Microbiol 2023; 14:1291868. [PMID: 38075876 PMCID: PMC10704101 DOI: 10.3389/fmicb.2023.1291868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2024] Open
Abstract
The Varicella Zoster Virus (VZV) presents a global health challenge due to its dual manifestations of chickenpox and shingles. Despite vaccination efforts, incomplete coverage, and waning immunity lead to recurrent infections, especially in aging and immunocompromised individuals. Existing vaccines prevent chickenpox but can trigger the reactivation of shingles. To address these limitations, we propose a polyvalent multiepitope subunit vaccine targeting key envelope glycoproteins of VZV. Through bioinformatics approaches, we selected six glycoproteins that are crucial for viral infection. Epitope mapping led to the identification of cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL), and B cell linear (LBL) epitopes. Incorporating strong immunostimulants, we designed two vaccine constructs, demonstrating high antigenicity, solubility, stability, and compatibility with Toll-like receptors (TLRs). Molecular docking and dynamics simulations underscored the stability and affinity of the vaccine constructs with TLRs. These findings lay the foundation for a comprehensive solution to VZV infections, addressing the challenges of incomplete immunity and shingles reactivation. By employing advanced immunoinformatics and dynamics strategies, we have developed a promising polyvalent multiepitope subunit vaccine candidate, poised to enhance protection against VZV and its associated diseases. Further validation through in vivo studies is crucial to confirm the effectiveness and potential of the vaccine to curb the spread of VZV. This innovative approach not only contributes to VZV control but also offers insights into tailored vaccine design strategies against complex viral pathogens.
Collapse
Affiliation(s)
- Nurul Amin Rani
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Rajesh Patil
- Sinhgad Technical Education Society’s, Department of Pharmaceutical Chemistry, Sinhgad College of Pharmacy, Pune, India
| | - Tanjin Barketullah Robin
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | | | - Nafisa Nawal
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Md. Razwan Sardar Sami
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Md Masud Morshed
- Department of Pharmacy, International Islamic University Chittagong, Chattogram, Bangladesh
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mohabbat Hossain
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Mohammed Abul Manchur
- Department of Microbiology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Nazneen Naher Islam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| |
Collapse
|
46
|
Wu A, Wang Y, Ali A, Xu Z, Zhang D, Zhumanov K, Sheng J, Yi J. Design of a multi-epitope vaccine against brucellosis fused to IgG-fc by an immunoinformatics approach. Front Vet Sci 2023; 10:1238634. [PMID: 37937155 PMCID: PMC10625910 DOI: 10.3389/fvets.2023.1238634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Brucella, a type of intracellular Gram-negative bacterium, has unique features and acts as a zoonotic pathogen. It can lead to abortion and infertility in animals. Eliminating brucellosis becomes very challenging once it spreads among both humans and animals, putting a heavy burden on livestock and people worldwide. Given the increasing spread of brucellosis, it is crucial to develop improved vaccines for susceptible animals to reduce the disease's impact. Methods In this study, we effectively used an immunoinformatics approach with advanced computer software to carefully identify and analyze important antigenic parts of Brucella abortus. Subsequently, we skillfully designed chimeric peptides to enhance the vaccine's strength and effectiveness. We used computer programs to find four important parts of the Brucella bacteria that our immune system recognizes. Then, we carefully looked for eight parts that are recognized by a type of white blood cell called cytotoxic T cells, six parts recognized by T helper cells, and four parts recognized by B cells. We connected these parts together using a special link, creating a strong new vaccine. To make the vaccine even better, we added some extra parts called molecular adjuvants. These included something called human β-defensins 3 (hBD-3) that we found in a database, and another part that helps the immune system called PADRE. We attached these extra parts to the beginning of the vaccine. In a new and clever way, we made the vaccine even stronger by attaching a part from a mouse's immune system to the end of it. This created a new kind of vaccine called MEV-Fc. We used advanced computer methods to study how well the MEV-Fc vaccine interacts with certain receptors in the body (TLR-2 and TLR-4). Results In the end, Immunosimulation predictions showed that the MEV-Fc vaccine can make the immune system respond strongly, both in terms of cells and antibodies. Discussion In summary, our results provide novel insights for the development of Brucella vaccines. Although further laboratory experiments are required to assess its protective effect.
