1
|
Tourdot S, Bloem K, Champion L, De Groot AS, Ducret A, Garidel P, Grudzinska-Goebel J, Gutknecht M, Hickling T, Horling F, Ichetovkin M, Johnson A, Jyamubandi I, Karle A, Kromminga A, Kurtulmus EA, Loeff F, Maillere B, Michaut L, Minelli F, Nielsen M, Nayak V, Nelson R, Pallardy M, Pattyn S, Pedras-Vasconelos J, Pepermans E, Poyau A, Reichel M, Rosenberg A, Sauna Z, Saxena M, Smith N, Snoeck V, Thoo L, Tovey M, Verthelyi D, Wuttke R, Yerly D, Kramer D. Proceedings of the 15 th European immunogenicity platform open symposium on immunogenicity of biopharmaceuticals. MAbs 2025; 17:2487604. [PMID: 40181240 DOI: 10.1080/19420862.2025.2487604] [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: 12/31/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
The European Immunogenicity Platform (EIP) celebrated the 15th edition of its Open Symposium on Immunogenicity of Biopharmaceuticals and its associated one-day workshop on 22-24 February 2024 in Lisbon. The meeting attracted experts and newcomers across industry, regulatory agencies, and academia, who actively participated in 3 days of discussion on risk assessment, monitoring, and mitigation of unwanted immunogenicity of biologics. Besides oral presentations, poster sessions were held to maximize scientific exchange and networking opportunities. Therapeutic proteins and emerging gene and cell-based therapies present promising therapeutic options for addressing unmet medical needs or when conventional treatment approaches have failed. Nonetheless, the development of an immune response against these therapeutic agents is a significant concern, as it occurs in a considerable number of cases across various products and indications. The specific anti-drug antibodies that develop can lead to adverse safety events, inhibition of drug activity, or accelerated clearance, all of which result in a loss of treatment efficacy. The EIP serves as a forum for experts and newcomers in the immunogenicity field, fostering discussion among scientists from industry and academia, encouraging interactions with regulatory agencies, and disseminating knowledge and advancements in immunogenicity sciences to the broader scientific community. This report covers the main topics discussed during the EIP 15th Open Symposium on Immunogenicity of Biopharmaceuticals, and the one-day workshop on practical aspects of immunogenicity held prior to the conference. Key topics included immunogenicity testing, clinical relevance of immunogenicity, immunogenicity risk assessment and mitigation, and current regulatory considerations.
Collapse
Affiliation(s)
- Sophie Tourdot
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Andover, MA, USA
| | - Karien Bloem
- Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - Lysie Champion
- Bioanalytical Services, Celerion Switzerland AG, Fehraltorf, Switzerland
| | | | - Axel Ducret
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Patrick Garidel
- IU-TIP, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach/Riss, Germany
| | | | - Michael Gutknecht
- Immunogenicity and Mechanistic Immunology, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Timothy Hickling
- Pharma Research and Early Development, Roche Innovation Centre Welwyn, Roche, Welwyn Garden City, UK
| | | | - Marina Ichetovkin
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Regulated Bioanalytics, Merck & Co., Inc., Rahway, NJ, USA
| | - Alison Johnson
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | | | - Anette Karle
- Immunogenicity and Mechanistic Immunology, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Floris Loeff
- R&D, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - Bernard Maillere
- Département Médicaments et Technologies pour la Santé, Université de Paris-Saclay, CEA, SIMoS, Gif-sur-Yvette, France
| | - Lydia Michaut
- PK Sciences, Novartis Biomedical Research, Basel, Switzerland
| | - Francesca Minelli
- NBE-DMPK Innovative BioAnalytics, RBM Merck S.p.A., An Affiliate of Merck KGaA, Darmstadt, Germany
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Vivek Nayak
- Precision Medicine, UCB, Braine-l'Alleud, Belgium
| | | | - Marc Pallardy
- INSERM, Inflammation, Microbiome and Immunosurveillance, University Paris-Saclay, Orsay, France
| | - Sofie Pattyn
- In Vitro Immunology, RIqvia Laboratories, Gosselies, Belgium
| | - Joao Pedras-Vasconelos
- Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Product Quality Assessment III, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Alain Poyau
- LBA Method Development, KCAS Bio, Lyon, France
| | | | | | - Zuben Sauna
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Manisha Saxena
- PK Sciences - Translational Medicine, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Lester Thoo
- Adverse Drug Reaction - Analysis & Consulting, ADR-AC GmbH, Bern, Switzerland
| | - Michael Tovey
- Chief Scientific Advisor Svar Life Science AB, Villejuif, France
| | - Daniela Verthelyi
