1
|
Feng H, Jin Y, Wu B. Strategies for neoantigen screening and immunogenicity validation in cancer immunotherapy (Review). Int J Oncol 2025; 66:43. [PMID: 40342048 PMCID: PMC12101193 DOI: 10.3892/ijo.2025.5749] [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: 02/01/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer immunotherapy stimulates and enhances antitumor immune responses to eliminate cancer cells. Neoantigens, which originate from specific mutations within tumor cells, are key targets in cancer immunotherapy. Neoantigens manifest as abnormal peptide fragments or protein segments that are uniquely expressed in tumor cells, making them highly immunogenic. As a result, they activate the immune system, particularly T cell‑mediated immune responses, effectively identifying and eliminating tumor cells. Certain tumor‑associated antigens that are abnormally expressed in normal host proteins in cancer cells are promising targets for immunotherapy. Neoantigens derived from mutated proteins in cancer cells offer true cancer specificity and are often highly immunogenic. Furthermore, most neoantigens are unique to each patient, highlighting the need for personalized treatment strategies. The precise identification and screening of neoantigens are key for improving treatment efficacy and developing individualized therapeutic plans. The neoantigen prediction process involves somatic mutation identification, human leukocyte antigen (HLA) typing, peptide processing and peptide‑HLA binding prediction. The present review summarizes the major current methods used for neoantigen screening, available computational tools and the advantages and limitations of various techniques. Additionally, the present review aimed to summarize experimental strategies for validating the immunogenicity of the predicted neoantigens, which will determine whether these neoantigens can effectively trigger immune responses, as well as challenges encountered during neoantigen screening, providing relevant recommendations for the optimization of neoantigen‑based immunotherapy.
Collapse
Affiliation(s)
- Hua Feng
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yuanting Jin
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, P.R. China
| | - Bin Wu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
2
|
da Silva GCS, Paraná VC, de Almeida Rego FF, Portela MM, Queiroz MB, Junior RC, da Silva CGR, Gois LL, Grassi MFR. Impact of mutations in immunodominant regions of SARS-CoV-2 variants on recognition by CD8+ T cell: An in silico analysis. J Infect Public Health 2025; 18:102803. [PMID: 40359819 DOI: 10.1016/j.jiph.2025.102803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/10/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND This study aimed to investigate whether mutations in the immunodominant regions of the S, M, and N proteins of the Gamma, Delta, and Omicron SARS-CoV-2 variants that circulated in Brazil affect the recognition of viral antigens by Brazilian HLA-I-restricted CD8+ T cell epitopes, using an in silico approach. METHODS Sequences of the Gamma (n = 36,174), Delta (n = 35,129), and Omicron (n = 336) variants were retrieved using GISAID. Consensus sequences were generated using Geneious software with NC045512 as a reference. Epitopes for the S, M, and N proteins of both the original and variant sequences were predicted using NetCTLpan 1.1, NetMHCpan 4.0, and VaxiJen v2.0. The positions occupied by these epitopes, with high probability of presentation, affinity to HLA molecules, and antigenicity, were identified as potentially immunodominant regions. RESULTS The S protein of the reference sequence (NC045512) and its variants contained 17 immunodominant regions. Delta showed the highest conservation (94.1 %, 16), followed by Gamma (82.3 %, 14) and Omicron (70.5 %, 12). Omicron exhibited the greatest mutational variability and had regions of increased antigenicity and two novel immunodominant regions with broader human leukocyte antigen (HLA) recognition. Additionally, Omicron lost two previously identified immunodominant regions and had one region of reduced antigenicity that did not affect HLA recognition. Gamma had mutations in three regions that increased both antigenicity and HLA recognition. Delta had only one mutated region with lower antigenicity, which did not affect HLA recognition. Notably, new immunodominant regions for the M and N proteins appeared in the Omicron variant. CONCLUSIONS Brazilian HLA-I-restricted CD8+ T cell epitopes from SARS-CoV-2 immunodominant regions are partially conserved in the Gamma, Delta, and Omicron variants circulating in Brazil, suggesting effective a cross-protective immune response that may help reduce COVID-19 severity and mortality.
Collapse
Affiliation(s)
| | - Victoria Cruz Paraná
- Fundação Oswaldo Cruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, Bahia, Brazil
| | - Filipe Ferreira de Almeida Rego
- Fundação Oswaldo Cruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, Bahia, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | | | - Mariana Barros Queiroz
- Universidade Federal da Bahia, Instituto de Ciências da Saúde, Departamento de Ciências de Biointeração, Salvador, Bahia, Brazil
| | - Raimundo Coutinho Junior
- Fundação Oswaldo Cruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, Bahia, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | | | - Luana Leandro Gois
- Fundação Oswaldo Cruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, Bahia, Brazil; Universidade Federal da Bahia, Instituto de Ciências da Saúde, Departamento de Ciências de Biointeração, Salvador, Bahia, Brazil
| | - Maria Fernanda Rios Grassi
- Fundação Oswaldo Cruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, Bahia, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil.
| |
Collapse
|
3
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
4
|
Khalaf WS, Morgan RN, Elkhatib WF. Clinical microbiology and artificial intelligence: Different applications, challenges, and future prospects. J Microbiol Methods 2025; 232-234:107125. [PMID: 40188989 DOI: 10.1016/j.mimet.2025.107125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/10/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Conventional clinical microbiological techniques are enhanced by the introduction of artificial intelligence (AI). Comprehensive data processing and analysis enabled the development of curated datasets that has been effectively used in training different AI algorithms. Recently, a number of machine learning (ML) and deep learning (DL) algorithms are developed and evaluated using diverse microbiological datasets. These datasets included spectral analysis (Raman and MALDI-TOF spectroscopy), microscopic images (Gram and acid fast stains), and genomic and protein sequences (whole genome sequencing (WGS) and protein data banks (PDBs)). The primary objective of these algorithms is to minimize the time, effort, and expenses linked to conventional analytical methods. Furthermore, AI algorithms are incorporated with quantitative structure-activity relationship (QSAR) models to predict novel antimicrobial agents that address the continuing surge of antimicrobial resistance. During the COVID-19 pandemic, AI algorithms played a crucial role in vaccine developments and the discovery of new antiviral agents, and introduced potential drug candidates via drug repurposing. However, despite their significant benefits, the implementation of AI encounters various challenges, including ethical considerations, the potential for bias, and errors related to data training. This review seeks to provide an overview of the most recent applications of artificial intelligence in clinical microbiology, with the intention of educating a wider audience of clinical practitioners regarding the current uses of machine learning algorithms and encouraging their implementation. Furthermore, it will discuss the challenges related to the incorporation of AI into clinical microbiology laboratories and examine future opportunities for AI within the realm of infectious disease epidemiology.
Collapse
Affiliation(s)
- Wafaa S Khalaf
- Department of Microbiology and Immunology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11751, Egypt.
| | - Radwa N Morgan
- National Centre for Radiation Research and Technology (NCRRT), Drug Radiation Research Department, Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt.
| | - Walid F Elkhatib
- Department of Microbiology & Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt; Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo 11566, Egypt.
| |
Collapse
|
5
|
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] [Download PDF] [Figures] [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
|
6
|
Xiong Z, Sneiderman CT, Kuminkoski CR, Reinheimer J, Schwegman L, Sever RE, Habib A, Hu B, Agnihotri S, Rajasundaram D, Zinn PO, Forsthuber TG, Pollack IF, Li X, Raphael I, Kohanbash G. Transcript-targeted antigen mapping reveals the potential of POSTN splicing junction epitopes in glioblastoma immunotherapy. Genes Immun 2025:10.1038/s41435-025-00326-6. [PMID: 40181162 DOI: 10.1038/s41435-025-00326-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Tumor antigens are crucial for T-cell mediated immunotherapy, but identified antigens for gliomas remain limited. Aberrant splicing variants are commonly expressed in tumors, resulting in unique tumor isoforms with potential antigenic properties. Herein, we analyzed multi-omics data from 587 glioma patients and assembled a library of putative tumor-enriched isoform antigens (TIA) and corresponding peptides presented on each HLA-I allele. We constructed an individual-specific TIA peptide candidate repertoire for each patient based on their TIA expression and HLA-I haplotypes. TIAs were highly expressed, enriched with glioma malignancy, and demonstrated strong HLA-binding affinity. We focused on periostin isoform-203 (POSTN-203), which was associated with poor survival of patients and contained multiple predicted HLA-restricted peptide epitopes. A selected HLA-A11-restricted peptide from POSTN-203 (POSTN-203A11) induced antigen-specific T-cell responses against both peptide-pulsed and POSTN-203-expressing glioma cells in an HLA-specific manner. Our findings highlight TIAs as a promising source of immunogenic antigens and POSTN-203 as a potential promising target for glioma immunotherapy.
Collapse
Affiliation(s)
- Zujian Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chaim T Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chloe R Kuminkoski
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jared Reinheimer
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lance Schwegman
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, USA
| | - ReidAnn E Sever
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ahmed Habib
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Pascal O Zinn
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas G Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Lara-Ramírez D, Santacruz-Tinoco CE, Ramón-Gallegos E, Muñoz-Medina JE. In silico design of Ebola virus Glycoprotein antigenic peptides as vaccine candidates. PLoS One 2025; 20:e0319496. [PMID: 40153397 PMCID: PMC11952221 DOI: 10.1371/journal.pone.0319496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/03/2025] [Indexed: 03/30/2025] Open
Abstract
Ebola virus (EBOV) is a filovirus that causes severe hemorrhagic fever and has a fatality rate between 50 and 90%. The vaccines were developed against the Ebola Zaire species; therefore, it is necessary to develop vaccines against other species to control future outbreaks. The objective of this work was to obtain vaccine candidate peptides against different EBOV species through the use of bioinformatics programs and servers that allow glycoprotein (GP) to be analyzed. GP sequences of various EBOV species that did not present gaps or unspecified amino acids or that were repeated (same year, region and laboratory) were downloaded from the NCBI database. A consensus sequence was generated and used to determine vaccine candidate peptides, which were evaluated, through a combination of servers and molecular dynamics, for their ability to interact with B and T lymphocytes, toxicity, allergenicity, solvent exposure, glycosylation, antigenicity, and presence in mature GP. Five vaccine candidate peptides were identified, of which PEP4 had the best characteristics evaluated in this study. PEP4 may be a potential candidate for the development of an EBOV vaccine.
Collapse
Affiliation(s)
- David Lara-Ramírez
- Environmental Cytopathology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico CityMexico
- División de Laboratorios Especializados. Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | | - Eva Ramón-Gallegos
- Environmental Cytopathology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico CityMexico
| | | |
Collapse
|
8
|
Iyyanar S, Ravi SN. Vaccine Development T-cell (MHC-I) Epitopes Identification Against the Indian HCV Genotype: An Approach Based on Immunoinformatic. Mol Biotechnol 2025:10.1007/s12033-025-01398-5. [PMID: 39994132 DOI: 10.1007/s12033-025-01398-5] [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: 08/07/2024] [Accepted: 01/30/2025] [Indexed: 02/26/2025]
Abstract
Hepatitis C virus (HCV) infects approximately 58 million individuals worldwide, often progressing to chronic liver disease, cirrhosis, and hepatocellular carcinoma. The viral envelope glycoproteins E1 and E2 are critical for HCV entry and serve as primary targets for neutralizing antibodies. Recent advancements in cryo-electron tomography have provided high-resolution structures (3.5 Å) of the E1E2 heterodimer, revealing interactions between the E1 and E2 ectodomains, as well as neutralizing antibody complexes (e.g., AR4A, AT1209, IGH505). This structural information facilitates the design of a synthetic peptide vaccine targeting conserved E1 and E2 regions. We suggest developing a vaccine tailored to the HLA-A*24:02 allele, the most prevalent in the Indian population. Epitope candidates will be screened using immunoinformatics tools, incorporating epitopes derived from epitope mapping with 7t6x protein structure modeling. Molecular docking studies will identify high-affinity interactions with human MHC-Class I alleles, using tools such as AutoDock and HADDOCK. GROMACS molecular dynamics simulations will assess peptide-HLA binding stability and dynamics. Among ten screened epitopes, KWEYVVLLF and QWQVLPCSF emerged as the most promising based on their toxicity profiles, conservation, and docking scores with HLA-A*24:02 (- 9.3 kcal/mol for KWEYVVLLF and - 225.34 kcal/mol for QWQVLPCSF). Molecular dynamics simulations indicated that the KWEY segment of KWEYVVLLF underwent structural changes, while the VVLLF region maintained stable binding to Chain A, suggesting immunogenic potential. These epitopes represent strong candidates for T-cell-based vaccines, and the reverse vaccinology approach, supported by computational tools, offers a population-specific strategy for HCV vaccine development, advancing precision immunotherapy.