Collapse
Affiliation(s)
- Aodi Wu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Yueli Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Adnan Ali
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Zhenyu Xu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Dongsheng Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Kairat Zhumanov
- College of Veterinary Medicine, Kazakhstan Kazakh State Agricultural University, Almaty, Kazakhstan
| | - Jinliang Sheng
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
47
|
Moin AT, Rani NA, Ullah MA, Patil RB, Robin TB, Nawal N, Zubair T, Mahamud SI, Sakib MN, Islam NN, Khaleque MA, Absar N, Shohael AM. An immunoinformatics and extended molecular dynamics approach for designing a polyvalent vaccine against multiple strains of Human T-lymphotropic virus (HTLV). PLoS One 2023; 18:e0287416. [PMID: 37682972 PMCID: PMC10490984 DOI: 10.1371/journal.pone.0287416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
Human T-lymphotropic virus (HTLV), a group of retroviruses belonging to the oncovirus family, has long been associated with various inflammatory and immunosuppressive disorders. At present, there is no approved vaccine capable of effectively combating all the highly pathogenic strains of HTLV that makes this group of viruses a potential threat to human health. To combat the devastating impact of any potential future outbreak caused by this virus group, our study employed a reverse vaccinology approach to design a novel polyvalent vaccine targeting the highly virulent subtypes of HTLV. Moreover, we comprehensively analyzed the molecular interactions between the designed vaccine and corresponding Toll-like receptors (TLRs), providing valuable insights for future research on preventing and managing HTLV-related diseases and any possible outbreaks. The vaccine was designed by focusing on the envelope glycoprotein gp62, a crucial protein involved in the infectious process and immune mechanisms of HTLV inside the human body. Epitope mapping identified T cell and B cell epitopes with low binding energies, ensuring their immunogenicity and safety. Linkers and adjuvants were incorporated to enhance the vaccine's stability, antigenicity, and immunogenicity. Initially, two vaccine constructs were formulated, and among them, vaccine construct-2 exhibited superior solubility and structural stability. Molecular docking analyses also revealed strong binding affinity between the vaccine construct-2 and both targeted TLR2 and TLR4. Molecular dynamics simulations demonstrated enhanced stability, compactness, and consistent hydrogen bonding within TLR-vaccine complexes, suggesting a strong binding affinity. The stability of the complexes was further corroborated by contact, free energy, structure, and MM-PBSA analyses. Consequently, our research proposes a vaccine targeting multiple HTLV subtypes, offering valuable insights into the molecular interactions between the vaccine and TLRs. These findings should contribute to developing effective preventive and treatment approaches against HTLV-related diseases and preventing possible outbreaks. However, future research should focus on in-depth validation through experimental studies to confirm the interactions identified in silico and to evaluate the vaccine's efficacy in relevant animal models and, eventually, in clinical trials.
Collapse
Affiliation(s)
- Abu Tayab Moin
- Faculty of Biological Sciences, Department of Genetic Engineering and Biotechnology, University of Chittagong, Chattogram, Bangladesh
| | - Nurul Amin Rani
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Md. Asad Ullah
- Faculty of Biological Sciences, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Rajesh B. Patil
- Department of Pharmaceutical Chemistry, Sinhgad Technical Education Society’s, Sinhgad College of Pharmacy, Maharashtra, India
| | - Tanjin Barketullah Robin
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Nafisa Nawal
- Faculty of Biological Sciences, Department of Genetic Engineering and Biotechnology, University of Chittagong, Chattogram, Bangladesh
| | | | - Syed Iftakhar Mahamud
- Faculty of Biological Sciences, Department of Microbiology, University of Chittagong, Chattogram, Bangladesh
| | - Mohammad Najmul Sakib
- Faculty of Biological Sciences, Department of Genetic Engineering and Biotechnology, University of Chittagong, Chattogram, Bangladesh
| | - Nafisa Nawal Islam
- Faculty of Biological Sciences, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Md. Abdul Khaleque
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| | - Nurul Absar
- Faculty of Basic Medical and Pharmaceutical Sciences, Department of Biochemistry and Biotechnology, University of Science & Technology Chittagong, Khulshi, Chittagong, Bangladesh
| | - Abdullah Mohammad Shohael
- Faculty of Biological Sciences, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka, Bangladesh
| |
Collapse
|
48
|
Matos AS, Invenção MDCV, Moura IAD, Freitas ACD, Batista MVDA. Immunoinformatics applications in the development of therapeutic vaccines against human papillomavirus-related infections and cervical cancer. Rev Med Virol 2023; 33:e2463. [PMID: 37291746 DOI: 10.1002/rmv.2463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/09/2023] [Accepted: 05/27/2023] [Indexed: 06/10/2023]
Abstract
The human papillomavirus (HPV) represents the most prevalent sexually transmitted infectious agent worldwide. HPV penetrates the epithelium through microlesions and establishes an infectious focus that can lead to the development of cervical cancer. Prophylactic HPV vaccines are available, but do not affect already-established infections. Using in silico prediction tools is a promising strategy for identifying and selecting vaccine candidate T cell epitopes. An advantage of this strategy is that epitopes can be selected according to the degree of conservation within a group of antigenic proteins. This makes achieving comprehensive genotypic coverage possible with a small set of epitopes. Therefore, this paper revises the general characteristics of HPV biology and the current knowledge on developing therapeutic peptide vaccines against HPV-related infections and cervical cancer.