- Division IV, Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Rene Wuttke
- Translational Medicine, Bioanalysis, Debiopharm International SA, Lausanne, Switzerland
| | - Daniel Yerly
- Adverse Drug Reaction - Analysis & Consulting, ADR-AC GmbH, Bern, Switzerland
| | - Daniel Kramer
- Translational Medicine Unit, Sanofi, Frankfurt am Main, Germany
| |
Collapse
|
2
|
Mandal S, Chanu WP, Natarajaseenivasan K. Development of a multi-epitope vaccine candidate to combat SARS-CoV-2 and dengue virus co-infection through an immunoinformatic approach. Front Immunol 2025; 16:1442101. [PMID: 40079004 PMCID: PMC11897530 DOI: 10.3389/fimmu.2025.1442101] [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: 06/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background Although the SARS-CoV-2 and dengue viruses seriously endanger human health, there is presently no vaccine that can stop a person from contracting both viruses at the same time. In this study, four antigens from SARS-CoV-2 and dengue virus were tested for immunogenicity, antigenicity, allergenicity, and toxicity and chosen to predict dominant T- and B-cell epitopes. Methods For designing a multi-epitope vaccine, the sequences were retrieved, and using bioinformatics and immunoinformatics, the physicochemical and immunological properties, as well as secondary structures, of the vaccine were predicted and studied. Additionally, the three-dimensional structure was estimated, improved upon, and confirmed using bioinformatics methods before being docked with TLR-2 and TLR-4. Eight helper T-cell lymphocyte (HTL) epitopes, ten cytotoxic T-cell lymphocyte (CTL) epitopes, nine B-cell epitopes, and TLR agonists were used to create a new multi-epitope vaccine. Furthermore, according to the immunological stimulation hypothesis, the vaccine could stimulate T and B cells to create large quantities of Th1 cytokines and antibodies. Results The study indicates that the developed vaccine is a favorable vaccine candidate with antigenicity, immunogenicity, non-toxicity, and non-allergenicity properties. The vaccine construct was made up of 460 amino acids, had an MW of 49391.51 Da, a theoretical pI of 9.86, and the formula C2203H3433N643O618S18, a lipid index of 39.84, a GRAVY of -0.473, an aliphatic index of 63.80, and an instability index of 39.84, which classifies the protein to be stable. Conclusion The acquired data showed that both vaccine designs had a considerable chance of preventing the co-infection of SARS-CoV-2 and dengue virus and that they demonstrate good results following in-silico testing. Furthermore, the vaccine may be an effective strategy in preventing SARS-CoV-2 and dengue since it can cause noticeably high levels of Th1 cytokines and antibodies.
Collapse
Affiliation(s)
- Saurav Mandal
- Division of Metabolomics, Proteomics & Imaging facility, Regional Medical Research Centre, Indian Council of Medical Research (ICMR), Dibrugarh, Assam, India
| | - Waribam Pratibha Chanu
- Department of Applied Physics, School of Vocational Studies and Applied Sciences (SoVSAS), Gautam Buddha University, Greater Noida, Uttar Pradesh, India
| | - Kalimuthusamy Natarajaseenivasan
- Division of Metabolomics, Proteomics & Imaging facility, Regional Medical Research Centre, Indian Council of Medical Research (ICMR), Dibrugarh, Assam, India
| |
Collapse
|
3
|
Sun Y, Pumroy RA, Mallik L, Chaudhuri A, Wang C, Hwang D, Danon JN, Dasteh Goli K, Moiseenkova-Bell VY, Sgourakis NG. CryoEM structure of an MHC-I/TAPBPR peptide-bound intermediate reveals the mechanism of antigen proofreading. Proc Natl Acad Sci U S A 2025; 122:e2416992122. [PMID: 39786927 PMCID: PMC11745410 DOI: 10.1073/pnas.2416992122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/05/2024] [Indexed: 01/30/2025] Open
Abstract
Class I major histocompatibility complex (MHC-I) proteins play a pivotal role in adaptive immunity by displaying epitopic peptides to CD8+ T cells. The chaperones tapasin and TAPBPR promote the selection of immunogenic antigens from a large pool of intracellular peptides. Interactions of chaperoned MHC-I molecules with incoming peptides are transient in nature, and as a result, the precise antigen proofreading mechanism remains elusive. Here, we leverage a high-fidelity TAPBPR variant and conformationally stabilized MHC-I, to determine the solution structure of the human antigen editing complex bound to a peptide decoy by cryogenic electron microscopy (cryo-EM) at an average resolution of 3.0 Å. Antigen proofreading is mediated by transient interactions formed between the nascent peptide binding groove with the P2/P3 peptide anchors, where conserved MHC-I residues stabilize incoming peptides through backbone-focused contacts. Finally, using our high-fidelity chaperone, we demonstrate robust peptide exchange on the cell surface across multiple clinically relevant human MHC-I allomorphs. Our work has important ramifications for understanding the selection of immunogenic epitopes for T cell screening and vaccine design applications.