Collapse
Affiliation(s)
- Sridevi Iyyanar
- Department of Biotechnology, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Sai Nandhini Ravi
- Department of Biotechnology, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, Tamil Nadu, India.
| |
Collapse
|
9
|
Sira EMJS, Banico EC, Fajardo LE, Odchimar NMO, Dela Cruz KM, Orosco FL. In silico design of multi-epitope vaccine candidate based on structural proteins of porcine reproductive and respiratory syndrome virus. Vet Immunol Immunopathol 2025; 280:110881. [PMID: 39847849 DOI: 10.1016/j.vetimm.2025.110881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/25/2025]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most common respiratory disease-causing viral agents. Swine infected with PRRSV exhibit severe respiratory symptoms and reproductive failure, leading to significant economic losses. To address this issue, inactivated and live-attenuated vaccines have been developed. However, the current commercially available PRRSV vaccines do not confer sufficient protection or have safety issues. The use of epitope-based subunit vaccines reduce safety risks by including only specific immunogenic portions of the antigens. To enhance immune protection, this study targeted multiple structural proteins of PRRSV, including GP2, GP3, GP4, GP5, membrane (M), envelope (E), GP5a, and nucleocapsid (N), to enable the discovery of novel epitopes. Thus, a reverse vaccinology approach was utilized to design a multi-epitope subunit vaccine construct against PRRSV. Using different tools, seven linear B cell, seven cytotoxic T cell, and three helper T cell epitopes were predicted to be safe, antigenic, and immunogenic. These epitopes were linked together, and a protein adjuvant, heparin-binding hemagglutinin, was added to increase the vaccine's immunogenicity. The construct was then docked to Toll-like receptor 4 (TLR4) to assess its ability to initiate the innate immune response. The final vaccine construct was determined to be antigenic, stable, non-allergenic, and soluble. Furthermore, the vaccine demonstrated stable binding to TLR4 based on coarse-grained and atomistic molecular dynamics simulations. Finally, the immune simulation of the vaccine construct showed a robust immune response against PRRSV. In this study, a candidate vaccine construct was successfully designed as a potential strategy against PRRSV.
Collapse
Affiliation(s)
- Ella Mae Joy S Sira
- Virology and Vaccine Research Program, Industrial Technology Development Institute, Department of Science and Technology, Bicutan, Taguig 1634, Philippines
| | - Edward C Banico
- Virology and Vaccine Research Program, Industrial Technology Development Institute, Department of Science and Technology, Bicutan, Taguig 1634, Philippines
| | - Lauren Emily Fajardo
- Virology and Vaccine Research Program, Industrial Technology Development Institute, Department of Science and Technology, Bicutan, Taguig 1634, Philippines
| | - Nyzar Mabeth O Odchimar
- Virology and Vaccine Research Program, Industrial Technology Development Institute, Department of Science and Technology, Bicutan, Taguig 1634, Philippines
| | | | - Fredmoore L Orosco
- Virology and Vaccine Research Program, Industrial Technology Development Institute, Department of Science and Technology, Bicutan, Taguig 1634, Philippines; Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila 1000, Philippines; S&T Fellows Program, Department of Science and Technology, Bicutan, Taguig 1634, Philippines.
| |
Collapse
|
10
|
Floudas CS, Sarkizova S, Ceccarelli M, Zheng W. Leveraging mRNA technology for antigen based immuno-oncology therapies. J Immunother Cancer 2025; 13:e010569. [PMID: 39848687 PMCID: PMC11784169 DOI: 10.1136/jitc-2024-010569] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/03/2025] [Indexed: 01/25/2025] Open
Abstract
The application of messenger RNA (mRNA) technology in antigen-based immuno-oncology therapies represents a significant advancement in cancer treatment. Cancer vaccines are an effective combinatorial partner to sensitize the host immune system to the tumor and boost the efficacy of immune therapies. Selecting suitable tumor antigens is the key step to devising effective vaccinations and amplifying the immune response. Tumor neoantigens are de novo epitopes derived from somatic mutations, avoiding T-cell central tolerance of self-epitopes and inducing immune responses to tumors. The identification and prioritization of patient-specific tumor neoantigens are based on advanced computational algorithms taking advantage of the profiling with next-generation sequencing considering factors involved in human leukocyte antigen (HLA)-peptide-T-cell receptor (TCR) complex formation, including peptide presentation, HLA-peptide affinity, and TCR recognition. This review discusses the development and clinical application of mRNA vaccines in oncology, with a particular focus on recent clinical trials and the computational workflows and methodologies for identifying both shared and individual antigens. While this review centers on therapeutic mRNA vaccines targeting existing tumors, it does not cover preventative vaccines. Preclinical experimental validations are crucial in cancer vaccine development, but we emphasize the computational approaches that facilitate neoantigen selection and design, highlighting their role in advancing mRNA vaccine development. The versatility and rapid development potential of mRNA make it an ideal platform for personalized neoantigen immunotherapy. We explore various strategies for antigen target identification, including tumor-associated and tumor-specific antigens and the computational tools used to predict epitopes capable of eliciting strong immune responses. We address key design considerations for enhancing the immunogenicity and stability of mRNA vaccines, as well as emerging trends and challenges in the field. This comprehensive overview highlights the therapeutic potential of mRNA-based cancer vaccines and underscores ongoing research efforts aimed at optimizing these therapies for improved clinical outcomes.
Collapse
Affiliation(s)
- Charalampos S Floudas
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Wei Zheng
- Moderna, Inc, Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Nejabat S, Khomartash MS, Mohammadimehr M, Adloo Z, Zanchi FB, Ghorbani M, Nezafat N. Immunoinformatics approach: Developing a multi-epitope vaccine with novel carriers targeting monkeypox virus. FASEB J 2024; 38:e70257. [PMID: 39679938 DOI: 10.1096/fj.202400757rr] [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: 10/12/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
Since May 2022, the global spread of monkeypox virus (MPXV) has presented a significant threat to public health. Despite this, there are limited preventive measures available. In this study, different computational tools were employed to design a multi-epitope vaccine targeting MPXV. Three key MPXV proteins, M1R, B6R, and F3L, were chosen for epitope selection, guided by bioinformatic analyses to identify immunodominant epitopes for T- and B-cell activation. To enhance immune stimulation and facilitate targeted delivery of the vaccine to specific cells, the selected epitopes were linked to novel carriers, including the extracellular domain of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), a 12-mer Clec9a binding peptide (CBP-12), and a Toll-like receptor 2 (TLR2) peptide ligand. The designed vaccine construct exhibited strong antigenicity along with nonallergenic and nontoxic properties, with favorable physicochemical characteristics. The validated vaccine's tertiary structure underwent evaluation for interactions with CD80/86, Clec9a, and TLR2 through molecular docking and molecular dynamics simulation. The results ensured the vaccine's stability and high affinity for the aforementioned receptors. In silico immune simulations studies revealed robust innate and adaptive immune responses, including enhanced mucosal immunity essential for protection against MPXV. Ultimately, the DNA sequence of the vaccine construct was synthesized and successfully cloned into the pET-22b(+) vector. Our study, through integration of computational predictions, suggests the proposed vaccine's potential efficacy in safeguarding against MPXV; however, further in vitro and in vivo validations are imperative to assess real-world effectiveness and safety.
Collapse
Affiliation(s)
- Sajjad Nejabat
- Science and Technology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mojgan Mohammadimehr
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences, Faculty of Paramedicine, AJA University of Medical Sciences, Tehran, Iran
| | - Zahra Adloo
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fernando Berton Zanchi
- Laboratório de Bioinformática e Química Medicinal (LABIOQUIM), Fundação Oswaldo Cruz Rondônia, Porto Velho, Brazil
| | - Mahdi Ghorbani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Mi X, Li S, Ye Z, Dai Z, Ding B, Sun B, Shen Y, Xiao Z. LRMAHpan: a novel tool for multi-allelic HLA presentation prediction using Resnet-based and LSTM-based neural networks. Front Immunol 2024; 15:1478201. [PMID: 39669561 PMCID: PMC11634944 DOI: 10.3389/fimmu.2024.1478201] [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: 08/09/2024] [Accepted: 10/30/2024] [Indexed: 12/14/2024] Open
Abstract
Introduction The identification of peptides eluted from HLA complexes by mass spectrometry (MS) can provide critical data for deep learning models of antigen presentation prediction and promote neoantigen vaccine design. A major challenge remains in determining which HLA allele eluted peptides correspond to. Methods To address this, we present a tool for prediction of multiple allele (MA) presentation called LRMAHpan, which integrates LSTM network and ResNet_CA network for antigen processing and presentation prediction. We trained and tested the LRMAHpan BA (binding affinity) and the LRMAHpan AP (antigen processing) models using mass spectrometry data, subsequently combined them into the LRMAHpan PS (presentation score) model. Our approach is based on a novel pHLA encoding method that enables the integration of neoantigen prediction tasks into computer vision methods. This method aggregates MA data into a multichannel matrix and incorporates peptide sequences to efficiently capture binding signals. Results LRMAHpan outperforms standard predictors such as NetMHCpan 4.1, MHCflurry 2.0, and TransPHLA in terms of positive predictive value (PPV) when applied to MA data. Additionally, it can accommodate peptides of variable lengths and predict HLA class I and II presentation. We also predicted neoantigens in a cohort of metastatic melanoma patients, identifying several shared neoantigens. Discussion Our results demonstrate that LRMAHpan significantly improves the accuracy of antigen presentation predictions.
Collapse
Affiliation(s)
- Xue Mi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Shaohao Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zheng Ye
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhu Dai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Bo Ding
- Department of Obstetrics and Gynecoloty, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bo Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yang Shen
- Department of Obstetrics and Gynecoloty, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Sports Health Research Institute, Institute of Sports and Health, Nanjing, China
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Jiangsu Sports Health Research Institute, Institute of Sports and Health, Nanjing, China
| |
Collapse
|
13
|
Pretti MAM, Vieira GF, Boroni M, Bonamino MH. Unveiling cross-reactivity: implications for immune response modulation in cancer. Brief Bioinform 2024; 26:bbaf012. [PMID: 39831892 PMCID: PMC11744606 DOI: 10.1093/bib/bbaf012] [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: 09/23/2024] [Revised: 12/03/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
Antigen recognition by CD8+ T-cell receptors (TCR) is crucial for immune responses to pathogens and tumors. TCRs are cross-reactive, a single TCR can recognize multiple peptide-Human Leukocyte Antigen (HLA) complexes. The study of cross-reactivity can support the development of therapies focusing on immune modulation, such as the expansion of pre-existing T-cell clones to fight pathogens and tumors. The peptide-HLA (pHLA) surface has previously been used to identify TCR cross-reactivities. In the present work, we sought to perform a comprehensive analysis of peptide-HLA by selecting thousands of human and viral epitopes. We profit from established docking models to identify features from different spatial perspectives of HLA-A*02:01, explore similarities between self and non-self epitopes, and list potential cross-reactive epitopes of therapeutic interest. A total of 2631 unique epitopes from representative viral proteins or human proteins were modeled. We were able to demonstrate that cross-reactive CDR3 sequences from public databases recognize epitopes with similar electrostatic potential, charge, and spatial location. Using data from published studies that measured T-cell reactivity to mutated epitopes, we observed a negative correlation between epitope dissimilarity and T-cell activation. Most analysed cancer epitopes were more similar to self epitopes, yet we identified features distinguishing those more similar to viral antigens. Finally, we enumerated potential cross-reactivities between tumoral and viral epitopes and highlighted some challenges in their identification for therapeutic use. Moreover, the thousands of peptide-HLA complexes generated in our work constitute a valuable resource to study T-cell cross-reactivity.