Collapse
Affiliation(s)
- Alexandre Santos Matos
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, Sao Cristovao, Brazil
| | - Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, Sao Cristovao, Brazil
| |
Collapse
|
49
|
Mishra SK, Priya P, Rai GP, Haque R, Shanker A. Coevolution based immunoinformatics approach considering variability of epitopes to combat different strains: A case study using spike protein of SARS-CoV-2. Comput Biol Med 2023; 163:107233. [PMID: 37422941 DOI: 10.1016/j.compbiomed.2023.107233] [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: 12/07/2022] [Revised: 06/03/2023] [Accepted: 07/01/2023] [Indexed: 07/11/2023]
Abstract
In the recent past several vaccines were developed to combat the COVID-19 disease. Unfortunately, the protective efficacy of the current vaccines has been reduced due to the high mutation rate in SARS-CoV-2. Here, we successfully implemented a coevolution based immunoinformatics approach to design an epitope-based peptide vaccine considering variability in spike protein of SARS-CoV-2. The spike glycoprotein was investigated for B- and T-cell epitope prediction. Identified T-cell epitopes were mapped on previously reported coevolving amino acids in the spike protein to introduce mutation. The non-mutated and mutated vaccine components were constructed by selecting epitopes showing overlapping with the predicted B-cell epitopes and highest antigenicity. Selected epitopes were linked with the help of a linker to construct a single vaccine component. Non-mutated and mutated vaccine component sequences were modelled and validated. The in-silico expression level of the vaccine constructs (non-mutated and mutated) in E. coli K12 shows promising results. The molecular docking analysis of vaccine components with toll-like receptor 5 (TLR5) demonstrated strong binding affinity. The time series calculations including root mean square deviation (RMSD), radius of gyration (RGYR), and energy of the system over 100 ns trajectory obtained from all atom molecular dynamics simulation showed stability of the system. The combined coevolutionary and immunoinformatics approach used in this study will certainly help to design an effective peptide vaccine that may work against different strains of SARS-CoV-2. Moreover, the strategy used in this study can be implemented on other pathogens.
Collapse
Affiliation(s)
- Saurav Kumar Mishra
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India
| | - Prerna Priya
- Department of Botany, Purnea Mahila College, Purnia, Bihar, India
| | - Gyan Prakash Rai
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya, Bihar, India
| | - Asheesh Shanker
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India.
| |
Collapse
|
50
|
Berner F, Flatz L. Autoimmunity in immune checkpoint inhibitor-induced immune-related adverse events: A focus on autoimmune skin toxicity and pneumonitis. Immunol Rev 2023; 318:37-50. [PMID: 37548043 DOI: 10.1111/imr.13258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy. However, their use is frequently associated with immune-related adverse events (irAEs) potentially affecting any organ. The mechanisms mediating such irAEs remain poorly understood and biomarkers to predict the development of irAEs are lacking. Growing evidence shows the importance of self-antigens in mediating irAEs during ICI therapy, in particular the well-described melanocyte differentiation antigens in melanoma patients. This review will focus on two novel classes of self-antigens involved in mediating autoimmune skin toxicity and pneumonitis in non-small cell lung cancer patients treated with immunotherapy. T cells specific for these self-antigens are thought to not only mediate irAEs but are thought to simultaneously mediate anti-tumor responses and are therefore associated with both autoimmune toxicity and response to ICI therapy. We further discuss emerging cellular and proteomic immune signatures of irAEs that may serve as biomarkers to help predict which patients are at higher risk of developing these irAEs. The determination of new tumor antigens involved in ICI therapy and the identification of related biomarkers brings us a step forward in the mechanistic understanding of ICIs and will help to monitor patients at higher risk of developing irAEs. Lastly, we discuss the current challenges in collecting research samples for the study of ICI-related mechanisms and in distinguishing between immune signatures of response and those of irAEs.
Collapse
Affiliation(s)
- Fiamma Berner
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
- Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|