Collapse
Affiliation(s)
- Marco Antônio M Pretti
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Program of Cell and Gene Therapy, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Gustavo Fioravanti Vieira
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil
- Postgraduate Program in Health and Human Development, La Salle University, Canoas, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martín H Bonamino
- Program of Cell and Gene Therapy, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Tuhin IA, Mia MR, Islam MM, Mahmud I, Gongora HF, Rios CU, Ashraf I, Samad MA. StackIL10: A stacking ensemble model for the improved prediction of IL-10 inducing peptides. PLoS One 2024; 19:e0313835. [PMID: 39541341 PMCID: PMC11563426 DOI: 10.1371/journal.pone.0313835] [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: 01/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Interleukin-10, a highly effective cytokine recognized for its anti-inflammatory properties, plays a critical role in the immune system. In addition to its well-documented capacity to mitigate inflammation, IL-10 can unexpectedly demonstrate pro-inflammatory characteristics under specific circumstances. The presence of both aspects emphasizes the vital need to identify the IL-10-induced peptide. To mitigate the drawbacks of manual identification, which include its high cost, this study introduces StackIL10, an ensemble learning model based on stacking, to identify IL-10-inducing peptides in a precise and efficient manner. Ten Amino-acid-composition-based Feature Extraction approaches are considered. The StackIL10, stacking ensemble, the model with five optimized Machine Learning Algorithm (specifically LGBM, RF, SVM, Decision Tree, KNN) as the base learners and a Logistic Regression as the meta learner was constructed, and the identification rate reached 91.7%, MCC of 0.833 with 0.9078 Specificity. Experiments were conducted to examine the impact of various enhancement techniques on the correctness of IL-10 Prediction. These experiments included comparisons between single models and various combinations of stacking-based ensemble models. It was demonstrated that the model proposed in this study was more effective than singular models and produced satisfactory results, thereby improving the identification of peptides that induce IL-10.
Collapse
Affiliation(s)
- Izaz Ahmmed Tuhin
- Department of Software Engineering, Daffodil International University, Daffodil Smart City (DSC), Savar, Dhaka, Bangladesh
| | - Md. Rajib Mia
- Department of Software Engineering, Daffodil International University, Daffodil Smart City (DSC), Savar, Dhaka, Bangladesh
| | - Md. Monirul Islam
- Department of Software Engineering, Daffodil International University, Daffodil Smart City (DSC), Savar, Dhaka, Bangladesh
| | - Imran Mahmud
- Department of Software Engineering, Daffodil International University, Daffodil Smart City (DSC), Savar, Dhaka, Bangladesh
| | - Henry Fabian Gongora
- Universidad Europea del Atlántico, Santander, Spain
- Universidad Internacional Iberoamericana Campeche, Campeche, México
- Universidad de La Romana, La Romana, República Dominicana
| | - Carlos Uc Rios
- Universidad Europea del Atlántico, Santander, Spain
- Universidad Internacional Iberoamericana Campeche, Campeche, México
- Universidad Internacional Iberoamericana Arecibo, Arecibo, Puerto Rico, United States of America
| | - Imran Ashraf
- Department of Information and Communication Engineering, Yeungnam University, Gyeongsangbuk-do, Gyeongsan-si, South Korea
| | - Md. Abdus Samad
- Department of Information and Communication Engineering, Yeungnam University, Gyeongsangbuk-do, Gyeongsan-si, South Korea
| |
Collapse
|
15
|
James SA, Joshua IA. Charting Peptide Shared Sequences Between 'Diabetes-Viruses' and Human Pancreatic Proteins, Their Structural and Autoimmune Implications. Bioinform Biol Insights 2024; 18:11779322241289936. [PMID: 39502449 PMCID: PMC11536397 DOI: 10.1177/11779322241289936] [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: 11/22/2023] [Accepted: 08/21/2024] [Indexed: 11/08/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic syndrome characterized by hyperglycaemia, polydipsia, polyuria, and weight loss, among others. The pathophysiology for the disorders is complex and results in pancreatic abnormal function. Viruses have also been implicated in the metabolic syndrome. This study charted peptides to investigate and predict the autoimmune potential of shared sequences between 8 viral species proteins (which we termed 'diabetes-viruses') and the human pancreatic proteins. The structure and immunological relevance of shared sequences between viruses reported in DM onset and human pancreatic proteins were analysed. At nonapeptide mapping between human pancreatic protein and 'diabetic-viruses', reveal 1064 shared sequences distributed among 454 humans and 4288 viral protein sequences. The viral results showed herpesviruses, enterovirus (EV), human endogenous retrovirus, influenza A viruses, rotavirus, and rubivirus sequences are hosted by the human pancreatic protein. The most common shared nonapeptide was AAAAAAAAA, present in 30 human nonredundant sequences. Among the viral species, the shared sequence NSLEVLFQG occurred in 18 nonredundant EVs protein, while occurring merely in 1 human protein, whereas LGLDIEIAT occurred in 8 influenza A viruses overlapping to 1 human protein and KDELSEARE occurred in 2 rotaviruses. The prediction of the location of the shared sequences in the protein structures, showed most of the shared sequences are exposed and located either on the surface or cleft relative to the entire protein structure. Besides, the peptides in the viral protein shareome were predicted computationally for binding to MHC molecules. Here analyses showed that the entire 1064 shared sequences predicted 203 to be either HLA-A or B supertype-restricted epitopes. Fifty-one of the putative epitopes matched reported HLA ligands/T-cell epitopes majorly coming from EV B supertype representative allele restrictions. These data, shared sequences, and epitope charts provide important insight into the role of viruses on the onset of DM and its implications.
Collapse
Affiliation(s)
- Stephen A James
- Department of Biochemistry, Kaduna State University, Kaduna, Nigeria
- School of Data Sciences, Centre of Bioinformatics, Perdana University, Kuala Lumpur, Malaysia
| | - Istifanus A Joshua
- Department of Community Medicine, College of Medicine, Kaduna State University, Kaduna, Nigeria
- Department of Community Medicine, College of Health Sciences, Federal University Wukari, Wukari, Nigeria
| |
Collapse
|
16
|
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: 6] [Impact Index Per Article: 6.0] [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
|
17
|
O’Brien H, Salm M, Morton LT, Szukszto M, O’Farrell F, Boulton C, King L, Bola SK, Becker PD, Craig A, Nielsen M, Samuels Y, Swanton C, Mansour MR, Hadrup SR, Quezada SA. A modular protein language modelling approach to immunogenicity prediction. PLoS Comput Biol 2024; 20:e1012511. [PMID: 39527593 PMCID: PMC11581412 DOI: 10.1371/journal.pcbi.1012511] [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: 06/14/2024] [Revised: 11/21/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
Neoantigen immunogenicity prediction is a highly challenging problem in the development of personalised medicines. Low reactivity rates in called neoantigens result in a difficult prediction scenario with limited training datasets. Here we describe ImmugenX, a modular protein language modelling approach to immunogenicity prediction for CD8+ reactive epitopes. ImmugenX comprises of a pMHC encoding module trained on three pMHC prediction tasks, an optional TCR encoding module and a set of context specific immunogenicity prediction head modules. Compared with state-of-the-art models for each task, ImmugenX's encoding module performs comparably or better on pMHC binding affinity, eluted ligand prediction and stability tasks. ImmugenX outperforms all compared models on pMHC immunogenicity prediction (Area under the receiver operating characteristic curve = 0.619, average precision: 0.514), with a 7% increase in average precision compared to the next best model. ImmugenX shows further improved performance on immunogenicity prediction with the integration of TCR context information. ImmugenX performance is further analysed for interpretability, which locates areas of weakness found across existing immunogenicity models and highlight possible biases in public datasets.
Collapse
Affiliation(s)
| | - Max Salm
- Achilles Therapeutics UK Ltd, United Kingdom
| | | | - Maciej Szukszto
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Charlotte Boulton
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Laurence King
- Achilles Therapeutics UK Ltd, United Kingdom
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Supreet Kaur Bola
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | | | | | | | | | | | - Marc R. Mansour
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
- Department of Developmental Biology and Cancer, Great Ormond Street Institute of Child Health, UCL
| | | | - Sergio A. Quezada
- Achilles Therapeutics UK Ltd, United Kingdom
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
18
|
Huber F, Arnaud M, Stevenson BJ, Michaux J, Benedetti F, Thevenet J, Bobisse S, Chiffelle J, Gehert T, Müller M, Pak H, Krämer AI, Altimiras ER, Racle J, Taillandier-Coindard M, Muehlethaler K, Auger A, Saugy D, Murgues B, Benyagoub A, Gfeller D, Laniti DD, Kandalaft L, Rodrigo BN, Bouchaab H, Tissot S, Coukos G, Harari A, Bassani-Sternberg M. A comprehensive proteogenomic pipeline for neoantigen discovery to advance personalized cancer immunotherapy. Nat Biotechnol 2024:10.1038/s41587-024-02420-y. [PMID: 39394480 DOI: 10.1038/s41587-024-02420-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
The accurate identification and prioritization of antigenic peptides is crucial for the development of personalized cancer immunotherapies. Publicly available pipelines to predict clinical neoantigens do not allow direct integration of mass spectrometry immunopeptidomics data, which can uncover antigenic peptides derived from various canonical and noncanonical sources. To address this, we present an end-to-end clinical proteogenomic pipeline, called NeoDisc, that combines state-of-the-art publicly available and in-house software for immunopeptidomics, genomics and transcriptomics with in silico tools for the identification, prediction and prioritization of tumor-specific and immunogenic antigens from multiple sources, including neoantigens, viral antigens, high-confidence tumor-specific antigens and tumor-specific noncanonical antigens. We demonstrate the superiority of NeoDisc in accurately prioritizing immunogenic neoantigens over recent prioritization pipelines. We showcase the various features offered by NeoDisc that enable both rule-based and machine-learning approaches for personalized antigen discovery and neoantigen cancer vaccine design. Additionally, we demonstrate how NeoDisc's multiomics integration identifies defects in the cellular antigen presentation machinery, which influence the heterogeneous tumor antigenic landscape.
Collapse
Affiliation(s)
- Florian Huber
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Marion Arnaud
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Brian J Stevenson
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Quartier Sorge, Bâtiment Amphipôle, Lausanne, Switzerland
| | - Justine Michaux
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Fabrizio Benedetti
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Jonathan Thevenet
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Sara Bobisse
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Johanna Chiffelle
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Talita Gehert
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Markus Müller
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Quartier Sorge, Bâtiment Amphipôle, Lausanne, Switzerland
| | - HuiSong Pak
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Anne I Krämer
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Emma Ricart Altimiras
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Julien Racle
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Quartier Sorge, Bâtiment Amphipôle, Lausanne, Switzerland
| | - Marie Taillandier-Coindard
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Katja Muehlethaler
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Aymeric Auger
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Damien Saugy
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Baptiste Murgues
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Abdelkader Benyagoub
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - David Gfeller
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Quartier Sorge, Bâtiment Amphipôle, Lausanne, Switzerland
| | - Denarda Dangaj Laniti
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Lana Kandalaft
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Blanca Navarro Rodrigo
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Hasna Bouchaab
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Department of Medical Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Stephanie Tissot
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
- AGORA Cancer Research Center, Lausanne, Switzerland.
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
19
|
Claudia B, Nugrahapraja H, Giri-Rachman EA. A multi-epitope self-amplifying mRNA SARS-CoV-2 vaccine design using a reverse vaccinology approach. Res Pharm Sci 2024; 19:520-548. [PMID: 39691299 PMCID: PMC11648349 DOI: 10.4103/rps.rps_91_23] [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: 05/25/2023] [Revised: 08/14/2024] [Accepted: 10/06/2024] [Indexed: 12/19/2024] Open
Abstract
Background and purpose Massive vaccine distribution is a crucial step to prevent the spread of SARS-CoV2 as the causative agent of COVID-19. This research aimed to design the multi-epitope self-amplifying mRNA (saRNA) vaccine from the spike and nucleocapsid proteins of SARS-CoV2. Experimental approach Commonly distributed constructions class I and II alleles of the Indonesian population were used to determine peptide sequences that trigger this population's high specificity T-cell response. The best vaccine candidate was selected through the analysis of tertiary structure validation and molecular docking of each candidate with TLR-4, TLR-8, HLA-A*24:02, and HLA-DRB1*04:05. The selected multi-epitope vaccine combined with the gene encoding the replication machinery that allows the RNA amplification in the host cell. Findings/Results Seven B-cell and four T-cell epitopes from the protein target were highly antigenic and conserved, non-allergen, non-toxic, and hydrophilic. Tertiary structure validation then determined the best multi-epitope construction with 269 AA in length containing hBD-2 adjuvant and PADRE. Most residues are predicted to be accessible by solvent and show high population coverage (99,26%). Molecular docking analysis demonstrated a stable and strong binding affinity with immune receptors. A recombinant plasmid as the template for mRNA production was constructed by inserting the multi-epitope DNA and non-structural polyprotein 1-4 gene of VEEV, which encodes the RNA replication complex to the cloning site of pcDNA3.1(+). Conclusion and implication In silico, design of self-amplifying mRNA could be a potential COVID-19 vaccine candidate since its ability to be amplified in the host cell can efficiently reduce the intake doses.
Collapse
Affiliation(s)
- Brigitta Claudia
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jalan Ganesa 10, Bandung 40132, Indonesia
| | - Husna Nugrahapraja
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jalan Ganesa 10, Bandung 40132, Indonesia
- University Center of Excellence for Nutraceuticals, Bioscience and Biotechnology Research Center, Institut Teknologi Bandung, Jalan Ganesa 10, Bandung 40132, Indonesia
| | - Ernawati Arifin Giri-Rachman
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jalan Ganesa 10, Bandung 40132, Indonesia
- University Center of Excellence for Nutraceuticals, Bioscience and Biotechnology Research Center, Institut Teknologi Bandung, Jalan Ganesa 10, Bandung 40132, Indonesia
| |
Collapse
|
20
|
An W, Li T, Tian X, Fu X, Li C, Wang Z, Wang J, Wang X. Allergies to Allergens from Cats and Dogs: A Review and Update on Sources, Pathogenesis, and Strategies. Int J Mol Sci 2024; 25:10520. [PMID: 39408849 PMCID: PMC11476515 DOI: 10.3390/ijms251910520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Inhalation allergies caused by cats and dogs can lead to a range of discomforting symptoms, such as rhinitis and asthma, in humans. With the increasing popularity of and care provided to these companion animals, the allergens they produce pose a growing threat to susceptible patients' health. Allergens from cats and dogs have emerged as significant risk factors for triggering asthma and allergic rhinitis worldwide; however, there remains a lack of systematic measures aimed at assisting individuals in recognizing and preventing allergies caused by these animals. This review provides comprehensive insights into the classification of cat and dog allergens, along with their pathogenic mechanisms. This study also discusses implementation strategies for prevention and control measures, including physical methods, gene-editing technology, and immunological approaches, as well as potential strategies for enhancing allergen immunotherapy combined with immunoinformatics. Finally, it presents future prospects for the prevention and treatment of human allergies caused by cats and dogs. This review will improve knowledge regarding allergies to cats and dogs while providing insights into potential targets for the development of next-generation treatments.
Collapse
Affiliation(s)
- Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, No. 20, Dongda Street, Beijing 100071, China;
| | - Xinya Tian
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiaoxin Fu
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Chunxiao Li
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
21
|
Banico EC, Sira EMJS, Fajardo LE, Dulay ANG, Odchimar NMO, Simbulan AM, Orosco FL. Advancing one health vaccination: In silico design and evaluation of a multi-epitope subunit vaccine against Nipah virus for cross-species immunization using immunoinformatics and molecular modeling. PLoS One 2024; 19:e0310703. [PMID: 39325755 PMCID: PMC11426463 DOI: 10.1371/journal.pone.0310703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
The resurgence of the Nipah virus (NiV) in 2023 has raised concerns for another potentially severe pandemic, given its history of high mortality from previous outbreaks. Unfortunately, no therapeutics and vaccines have been available for the virus. This study used immunoinformatics and molecular modeling to design and evaluate a multi-epitope subunit vaccine targeting NiV. The designed vaccine construct aims to stimulate immune responses in humans and two other intermediate animal hosts of the virus-swine and equine. Using several epitope prediction tools, ten peptides that induced B-lymphocyte responses, 17 peptides that induced cytotoxic T-lymphocyte (CTL) responses, and 12 peptides that induced helper T-lymphocyte (HTL) responses were mapped from nine NiV protein sequences. However, the CTL and HTL-inducing peptides were reduced to ten and eight, respectively, following molecular docking and dynamics. These screened peptides exhibited stability with 30 common major histocompatibility complex (MHC) receptors found in humans, swine, and equine. All peptides were linked using peptide linkers to form the multi-epitope construct and various adjuvants were tested to enhance its immunogenicity. The vaccine construct with resuscitation-promoting factor E (RpfE) adjuvant was selected as the final design based on its favorable physicochemical properties and superior immune response profile. Molecular docking was used to visualize the interaction of the vaccine to toll-like receptor 4 (TLR4), while molecular dynamics confirmed the structural stability of this interaction. Physicochemical property evaluation and computational simulations showed that the designed vaccine construct exhibited favorable properties and elicited higher antibody titers than the six multi-epitope NiV vaccine designs available in the literature. Further in vivo and in vitro experiments are necessary to validate the immunogenicity conferred by the designed vaccine construct and its epitope components. This study demonstrates the capability of computational methodologies in rational vaccine design and highlights the potential of cross-species vaccination strategies for mitigating potential NiV threats.
Collapse
Affiliation(s)
- Edward Coralde Banico
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Ella Mae Joy Sinco Sira
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Lauren Emily Fajardo
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Albert Neil Gura Dulay
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Nyzar Mabeth Obenio Odchimar
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Alea Maurice Simbulan
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Fredmoore Legaspi Orosco
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
- Department of Science and Technology, S&T Fellows Program, Taguig City, Metro Manila, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila City, Metro Manila, Philippines
| |
Collapse
|
22
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
23
|
Thongchot S, Aksonnam K, Prasopsiri J, Warnnissorn M, Sa-Nguanraksa D, O-Charoenrat P, Thuwajit P, Yenchitsomanus PT, Thuwajit C. Mesothelin- and nucleolin-specific T cells from combined short peptides effectively kill triple-negative breast cancer cells. BMC Med 2024; 22:400. [PMID: 39294656 PMCID: PMC11411782 DOI: 10.1186/s12916-024-03625-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC), known for its aggressiveness and limited treatment options, presents a significant challenge. Adoptive cell transfer, involving the ex vivo generation of antigen-specific T cells from peripheral blood mononuclear cells (PBMCs), emerges as a promising approach. The overexpression of mesothelin (MSLN) and nucleolin (NCL) in TNBC samples underscores their potential as targets for T cell therapy. This study explored the efficacy of multi-peptide pulsing of PBMCs to generate MSLN/NCL-specific T cells targeting MSLN+/NCL+ TNBC cells. METHODS TNBC patient samples were confirmed for both MSLN and NCL expression via immunohistochemistry. Synthesized MSLN and NCL peptides were combined and administered to activate PBMCs from healthy donors. The cancer-killing ability of the resultant T cells was assessed using crystal violet staining, and their subtypes and cytotoxic cytokines were characterized through flow cytometry and cytokine bead array. RESULTS Findings showed that 85.3% (127/149) of TNBC cases were positive for either MSLN or NCL, or both; with single positivity rates for MSLN and NCL of 14.1% and 28.9%, respectively. MSLN and NCL peptides, with high binding affinity for HLA-A*02, were combined and introduced to activated PBMCs from healthy donors. The co-pulsed PBMCs significantly induced TEM and TEMRA CD3+/CD8+ T cells and IFN-γ production, compared to single-peptide pulsed or unpulsed conditions. Notably, MSLN/NCL-specific T cells successfully induced cell death in MSLN+/NCL+ MDA-MB-231 cells, releasing key cytotoxic factors such as perforin, granzymes A and B, Fas ligand, IFN-γ, and granulysin. CONCLUSIONS These findings serve as a proof-of-concept for using multiple immunogenic peptides as a novel therapeutic approach in TNBC patients.
Collapse
Affiliation(s)
- Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Krittaya Aksonnam
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jaturawitt Prasopsiri
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Malee Warnnissorn
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Doonyapat Sa-Nguanraksa
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | | | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
24
|
Fan Y, He Y, Li Y, Yin Z, Shi J, Tian T, Shang K, Shi H, Zhang F, Wen H. Design of a novel EmTSP-3 and EmTIP based multi-epitope vaccine against Echinococcus multilocularis infection. Front Immunol 2024; 15:1425603. [PMID: 39351224 PMCID: PMC11439721 DOI: 10.3389/fimmu.2024.1425603] [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: 04/30/2024] [Accepted: 08/20/2024] [Indexed: 10/04/2024] Open
Abstract
Background Current treatments and prevention strategies for echinococcosis are inadequate. Recent advancements in molecular vaccine development show promise against Echinococcus granulosus; however, Echinococcus multilocularis remains a challenge. A Multi-epitope Vaccine could potentially induce specific B and T lymphocyte responses, thereby offering protection against Echinococcus multilocularis infection. Methods This study aimed to develop a MEV against alveolar echinococcosis. Key epitopes from the Echinococcus multilocularis proteins EmTSP3 and EmTIP were identified using immunoinformatics analyses. These analyses were conducted to assess the MEV feasibility, structural characteristics, molecular docking, molecular dynamics simulations, and immune simulations. The immunogenicity and antigenicity of the vaccine were evaluated through in vitro and in vivo experiments, employing ELISA, Western blotting, FCM, challenge infection experiments, and ELISPOT. Results The effective antigenicity and immunogenicity of MEV were demonstrated through immunoinformatics, as well as in vitro and in vivo experiments. In vitro experiments revealed that MEV increased the secretion of IFN-γ and IL-4 in PBMC and successfully bound to specific antibodies in patient serum. Furthermore, mice immunized with MEV developed a robust immune response, characterized by elevated levels of CD4+ and CD8+ T-cells, increased secretion of IFN-γ and IL-4 by specific Th1 and Th2 cells, and heightened serum antibody levels. Importantly, MEV reduced the weight of cysts by conferring resistance against echinococcosis. These findings suggest that MEV is a promising candidate for the prevention of Echinococcus multilocularis infection. Conclusion A total of 7 CTL, 7 HTL, 5 linear B-cell, and 2 conformational B-cell epitopes were identified. The vaccine has demonstrated effective antigenicity and immunogenicity against AE through molecular docking, immune simulation, molecular dynamics studies, and both in vitro and in vivo experiments. It provides effective protection against Echinococcus multilocularis infection, thereby laying a foundation for further development.
Collapse
Affiliation(s)
- Yichen Fan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yueyue He
- Department of Immunology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Yujiao Li
- Department of Blood Transfusion, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhengwei Yin
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Juan Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tingting Tian
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Kaiyu Shang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Huidong Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fengbo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
25
|
Shi H, Zhu Y, Shang K, Tian T, Yin Z, Shi J, He Y, Ding J, Wang Q, Zhang F. Development of innovative multi-epitope mRNA vaccine against central nervous system tuberculosis using in silico approaches. PLoS One 2024; 19:e0307877. [PMID: 39240891 PMCID: PMC11379207 DOI: 10.1371/journal.pone.0307877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/14/2024] [Indexed: 09/08/2024] Open
Abstract
Tuberculosis(TB) of the Central nervous system (CNS) is a rare and highly destructive disease. The emergence of drug resistance has increased treatment difficulty, leaving the Bacillus Calmette-Guérin (BCG) vaccine as the only licensed preventative immunization available. This study focused on identifying the epitopes of PknD (Rv0931c) and Rv0986 from Mycobacterium tuberculosis(Mtb) strain H37Rv using an in silico method. The goal was to develop a therapeutic mRNA vaccine for preventing CNS TB. The vaccine was designed to be non-allergenic, non-toxic, and highly antigenic. Codon optimization was performed to ensure effective translation in the human host. Additionally, the secondary and tertiary structures of the vaccine were predicted, and molecular docking with TLR-4 was carried out. A molecular dynamics simulation confirmed the stability of the complex. The results indicate that the vaccine structure shows effectiveness. Overall, the constructed vaccine exhibits ideal physicochemical properties, immune response, and stability, laying a theoretical foundation for future laboratory experiments.
Collapse
Affiliation(s)
- Huidong Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yuejie Zhu
- Reproductive Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Kaiyu Shang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tingting Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhengwei Yin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Juan Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yueyue He
- Department of Immunology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Jianbing Ding
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Quan Wang
- Department of Clinical Laboratory, The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fengbo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
26
|
Ahmadi N, Aghasadeghi M, Hamidi-Fard M, Motevalli F, Bahramali G. Reverse Vaccinology and Immunoinformatic Approach for Designing a Bivalent Vaccine Candidate Against Hepatitis A and Hepatitis B Viruses. Mol Biotechnol 2024; 66:2362-2380. [PMID: 37715882 DOI: 10.1007/s12033-023-00867-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 08/21/2023] [Indexed: 09/18/2023]
Abstract
Hepatitis A and B are two crucial viral infections that still dramatically affect public health worldwide. Hepatitis A Virus (HAV) is the main cause of acute hepatitis, whereas Hepatitis B Virus (HBV) leads to the chronic form of the disease, possibly cirrhosis or liver failure. Therefore, vaccination has always been considered the most effective preventive method against pathogens. At this moment, we aimed at the immunoinformatic analysis of HAV-Viral Protein 1 (VP1) as the major capsid protein to come up with the most conserved immunogenic truncated protein to be fused by HBV surface antigen (HBs Ag) to achieve a bivalent vaccine against HAV and HBV using an AAY linker. Various computational approaches were employed to predict highly conserved regions and the most immunogenic B-cell and T-cell epitopes of HAV-VP1 capsid protein in both humans and BALB/c. Moreover, the predicted fusion protein was analyzed regarding primary and secondary structures and also homology validation. Afterward, the three-dimensional structure of vaccine constructs docked with various toll-like receptors (TLR) 2, 4 and 7. According to the bioinformatics tools, the region of 99-259 amino acids of VP1 was selected with high immunogenicity and conserved epitopes. T-cell epitope prediction showed that this region contains 32 antigenic peptides for Human leukocyte antigen (HLA) class I and 20 antigenic peptides in terms of HLA class II which are almost fully conserved in the Iranian population. The vaccine design includes 5 linear and 4 conformational B-cell lymphocyte (BCL) epitopes to induce humoral immune responses. The designed VP1-AAY-HBsAg fusion protein has the potency to be constructed and expressed to achieve a bivalent vaccine candidate, especially in the Iranian population. These findings led us to claim that the designed vaccine candidate provides potential pathways for creating an exploratory vaccine against Hepatitis A and Hepatitis B Viruses with high confidence for the identified strains.
Collapse
Affiliation(s)
- Neda Ahmadi
- Department of Microbiology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohammadreza Aghasadeghi
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, No: 69, Pasteur Ave, Tehran, 13165, Iran
- Viral Vaccine Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mojtaba Hamidi-Fard
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, No: 69, Pasteur Ave, Tehran, 13165, Iran
- Viral Vaccine Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Motevalli
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, No: 69, Pasteur Ave, Tehran, 13165, Iran
| | - Golnaz Bahramali
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, No: 69, Pasteur Ave, Tehran, 13165, Iran.
- Viral Vaccine Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
27
|
Moura DMD, Carvalho AMRS, Brito RCFD, Roatt BM, Lage DP, Martins VT, Cruz LDR, Medeiros FAC, Batista SD, Pinheiro GRG, da Costa Rocha MO, Coelho EAF, Duarte MC, Mendes TADO, Menezes-Souza D. CD4 + and CD8 + T-cell multi-epitope chimeric protein associated with an MPLA adjuvant induce protective efficacy and long-term immunological memory for the immunoprophylaxis of American Tegumentary Leishmaniasis. Vaccine 2024; 42:126178. [PMID: 39096765 DOI: 10.1016/j.vaccine.2024.126178] [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: 03/11/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
American Tegumentary Leishmaniasis (ATL) is a disease of high severity and incidence in Brazil, in addition to being a worldwide concern in public health. Leishmania amazonensis is one of the etiological agents of ATL, and the inefficiency of control measures, associated with the high toxicity of the treatment and the lack of effective immunoprophylactic strategies, makes the development of vaccines indispensable and imminent. In this light, the present study proposes to elaborate a chimeric protein (rChiP), based on the fusion of multiple epitopes of CD4+/CD8+ T cells, identified in the immunoproteome of the parasites L. amazonensis and L. braziliensis. The designed chimeric protein was tested in the L. amazonensis murine model of infection using the following formulations: 25 μg of the rChiP in saline (rChiP group) and 25 μg of the rChiP plus 25 μg of MPLA-PHAD® (rChiP+MPLA group). After completing immunization, CD4+ and CD8+ T cells, stimulated with SLa-Antigen or rChiP, showed an increased production of nitric oxide and intracytoplasmic pro-inflammatory cytokines, in addition to the generation of central and effector memory T cells. rChiP and rChiP+MPLA formulations were able to promote an effective protection against L. amazonensis infection determined by a reduction in the development of skin lesions and lower parasitic burden. Reduction in the development of skin lesions and lower parasitic burden in the vaccinated groups were associated with an increase of nitrite, CD4+/CD8+IFN-γ+TNF-α+ and CD4+/CD8+CD44highCD62Lhigh/low T cells, IgGTotal, IgG2a, and lower rates of IgG1 and CD4+/CD8+IL-10+. This data suggests that proposed formulations could be considered potential tools to prevent ATL.
Collapse
Affiliation(s)
- Dênia Monteiro de Moura
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Ana Maria Ravena Severino Carvalho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Rory Cristiane Fortes de Brito
- Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Universidade Federal de Ouro Preto, Ouro Preto 35400-000, Minas Gerais, Brazil
| | - Bruno Mendes Roatt
- Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Universidade Federal de Ouro Preto, Ouro Preto 35400-000, Minas Gerais, Brazil
| | - Daniela Pagliara Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Vivian Tamietti Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Luiza Dos Reis Cruz
- Laboratório de Química Orgânica Sintética, Instituto de Química, Universidade Estadual de Campinas, Campinas, 13083-970 São Paulo, Brazil
| | - Fernanda Alvarenga Cardoso Medeiros
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Sarah Dutra Batista
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Guilherme Rafael Gomide Pinheiro
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Manoel Otávio da Costa Rocha
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Eduardo Antonio Ferraz Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Mariana Costa Duarte
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | | | - Daniel Menezes-Souza
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; Programa de Pós-Graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil.
| |
Collapse
|
28
|
Arshad NF, Nordin FJ, Foong LC, In LLA, Teo MYM. Engineering receptor-binding domain and heptad repeat domains towards the development of multi-epitopes oral vaccines against SARS-CoV-2 variants. PLoS One 2024; 19:e0306111. [PMID: 39146295 PMCID: PMC11326571 DOI: 10.1371/journal.pone.0306111] [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: 08/29/2023] [Accepted: 06/11/2024] [Indexed: 08/17/2024] Open
Abstract
The inability of existing vaccines to cope with the mutation rate has highlighted the need for effective preventative strategies for COVID-19. Through the secretion of immunoglobulin A, mucosal delivery of vaccines can effectively stimulate mucosal immunity for better protection against SARS-CoV-2 infection. In this study, various immunoinformatic tools were used to design a multi-epitope oral vaccine against SARS-CoV-2 based on its receptor-binding domain (RBD) and heptad repeat (HR) domains. T and B lymphocyte epitopes were initially predicted from the RBD and HR domains of SARS-CoV-2, and potential antigenic, immunogenic, non-allergenic, and non-toxic epitopes were identified. Epitopes that are highly conserved and have no significant similarity to human proteome were selected. The epitopes were joined with appropriate linkers, and an adjuvant was added to enhance the vaccine efficacy. The vaccine 3D structure constructs were docked with toll-like receptor 4 (TLR-4) and TLR1-TLR2, and the binding affinity was calculated. The designed multi-epitope vaccine construct (MEVC) consisted of 33 antigenic T and B lymphocyte epitopes. The results of molecular dockings and free binding energies confirmed that the MEVC effectively binds to TLR molecules, and the complexes were stable. The results suggested that the designed MEVC is a potentially safe and effective oral vaccine against SARS-CoV-2. This in silico study presents a novel approach for creating an oral multi-epitope vaccine against the rapidly evolving SARS-CoV-2 variants. These findings offer valuable insights for developing an effective strategy to combat COVID-19. Further preclinical and clinical studies are required to confirm the efficacy of the MEVC vaccine.
Collapse
Affiliation(s)
- Nur Farhanah Arshad
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Fariza Juliana Nordin
- Department of Biological Sciences and Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Lian Chee Foong
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Michelle Yee Mun Teo
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Rastogi A, Gautam S, Kumar M. Bioinformatic elucidation of conserved epitopes to design a potential vaccine candidate against existing and emerging SARS-CoV-2 variants of concern. Heliyon 2024; 10:e35129. [PMID: 39157328 PMCID: PMC11328099 DOI: 10.1016/j.heliyon.2024.e35129] [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: 09/27/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 poses a significant adverse effects on health and economy globally. Due to mutations in genome, COVID-19 vaccine efficacy decreases. We used immuno-informatics to design a Multi epitope vaccine (MEV) candidate for SARS-CoV-2 variants of concern (VOCs). Hence, we predicted binders/epitopes MHC-I, CD8+, MHC-II, CD4+, and CTLs from spike, membrane and envelope proteins of VOCs. In addition, we assessed the conservation of these binders and epitopes across different VOCs. Subsequently, we designed MEV by combining the predicted CTL and CD4+ epitopes from spike protein, peptide linkers, and an adjuvant. Further, we evaluated the binding of MEV candidate against immune receptors namely HLA class I histocompatibility antigen, HLA class II histocompatibility antigen, and TLR4, achieving binding scores of -1265.3, -1330.7, and -1337.9. Molecular dynamics and normal mode analysis revealed stable docking complexes. Moreover, immune simulation suggested MEV candidate elicits both innate and adaptive immune response. We anticipate that this conserved MEV candidate will provide protection from VOCs and emerging strains.
Collapse
Affiliation(s)
- Amber Rastogi
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh, 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sakshi Gautam
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh, 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh, 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
30
|
Kant R, Khan MS, Chopra M, Saluja D. Artificial intelligence-driven reverse vaccinology for Neisseria gonorrhoeae vaccine: Prioritizing epitope-based candidates. Front Mol Biosci 2024; 11:1442158. [PMID: 39193221 PMCID: PMC11347834 DOI: 10.3389/fmolb.2024.1442158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/04/2024] [Indexed: 08/29/2024] Open
Abstract
Neisseria gonorrhoeae is the causative agent of the sexually transmitted disease gonorrhea. The increasing prevalence of this disease worldwide, the rise of antibiotic-resistant strains, and the difficulties in treatment necessitate the development of a vaccine, highlighting the significance of preventative measures to control and eradicate the infection. Currently, there is no widely available vaccine, partly due to the bacterium's ability to evade natural immunity and the limited research investment in gonorrhea compared to other diseases. To identify distinct vaccine candidates, we chose to focus on the uncharacterized, hypothetical proteins (HPs) as our initial approach. Using the in silico method, we first carried out a comprehensive assessment of hypothetical proteins of Neisseria gonorrhoeae, encompassing assessments of physicochemical properties, cellular localization, secretary pathways, transmembrane regions, antigenicity, toxicity, and prediction of B-cell and T-cell epitopes, among other analyses. Detailed analysis of all HPs resulted in the functional annotation of twenty proteins with a great degree of confidence. Further, using the immuno-informatics approach, the prediction pipeline identified one CD8+ restricted T-cell epitope, seven linear B-cell epitopes, and seven conformational B-cell epitopes as putative epitope-based peptide vaccine candidates which certainly require further validation in laboratory settings. The study accentuates the promise of functional annotation and immuno-informatics in the systematic design of epitope-based peptide vaccines targeting Neisseria gonorrhoeae.
Collapse
Affiliation(s)
- Ravi Kant
- Medical Biotechnology Laboratory, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
- Delhi School of Public Health, Institute of Eminence (IoE), University of Delhi, Delhi, India
| | - Mohd. Shoaib Khan
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Madhu Chopra
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Daman Saluja
- Medical Biotechnology Laboratory, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
- Delhi School of Public Health, Institute of Eminence (IoE), University of Delhi, Delhi, India
| |
Collapse
|
31
|
Shang K, Zhu Y, Tian T, Shi H, Yin Z, He Y, Shi J, Ding J, Zhang F. Development of a novel multi-epitope vaccine for brucellosis prevention. Heliyon 2024; 10:e34721. [PMID: 39148966 PMCID: PMC11325379 DOI: 10.1016/j.heliyon.2024.e34721] [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: 03/13/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Brucellosis, a zoonotic disease caused by Brucella, presents a significant threat to both animal and human health. In animals, the disease can lead to infertility, miscarriage, and high fever, while in humans, symptoms may include recurrent fever, fatigue, sweating, hepatosplenomegaly, and joint and muscle pain following infection. Treatment often involves long-term antibiotic therapy, placing a substantial psychological and financial burden on patients. While vaccination is crucial for prevention, current animal vaccines have drawbacks such as residual virulence, and a safe and effective human vaccine is lacking. Hence, the development of a vaccine for brucellosis is imperative. In this study, we utilized bioinformatics methods to design a multi-epitope vaccine targeting Brucella. Targeting Heme transporter BhuA and polysaccharide export protein, we identified antigenic epitopes, including six cytotoxic T lymphocyte (CTL) dominant epitopes, six helper T lymphocyte (HTL) dominant epitopes, one conformation B cell dominant epitope, and three linear B cell dominant epitopes. By linking these epitopes with appropriate linkers and incorporating a Toll-like receptor (TLR) agonist (human beta-defensin-2) and an auxiliary peptide (Pan HLA-DR epitopes), we constructed the multi-epitope vaccine (MEV). The MEV demonstrated high antigenicity, non-toxicity, non-allergenicity, non-human homology, stability, and solubility. Molecular docking analysis and molecular dynamics simulations confirmed the interaction and stability of the MEV with receptors (MHCI, MHCII, TLR4). Codon optimization and in silico cloning validated the translation efficiency and successful expression of MEV in Escherichia coli. Immunological simulations further demonstrated the efficacy of MEV in inducing robust immune responses. In conclusion, our findings suggest that the engineered MEVs have the potential to stimulate both humoral and cellular immune responses, offering valuable insights for the future development of safe and efficient Brucella vaccines.
Collapse
Affiliation(s)
- Kaiyu Shang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Yuejie Zhu
- Reproductive Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Tingting Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Huidong Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Zhengwei Yin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Yueyue He
- Department of Immunology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Juan Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Jianbing Ding
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Fengbo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| |
Collapse
|
32
|
Pan X, Guo X, Shi J. Design of a novel multiepitope vaccine with CTLA-4 extracellular domain against Mycoplasma pneumoniae: A vaccine-immunoinformatics approach. Vaccine 2024; 42:3883-3898. [PMID: 38777697 DOI: 10.1016/j.vaccine.2024.04.098] [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: 09/17/2023] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Community-acquired pneumonia often stems from the macrolide-resistant strain of Mycoplasma pneumoniae, yet no effective vaccine exists against it. METHODS This study proposes a vaccine-immunoinformatics strategy for Mycoplasma pneumoniae and other pathogenic microbes. Specifically, dominant B and T cell epitopes of the Mycoplasma pneumoniae P30 adhesion protein were identified through immunoinformatics method. The vaccine sequence was then constructed by coupling with CTLA-4 extracellular region, a novel molecular adjuvant for antigen-presenting cells. Subsequently, the vaccine's physicochemical properties, antigenicity, and allergenicity were verified. Molecular dynamics modeling was employed to confirm interaction with TLR-2, TLR-4, B7-1, and B7-2. Finally, the vaccine underwent in silico cloning for expression. RESULTS The vaccine exhibited both antigenicity and non-allergenicity. Molecular dynamics simulation, post-docking with TLR-2, TLR-4, B7-1, and B7-2, demonstrated stable interaction between the vaccine and these molecules. In silico cloning confirmed effective expression of the vaccine gene in insect baculovirus vectors. CONCLUSION This vaccine-immunoinformatics approach holds promise for the development of vaccines against Mycoplasma pneumoniae and other pathogenic non-viral and non-bacterial microbes.
Collapse
Affiliation(s)
- Xiaohong Pan
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| | - Xiaomei Guo
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China; Kunming Medical University, Kunming, Yunnan, China
| | - Jiandong Shi
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China; National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan China.
| |
Collapse
|
33
|
Ananya, Panchariya DC, Karthic A, Singh SP, Mani A, Chawade A, Kushwaha S. Vaccine design and development: Exploring the interface with computational biology and AI. Int Rev Immunol 2024; 43:361-380. [PMID: 38982912 DOI: 10.1080/08830185.2024.2374546] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/29/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
Computational biology involves applying computer science and informatics techniques in biology to understand complex biological data. It allows us to collect, connect, and analyze biological data at a large scale and build predictive models. In the twenty first century, computational resources along with Artificial Intelligence (AI) have been widely used in various fields of biological sciences such as biochemistry, structural biology, immunology, microbiology, and genomics to handle massive data for decision-making, including in applications such as drug design and vaccine development, one of the major areas of focus for human and animal welfare. The knowledge of available computational resources and AI-enabled tools in vaccine design and development can improve our ability to conduct cutting-edge research. Therefore, this review article aims to summarize important computational resources and AI-based tools. Further, the article discusses the various applications and limitations of AI tools in vaccine development.
Collapse
Affiliation(s)
- Ananya
- National Institute of Animal Biotechnology, Hyderabad, India
| | | | | | | | - Ashutosh Mani
- Motilal Nehru National Institute of Technology, Prayagraj, India
| | - Aakash Chawade
- Swedish University of Agricultural Sciences, Alnarp, Sweden
| | | |
Collapse
|
34
|
Guasp P, Reiche C, Sethna Z, Balachandran VP. RNA vaccines for cancer: Principles to practice. Cancer Cell 2024; 42:1163-1184. [PMID: 38848720 DOI: 10.1016/j.ccell.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024]
Abstract
Vaccines are the most impactful medicines to improve health. Though potent against pathogens, vaccines for cancer remain an unfulfilled promise. However, recent advances in RNA technology coupled with scientific and clinical breakthroughs have spurred rapid discovery and potent delivery of tumor antigens at speed and scale, transforming cancer vaccines into a tantalizing prospect. Yet, despite being at a pivotal juncture, with several randomized clinical trials maturing in upcoming years, several critical questions remain: which antigens, tumors, platforms, and hosts can trigger potent immunity with clinical impact? Here, we address these questions with a principled framework of cancer vaccination from antigen detection to delivery. With this framework, we outline features of emergent RNA technology that enable rapid, robust, real-time vaccination with somatic mutation-derived neoantigens-an emerging "ideal" antigen class-and highlight latent features that have sparked the belief that RNA could realize the enduring vision for vaccines against cancer.
Collapse
Affiliation(s)
- Pablo Guasp
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Reiche
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zachary Sethna
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
35
|
Yan Z, Kim K, Kim H, Ha B, Gambiez A, Bennett J, de Almeida Mendes M, Trevizani R, Mahita J, Richardson E, Marrama D, Blazeska N, Koşaloğlu-Yalçın Z, Nielsen M, Sette A, Peters B, Greenbaum J. Next-generation IEDB tools: a platform for epitope prediction and analysis. Nucleic Acids Res 2024; 52:W526-W532. [PMID: 38783079 PMCID: PMC11223806 DOI: 10.1093/nar/gkae407] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
The Next-Generation (NG) IEDB Tools website (https://nextgen-tools.iedb.org) provides users with a redesigned interface to many of the algorithms for epitope prediction and analysis that were originally released on the legacy IEDB Tools website. The initial release focuses on consolidation of all tools related to HLA class I epitopes (MHC binding, elution, immunogenicity, and processing), making all of these predictions accessible from a single application and allowing for their simultaneous execution with minimal user inputs. Additionally, the PEPMatch tool for identifying highly similar epitopes in a set of curated proteomes, as well as a tool for epitope clustering, are available on the site. The NG Tools site allows users to build data pipelines by sending the output of one tool as input for the next. Over the next several years, all pre-existing IEDB Tools, and any newly developed tools, will be integrated into this new site. Here we describe the philosophy behind the redesign and demonstrate the utility and productivity enhancements that are enabled by the new interface.
Collapse
Affiliation(s)
- Zhen Yan
- Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kevin Kim
- Information Technology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Haeuk Kim
- Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Brendan Ha
- Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Anaïs Gambiez
- Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Jason Bennett
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | - Raphael Trevizani
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Fiocruz Ceará, Fundação Oswaldo Cruz, Rua São José s/n, Precabura, Eusébio/CE, Brazil
| | - Jarjapu Mahita
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Eve Richardson
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Daniel Marrama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Nina Blazeska
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, DK-2800 Kgs, Lyngby, Denmark
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, B1650 Buenos Aires, Argentina
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jason A Greenbaum
- Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| |
Collapse
|
36
|
Richardson E, Trevizani R, Greenbaum JA, Carter H, Nielsen M, Peters B. The receiver operating characteristic curve accurately assesses imbalanced datasets. PATTERNS (NEW YORK, N.Y.) 2024; 5:100994. [PMID: 39005487 PMCID: PMC11240176 DOI: 10.1016/j.patter.2024.100994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/05/2024] [Accepted: 05/03/2024] [Indexed: 07/16/2024]
Abstract
Many problems in biology require looking for a "needle in a haystack," corresponding to a binary classification where there are a few positives within a much larger set of negatives, which is referred to as a class imbalance. The receiver operating characteristic (ROC) curve and the associated area under the curve (AUC) have been reported as ill-suited to evaluate prediction performance on imbalanced problems where there is more interest in performance on the positive minority class, while the precision-recall (PR) curve is preferable. We show via simulation and a real case study that this is a misinterpretation of the difference between the ROC and PR spaces, showing that the ROC curve is robust to class imbalance, while the PR curve is highly sensitive to class imbalance. Furthermore, we show that class imbalance cannot be easily disentangled from classifier performance measured via PR-AUC.
Collapse
Affiliation(s)
- Eve Richardson
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Raphael Trevizani
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Fiocruz Ceará, Fundação Oswaldo Cruz, Rua São José s/n, Precabura, Eusébio/CE, Brazil
| | - Jason A Greenbaum
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Hannah Carter
- Department of Medicine, University of California, La Jolla, CA, USA
| | - Morten Nielsen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Lyngby, Denmark
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| |
Collapse
|
37
|
Yao P, Gao M, Hu W, Wang J, Wang Y, Wang Q, Ji J. Proteogenomic analysis identifies neoantigens and bacterial peptides as immunotherapy targets in colorectal cancer. Pharmacol Res 2024; 204:107209. [PMID: 38740147 DOI: 10.1016/j.phrs.2024.107209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Considerable progress has recently been made in cancer immunotherapy, including immune checkpoint blockade, cancer vaccine, and adoptive T cell methods. The lack of effective targets is a major cause of the low immunotherapy response rate in colorectal cancer (CRC). Here, we used a proteogenomic strategy comprising immunopeptidomics, whole exome sequencing, and 16 S ribosomal DNA sequencing analyses of 8 patients with CRC to identify neoantigens and bacterial peptides that can serve as antitumor targets. This study directly identified several personalized neoantigens and bacterial immunopeptides. Immunoassays showed that all neoantigens and 5 of 8 bacterial immunopeptides could be recognized by autologous T cells. Additionally, T cell receptor (TCR) αβ sequencing revealed the TCR repertoire of epitope-reactive CD8+ T cells. Functional studies showed that T cell receptor-T (TCR-T) could be activated by epitope pulsed lymphoblastoid cells. Overall, this study comprehensively profiled the CRC immunopeptidome, revealing several neoantigens and bacterial peptides with potential to serve as immunotherapy targets in CRC.
Collapse
Affiliation(s)
- Pengju Yao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Mingjie Gao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Weiyi Hu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Jiahao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Yuhao Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
38
|
Amarajeewa AWP, Özcan A, Mukhtiar A, Ren X, Wang Q, Ozbek P, Garstka MA, Serçinoğlu O. Polymorphism in F pocket affects peptide selection and stability of type 1 diabetes-associated HLA-B39 allotypes. Eur J Immunol 2024; 54:e2350683. [PMID: 38549458 DOI: 10.1002/eji.202350683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 10/30/2024]
Abstract
HLA-B*39:06, HLA-B*39:01, and HLA-B*38:01 are closely related HLA allotypes differentially associated with type 1 diabetes (T1D) risk and progression. B*39:06 is highly predisposing, while B*39:01 and B*38:01 are weakly predisposing and protective allotypes, respectively. Here, we aimed to decipher molecular mechanisms underlying the differential association of these allotypes with T1D pathogenesis. We addressed peptide binding and conformational stability of HLA-B allotypes using computational and experimental approaches. Computationally, we found that B*39:06 and B*39:01 allotypes had more rigid peptide-binding grooves and were more promiscuous in binding peptides than B*38:01. Peptidomes of B*39:06 and B*39:01 contained fewer strong binders and were of lower affinity than that of B*38:01. Experimentally, we demonstrated that B*39:06 and B*39:01 had a higher capacity to bind peptides and exit to the cell surface but lower surface levels and were degraded faster than B*38:01. In summary, we propose that promiscuous B*39:06 and B*39:01 may bind suboptimal peptides and transport them the cell surface, where such unstable complexes may contribute to the pathogenesis of T1D.
Collapse
Affiliation(s)
- A W Peshala Amarajeewa
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Core Research Laboratory, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aslihan Özcan
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Türkiye
| | - Alveena Mukhtiar
- Core Research Laboratory, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xu Ren
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Core Research Laboratory, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qianyu Wang
- Core Research Laboratory, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pemra Ozbek
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Türkiye
| | - Malgorzata A Garstka
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Core Research Laboratory, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Endocrinology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Tumor and Immunology, Precision Medical Institute, Western China Science and Technology Innovation Port, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Onur Serçinoğlu
- Department of Bioengineering, Faculty of Engineering, Gebze Technical University, Gebze, Türkiye
| |
Collapse
|
39
|
Samri A, Bandeira AC, Gois LL, Silva CGR, Rousseau A, Corneau A, Tarantino N, Maucourant C, Queiroz GAN, Vieillard V, Yssel H, Campos GS, Sardi S, Autran B, Rios Grassi MF. Comprehensive analysis of early T cell responses to acute Zika Virus infection during the first epidemic in Bahia, Brazil. PLoS One 2024; 19:e0302684. [PMID: 38722858 PMCID: PMC11081376 DOI: 10.1371/journal.pone.0302684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/05/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND In most cases, Zika virus (ZIKV) causes a self-limited acute illness in adults, characterized by mild clinical symptoms that resolve within a few days. Immune responses, both innate and adaptive, play a central role in controlling and eliminating virus-infected cells during the early stages of infection. AIM To test the hypothesis that circulating T cells exhibit phenotypic and functional activation characteristics during the viremic phase of ZIKV infection. METHODS A comprehensive analysis using mass cytometry was performed on peripheral blood mononuclear cells obtained from patients with acute ZIKV infection (as confirmed by RT-PCR) and compared with that from healthy donors (HD). The frequency of IFN-γ-producing T cells in response to peptide pools covering immunogenic regions of structural and nonstructural ZIKV proteins was quantified using an ELISpot assay. RESULTS Circulating CD4+ and CD8+ T lymphocytes from ZIKV-infected patients expressed higher levels of IFN-γ and pSTAT-5, as well as cell surface markers associated with proliferation (Ki-67), activation ((HLA-DR, CD38) or exhaustion (PD1 and CTLA-4), compared to those from HD. Activation of CD4+ and CD8+ memory T cell subsets, including Transitional Memory T Cells (TTM), Effector Memory T cells (TEM), and Effector Memory T cells Re-expressing CD45RA (TEMRA), was prominent among CD4+ T cell subset of ZIKV-infected patients and was associated with increased levels of IFN-γ, pSTAT-5, Ki-67, CTLA-4, and PD1, as compared to HD. Additionally, approximately 30% of ZIKV-infected patients exhibited a T cell response primarily directed against the ZIKV NS5 protein. CONCLUSION Circulating T lymphocytes spontaneously produce IFN-γ and express elevated levels of pSTAT-5 during the early phase of ZIKV infection whereas recognition of ZIKV antigen results in the generation of virus-specific IFN-γ-producing T cells.
Collapse
Affiliation(s)
- Assia Samri
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Antonio Carlos Bandeira
- Secretaria de Saúde da Bahia, Salvador, Bahia, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Luana Leandro Gois
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
- Departamento de Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Alice Rousseau
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Aurelien Corneau
- Faculté de Médecine Pierre et Marie Curie, Plateforme de Cytométrie (CyPS), UMS30–LUMIC, Paris, France
| | - Nadine Tarantino
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Christopher Maucourant
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Gabriel Andrade Nonato Queiroz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| | - Vincent Vieillard
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Hans Yssel
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Gubio Soares Campos
- Departamento de Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Silvia Sardi
- Departamento de Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Brigitte Autran
- Sorbonne-Université, Inserm 1135, CNRS ERL8255, Centre d’immunologie et des Maladies Infectieuses, Cimi, Paris, France
| | - Maria Fernanda Rios Grassi
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| |
Collapse
|
40
|
Sira EMJS, Banico EC, Odchimar NMO, Fajardo LE, Fremista FF, Refuerzo HAB, Dictado APA, Orosco FL. Immunoinformatics approach for designing a multiepitope subunit vaccine against porcine epidemic diarrhea virus genotype IIA spike protein. Open Vet J 2024; 14:1224-1242. [PMID: 38938443 PMCID: PMC11199741 DOI: 10.5455/ovj.2024.v14.i5.18] [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: 03/20/2024] [Accepted: 04/26/2024] [Indexed: 06/29/2024] Open
Abstract
Background Porcine epidemic diarrhea (PED), caused by the porcine epidemic diarrhea virus (PEDV), is associated with high mortality and morbidity rates, especially in neonatal pigs. This has resulted in significant economic losses for the pig industry. PEDV genotype II-based vaccines were found to confer better immunity against both heterologous and homologous challenges; specifically, spike (S) proteins, which are known to play a significant role during infection, are ideal for vaccine development. Aim This study aims to design a multi-epitope subunit vaccine targeting the S protein of the PEDV GIIa strain using an immunoinformatics approach. Methods Various bioinformatics tools were used to predict HTL, CTL, and B-cell epitopes. The epitopes were connected using appropriate linkers and conjugated with the CTB adjuvant and M-ligand. The final multiepitope vaccine construct (fMEVc) was then docked to toll-like receptor 4 (TLR4). The stability of the fMEVc-TLR4 complex was then simulated using GROMACS. C-immsim was then used to predict the in vitro immune response of the fMEVc. Results Six epitopes were predicted to induce antibody production, ten epitopes were predicted to induce CTL responses, and four epitopes were predicted to induce HTL responses. The assembled epitopes conjugated with the CTB adjuvant and M-ligand, fMEVc, is antigenic, non-allergenic, stable, and soluble. The construct showed a favorable binding affinity for TLR4, and the protein complex was shown to be stable through molecular dynamics simulations. A robust immune response was induced after immunization, as demonstrated through immune stimulation. Conclusion In conclusion, the multi-epitope subunit vaccine construct for PEDV designed in this study exhibits promising antigenicity, stability, and immunogenicity, eliciting robust immune responses and suggesting its potential as a candidate for further vaccine development.
Collapse
Affiliation(s)
- Ella Mae Joy S. Sira
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Edward C. Banico
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Nyzar Mabeth O. Odchimar
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Lauren Emily Fajardo
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Ferdinand F. Fremista
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Philippines
| | | | - Ana Patrisha A. Dictado
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Philippines
| | - Fredmoore L. Orosco
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Philippines
- S&T Fellows Program, Department of Science and Technology, Taguig City, Philippines
| |
Collapse
|
41
|
Almanaa TN, Mubarak A, Sajjad M, Ullah A, Hassan M, Waheed Y, Irfan M, Khan S, Ahmad S. Design and validation of a novel multi-epitopes vaccine against hantavirus. J Biomol Struct Dyn 2024; 42:4185-4195. [PMID: 37261466 DOI: 10.1080/07391102.2023.2219324] [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: 03/21/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Hantavirus is a member of the order Bunyavirales and an emerging global pathogen. Hantavirus infections have affected millions of people globally based on available epidemiological data and research studies. Hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS) are the two main human diseases associated with hantavirus infections. Hence, efforts are required to develop a potent vaccine against the pathogen. The only vaccine that is in use for hantavirus is an inactivated virus vaccine, "Hantavax", but it failed to produce neutralizing antibodies. Vaccine development is of much importance in dealing with the surge of hantavirus globally. In this study, hantavirus five proteins (N protein, G1 and G2, L protein, and non-structural proteins) were used in NetCTL 1.2 program to predict T-cell epitopes. To predict major histocompatibility complex (MHC) binding alleles, an immune epitope database (IEDB) was used. All predicted epitopes were then investigated for different immunoinformatics analyses such as antigenicity and toxicity analyses. The good water-soluble, non-toxic, probable antigenic, and DRB*0101 binder was selected. A multi-epitopes-based vaccine designing was then done where linkers were used to connect the shortlisted epitopes. In addition, an adjuvant molecule was supplementary to the multi-epitopes peptide to improve the vaccine's immunogenic potential. The final vaccine construct's three-dimensional structure was modeled by ab initio method. The vaccine molecule was then evaluated for its binding potential with TLR-3 immune receptor, which is key for its recognition and processing by the host immune system. Docking studies were performed using HADDOCK software. The best-docked complex was selected and visualized for intermolecular binding and interactions using UCSF Chimera 1.16 software. The findings revealed that the designed vaccine might be a potential vaccine against hantavirus and can be used in experimental animal model testings.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ayman Mubarak
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Sajjad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Asad Ullah
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Muhammad Hassan
- Department of Pharmacy, Bacha Khan University, Charsadda, Pakistan
| | - Yasir Waheed
- Office of Research, Innovation and Commercialization, Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Muhammad Irfan
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Saifullah Khan
- Institute of Biotechnology and Microbiology, Bacha Khan University, Charsadda, Pakistan
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
- Department of Computer Science, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
42
|
Quiros-Fernandez I, Libório-Ramos S, Leifert L, Schönfelder B, Vlodavsky I, Cid-Arregui A. Dual T cell receptor/chimeric antigen receptor engineered NK-92 cells targeting the HPV16 E6 oncoprotein and the tumor-associated antigen L1CAM exhibit enhanced cytotoxicity and specificity against tumor cells. J Med Virol 2024; 96:e29630. [PMID: 38659368 DOI: 10.1002/jmv.29630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/08/2024] [Accepted: 04/13/2024] [Indexed: 04/26/2024]
Abstract
The human papillomavirus type 16 (HPV16) causes a large fraction of genital and oropharyngeal carcinomas. To maintain the transformed state, the tumor cells must continuously synthesize the E6 and E7 viral oncoproteins, which makes them tumor-specific antigens. Indeed, specific T cell responses against them have been well documented and CD8+ T cells engineered to express T cell receptors (TCRs) that recognize epitopes of E6 or E7 have been tested in clinical studies with promising results, yet with limited clinical success. Using CD8+ T cells from peripheral blood of healthy donors, we have identified two novel TCRs reactive to an unexplored E618-26 epitope. These TCRs showed limited standalone cytotoxicity against E618-26-HLA-A*02:01-presenting tumor cells. However, a single-signaling domain chimeric antigen receptor (ssdCAR) targeting L1CAM, a cell adhesion protein frequently overexpressed in HPV16-induced cancer, prompted a synergistic effect that significantly enhanced the cytotoxic capacity of NK-92/CD3/CD8 cells armored with both TCR and ssdCAR when both receptors simultaneously engaged their respective targets, as shown by live microscopy of 2-D and 3-D co-cultures. Thus, virus-specific TCRs from the CD8+ T cell repertoire of healthy donors can be combined with a suitable ssdCAR to enhance the cytotoxic capacity of the effector cells and, indirectly, their specificity.
Collapse
MESH Headings
- Humans
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Repressor Proteins/immunology
- Repressor Proteins/genetics
- CD8-Positive T-Lymphocytes/immunology
- Killer Cells, Natural/immunology
- Human papillomavirus 16/immunology
- Human papillomavirus 16/genetics
- Cytotoxicity, Immunologic
- Cell Line, Tumor
Collapse
Affiliation(s)
- Isaac Quiros-Fernandez
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Research Center on Tropical Diseases (CIET)/Research Center on Surgery and Cancer (CICICA), Faculty of Microbiology, Universidad de Costa Rica, San Jose, Costa Rica
| | - Sofia Libório-Ramos
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Leifert
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bruno Schönfelder
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
43
|
Truex N, Mohapatra S, Melo M, Rodriguez J, Li N, Abraham W, Sementa D, Touti F, Keskin DB, Wu CJ, Irvine DJ, Gómez-Bombarelli R, Pentelute BL. Design of Cytotoxic T Cell Epitopes by Machine Learning of Human Degrons. ACS CENTRAL SCIENCE 2024; 10:793-802. [PMID: 38680558 PMCID: PMC11046456 DOI: 10.1021/acscentsci.3c01544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 05/01/2024]
Abstract
Antigen processing is critical for therapeutic vaccines to generate epitopes for priming cytotoxic T cell responses against cancer and pathogens, but insufficient processing often limits the quantity of epitopes released. We address this challenge using machine learning to ascribe a proteasomal degradation score to epitope sequences. Epitopes with varying scores were translocated into cells using nontoxic anthrax proteins. Epitopes with a low score show pronounced immunogenicity due to antigen processing, but epitopes with a high score show limited immunogenicity. This work sheds light on the sequence-activity relationships between proteasomal degradation and epitope immunogenicity. We anticipate that future efforts to incorporate proteasomal degradation signals into vaccine designs will lead to enhanced cytotoxic T cell priming by these vaccines in clinical settings.
Collapse
Affiliation(s)
- Nicholas
L. Truex
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
- Department
of Chemistry and Biochemistry, University
of South Carolina, Columbia, South Carolina 29208, United States
| | - Somesh Mohapatra
- Department
of Materials Science and Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Machine
Intelligence and Manufacturing Operations Group, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Mariane Melo
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Ragon Institute
of Massachusetts General Hospital, Massachusetts
Institute of Technology, and Harvard University, Cambridge, Massachusetts 02139, United States
| | - Jacob Rodriguez
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Na Li
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Wuhbet Abraham
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Deborah Sementa
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Faycal Touti
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Derin B. Keskin
- Department
of Medical Oncology, Dana-Farber Cancer
Institute, Boston, Massachusetts 02215, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Translational
Immunogenomics Laboratory (TIGL), Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
- Department
of Computer Science, Metropolitan College, Boston University, Boston, Massachusetts 02215, United States
- Section
for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby DK-2800, Denmark
| | - Catherine J. Wu
- Department
of Medical Oncology, Dana-Farber Cancer
Institute, Boston, Massachusetts 02215, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States
| | - Darrell J. Irvine
- Department
of Materials Science and Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Ragon Institute
of Massachusetts General Hospital, Massachusetts
Institute of Technology, and Harvard University, Cambridge, Massachusetts 02139, United States
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| | - Rafael Gómez-Bombarelli
- Department
of Materials Science and Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley L. Pentelute
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Center
for Environmental Health Sciences, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
44
|
Shao W, Yao Y, Yang L, Li X, Ge T, Zheng Y, Zhu Q, Ge S, Gu X, Jia R, Song X, Zhuang A. Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy. Exp Hematol Oncol 2024; 13:37. [PMID: 38570883 PMCID: PMC10988985 DOI: 10.1186/s40164-024-00504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Weihuan Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Ludi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiaoran Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Qiuyi Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
45
|
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
|
46
|
Ren X, Amarajeewa AWP, Jayasinghe MDT, Garstka MA. Differences in F pocket impact on HLA I genetic associations with autoimmune diabetes. Front Immunol 2024; 15:1342335. [PMID: 38596688 PMCID: PMC11003304 DOI: 10.3389/fimmu.2024.1342335] [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: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Human leukocyte antigen (HLA) I molecules present antigenic peptides to activate CD8+ T cells. Type 1 Diabetes (T1D) is an auto-immune disease caused by aberrant activation of the CD8+ T cells that destroy insulin-producing pancreatic β cells. Some HLA I alleles were shown to increase the risk of T1D (T1D-predisposing alleles), while some reduce this risk (T1D-protective alleles). Methods Here, we compared the T1D-predisposing and T1D-protective allotypes concerning peptide binding, maturation, localization and surface expression and correlated it with their sequences and energetic profiles using experimental and computational methods. Results T1D-predisposing allotypes had more peptide-bound forms and higher plasma membrane levels than T1D-protective allotypes. This was related to the fact that position 116 within the F pocket was more conserved and made more optimal contacts with the neighboring residues in T1D-predisposing allotypes than in protective allotypes. Conclusion Our work uncovers that specific polymorphisms in HLA I molecules potentially influence their susceptibility to T1D.
Collapse
Affiliation(s)
- Xu Ren
- Department of Urology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Core Research Laboratory, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of Endocrinology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - A. W. Peshala Amarajeewa
- Core Research Laboratory, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | | | - Malgorzata A. Garstka
- Department of Urology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Core Research Laboratory, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of Endocrinology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of Tumor and Immunology, Precision Medical Institute, Western China Science and Technology Innovation Port, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
47
|
Lu Q, Zhou W, Fan L, Ding T, Wang W, Zhang X. Tumor neoantigens derived from RNA editing events show significant clinical relevance in melanoma patients treated with immunotherapy. Anticancer Drugs 2024; 35:305-314. [PMID: 38170793 DOI: 10.1097/cad.0000000000001565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
This study aimed to investigate the clinical significance of RNA editing (RE) and RNA editing derived (RED-) neoantigens in melanoma patients treated with immunotherapy. Vardict and VEP were used to identify the somatic mutations. RE events were identified by Reditools2 and filtered by the custom pipeline. miRTar2GO was implemented to predict the RE whether located in miRNA targets within the 3' UTR region. NetMHCpan and NetCTLpan were used to identify and characterize RED-neoantigens. In total, 7116 RE events were identified, most of which were A-to-I events. Using our custom pipeline, 631 RED-neoantigens were identified that show a significantly greater peptide-MHC affinity, and facilitate epitope processing and presentation than wild-type peptides. The OS of the patients with high RED-neoantigens burden was significantly longer ( P = 0.035), and a significantly higher RED-neoantigens burden was observed in responders ( P = 0.048). The area under the curve of the RED-neoantigen was 0.831 of OS. Then, we validated the reliability of RED-neoantigens in predicting the prognosis in an independent cohort and found that patients with high RED-neoantigens exhibited a longer OS ( P = 0.008). To our knowledge, this is the first study to systematically assess the clinical relevance of RED-neoantigens in melanoma patients treated with immunotherapy.
Collapse
Affiliation(s)
- Qicheng Lu
- Department of Gastrointestinal Surgery, Changzhou First People's Hospital, Changzhou, Jiangsu
| | - Wenhao Zhou
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd., Shenzhen, Guangdong
| | - Ligang Fan
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou
| | - Tian Ding
- Department of Clinical Medicine, Medical School, Nantong University
| | - Wei Wang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd., Shenzhen, Guangdong
| | - Xiaodong Zhang
- Department of Medical Oncology, Tumor Hospital Affiliated To Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
48
|
Nie J, Zhou Y, Ding F, Liu X, Yao X, Xu L, Chang Y, Li Z, Wang Q, Zhan L, Zhu L, Xie K, Li C, Shi Y, Zhao Q, Shan Y. Self-adjuvant multiepitope nanovaccine based on ferritin induced long-lasting and effective mucosal immunity against H3N2 and H1N1 viruses in mice. Int J Biol Macromol 2024; 259:129259. [PMID: 38191112 DOI: 10.1016/j.ijbiomac.2024.129259] [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: 12/11/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
The influenza A virus (IAV) is a ubiquitous and continuously evolving respiratory pathogen. The intranasal vaccination mimicking natural infections is an attractive strategy for controlling IAVs. Multiepitope vaccines accurately targeting multiple conserved domains have the potential to broaden the protective scope of current seasonal influenza vaccines and reduce the risk of generating escape mutants. Here, multiple linear epitopes from the matrix protein 2 ectodomain (M2e) and the hemagglutinin stem domain (HA2) are fused with the Helicobacter pylori ferritin, a self-assembled nanocarrier and mucosal adjuvant, to develop a multiepitope nanovaccine. Through intranasal delivery, the prokaryotically expressed multiepitope nanovaccine elicits long-lasting mucosal immunity, broad humoral immunity, and robust cellular immunity without any adjuvants, and confers complete protection against H3N2 and H1N1 subtypes of IAV in mice. Importantly, this intranasal multiepitope nanovaccine triggers memory B-cell responses, resulting in secretory immunoglobulin A (sIgA) and serum immunoglobulin G (IgG) levels persisting for more than five months post-immunization. Therefore, this intranasal ferritin-based multiepitope nanovaccine represents a promising approach to combating respiratory pathogens.
Collapse
Affiliation(s)
- Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Yongfei Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Fan Ding
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xiaoxi Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Lipeng Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yaotian Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Zeyu Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Li Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Lvzhou Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Kunpeng Xie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Chenxi Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China; Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| |
Collapse
|
49
|
Wang Y, Wu A, Xu Z, Zhang H, Li H, Fu S, Liu Y, Cui L, Miao Y, Wang Y, Zhumanov K, Xu Y, Sheng J, Yi J, Chen C. A multi-epitope subunit vaccine based on CU/ZN-SOD, OMP31 and BP26 against Brucella melitensis infection in BALB/C mice. Int Immunopharmacol 2024; 127:111351. [PMID: 38113688 DOI: 10.1016/j.intimp.2023.111351] [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: 10/09/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Brucellosis, a zoonosis caused by Brucella, is highly detrimental to both humans and animals. Most existing vaccines are live attenuated vaccines with safety flaws for people and animals. Therefore, it is advantageous to design a multi-epitope subunit vaccine (MEV) to prevent Brucella infection. To this end, we applied a reverse vaccinology approach. Six cytotoxic T cell (CTL) epitopes, seven T helper cell (HTL) epitopes, and four linear B cell epitopes from CU/ZN-SOD, Omp31, and BP26 were obtained. We linked the CTL, HTL, B-cell epitopes, the appropriate CTB molecular adjuvant, and the universal T helper lymphocyte epitope, PADRE, with linkers AAY, GPPGG, and KK, respectively. This yielded a 412-amino acid MEV construct, which we named MEVcob. The immunogenicity, stability, safety, and feasibility of the construct were evaluated by bioinformatics tools (including the AlphaFold2 prediction tool, the AlphaFold2 tool, NetMHC-I pan 4.0 server, IEDB MHC-I server, ABCpred service, and C-ImmSim server); the physicochemical properties, secondary and tertiary structures, and binding ability of MEVocb to toll-like receptor 4 (TLR4) was analyzed. Then, codon adaptation and computer cloning studies were performed. MEVocb is highly immunogenic in immunostimulation experiments, The proteins translated by these sequences were relatively stable, exhibiting a high antigenic index. Furthermore, mouse experiments confirmed that the MEVocb construct could raise IFN-γ, IgG, IgG2a, IgG1, IL-2, TNF-α levels in mice, indicating that induced a specific humoral and cellular immune response in BALB/c mice. This vaccine induced a statistically significant level of protection in BALB/c mice when challenged with Brucella melitensis 043 in Xinjiang. Briefly, we utilized immunoinformatic tools to design a novel multi-epitope subunit candidate vaccine against Brucella. This vaccine aims to induce host immune responses and confer specific protective effects. The study results offer a theoretical foundation for the development of a novel Brucella subunit vaccine.
Collapse
Affiliation(s)
- Yueli Wang
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Aodi Wu
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Zhenyu Xu
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Huan Zhang
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Honghuan Li
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Shuangshuang Fu
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Yajing Liu
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Lijin Cui
- Fujian Biotechnology Co., LTD., 353000 Nanping, Fujian, China
| | - Yuhe Miao
- Fujian Biotechnology Co., LTD., 353000 Nanping, Fujian, China
| | - Yong Wang
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China
| | - Kaiat Zhumanov
- Kazakh National Agrarian University, The Republic of Kazakhstan
| | - Yimei Xu
- Xinjiang Center for Disease Control and Prevention, 830000 Urumqi, Xinjiang, China
| | - Jinliang Sheng
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China.
| | - Jihai Yi
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China.
| | - Chuangfu Chen
- School of Animal Science and Technology, Shihezi University, 832000 Shihezi City, Xinjiang, China.
| |
Collapse
|
50
|
Simbulan AM, Banico EC, Sira EMJS, Odchimar NMO, Orosco FL. Immunoinformatics-guided approach for designing a pan-proteome multi-epitope subunit vaccine against African swine fever virus. Sci Rep 2024; 14:1354. [PMID: 38228670 DOI: 10.1038/s41598-023-51005-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024] Open
Abstract
Despite being identified over a hundred years ago, there is still no commercially available vaccine for the highly contagious and deadly African swine fever virus (ASFV). This study used immunoinformatics for the rapid and inexpensive designing of a safe and effective multi-epitope subunit vaccine for ASFV. A total of 18,858 proteins from 100 well-annotated ASFV proteomes were screened using various computational tools to identify potential epitopes, or peptides capable of triggering an immune response in swine. Proteins from genotypes I and II were prioritized for their involvement in the recent global ASFV outbreaks. The screened epitopes exhibited promising qualities that positioned them as effective components of the ASFV vaccine. They demonstrated antigenicity, immunogenicity, and cytokine-inducing properties indicating their ability to induce potent immune responses. They have strong binding affinities to multiple swine allele receptors suggesting a high likelihood of yielding more amplified responses. Moreover, they were non-allergenic and non-toxic, a crucial prerequisite for ensuring safety and minimizing any potential adverse effects when the vaccine is processed within the host. Integrated with an immunogenic 50S ribosomal protein adjuvant and linkers, the epitopes formed a 364-amino acid multi-epitope subunit vaccine. The ASFV vaccine construct exhibited notable immunogenicity in immune simulation and molecular docking analyses, and stable profiles in secondary and tertiary structure assessments. Moreover, this study designed an optimized codon for efficient translation of the ASFV vaccine construct into the Escherichia coli K-12 expression system using the pET28a(+) vector. Overall, both sequence and structural evaluations suggested the potential of the ASFV vaccine construct as a candidate for controlling and eradicating outbreaks caused by the pathogen.
Collapse
Affiliation(s)
- Alea Maurice Simbulan
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Edward C Banico
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Ella Mae Joy S Sira
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Nyzar Mabeth O Odchimar
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines
| | - Fredmoore L Orosco
- Department of Science and Technology, Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Bicutan, 1634, Taguig, Metro Manila, Philippines.
- Department of Science and Technology, S&T Fellows Program, Bicutan, 1634, Taguig, Metro Manila, Philippines.
- Department of Biology, University of the Philippines Manila, 1000, Manila, Philippines.
| |
Collapse
|