Collapse
Affiliation(s)
- Yi Sun
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Ruth A. Pumroy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Leena Mallik
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Apala Chaudhuri
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Chloe Wang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Immunology Graduate Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
| | - Daniel Hwang
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Julia N. Danon
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Kimia Dasteh Goli
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Vera Y. Moiseenkova-Bell
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Nikolaos G. Sgourakis
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| |
Collapse
|
4
|
Conte A, Gulmini N, Costa F, Cartura M, Bröhl F, Patanè F, Filippini F. NERVE 2.0: boosting the new enhanced reverse vaccinology environment via artificial intelligence and a user-friendly web interface. BMC Bioinformatics 2024; 25:378. [PMID: 39695945 PMCID: PMC11654298 DOI: 10.1186/s12859-024-06004-0] [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: 07/14/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Vaccines development in this millennium started by the milestone work on Neisseria meningitidis B, reporting the invention of Reverse Vaccinology (RV), which allows to identify vaccine candidates (VCs) by screening bacterial pathogens genome or proteome through computational analyses. When NERVE (New Enhanced RV Environment), the first RV software integrating tools to perform the selection of VCs, was released, it prompted further development in the field. However, the problem-solving potential of most, if not all, RV programs is still largely unexploited by experimental vaccinologists that impaired by somehow difficult interfaces, requiring bioinformatic skills. RESULTS We report here on the development and release of NERVE 2.0 (available at: https://nerve-bio.org ) which keeps the original integrative and modular approach of NERVE, while showing higher predictive performance than its previous version and other web-RV programs (Vaxign and Vaxijen). We renewed some of its modules and added innovative ones, such as Loop-Razor, to recover fragments of promising vaccine candidates or Epitope Prediction for the epitope prediction binding affinities and population coverage. Along with two newly built AI (Artificial Intelligence)-based models: ESPAAN and Virulent. To improve user-friendliness, NERVE was shifted to a tutored, web-based interface, with a noSQL-database to consent the user to submit, obtain and retrieve analysis results at any moment. CONCLUSIONS With its redesigned and updated environment, NERVE 2.0 allows customisable and refinable bacterial protein vaccine analyses to all different kinds of users.
Collapse
Affiliation(s)
- Andrea Conte
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, Padua, Italy
| | - Nicola Gulmini
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, Padua, Italy
| | - Francesco Costa
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, Padua, Italy
- EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, UK
| | - Matteo Cartura
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, Padua, Italy
| | | | - Francesco Patanè
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, Padua, Italy
| | - Francesco Filippini
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
5
|
Sun Y, Pumroy RA, Mallik L, Chaudhuri A, Wang C, Hwang D, Danon JN, Goli KD, Moiseenkova-Bell V, Sgourakis NG. CryoEM structure of an MHC-I/TAPBPR peptide bound intermediate reveals the mechanism of antigen proofreading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606663. [PMID: 39211162 PMCID: PMC11361172 DOI: 10.1101/2024.08.05.606663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Class I major histocompatibility complex (MHC-I) proteins play a pivotal role in adaptive immunity by displaying epitopic peptides to CD8+ T cells. The chaperones tapasin and TAPBPR promote the selection of immunogenic antigens from a large pool of intracellular peptides. Interactions of chaperoned MHC-I molecules with incoming peptides are transient in nature, and as a result, the precise antigen proofreading mechanism remains elusive. Here, we leverage a high-fidelity TAPBPR variant and conformationally stabilized MHC-I, to determine the solution structure of the human antigen editing complex bound to a peptide decoy by cryogenic electron microscopy (cryo-EM) at an average resolution of 3.0 Å. Antigen proofreading is mediated by transient interactions formed between the nascent peptide binding groove with the P2/P3 peptide anchors, where conserved MHC-I residues stabilize incoming peptides through backbone-focused contacts. Finally, using our high-fidelity chaperone, we demonstrate robust peptide exchange on the cell surface across multiple clinically relevant human MHC-I allomorphs. Our work has important ramifications for understanding the selection of immunogenic epitopes for T cell screening and vaccine design applications.
Collapse
|
6
|
Yang Q, Xu L, Dong W, Li X, Wang K, Dong S, Zhang X, Yang T, Jiang F, Zhang B, Luo G, Gao X, Wang G. HLAIImaster: a deep learning method with adaptive domain knowledge predicts HLA II neoepitope immunogenic responses. Brief Bioinform 2024; 25:bbae302. [PMID: 38920343 PMCID: PMC11200192 DOI: 10.1093/bib/bbae302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
While significant strides have been made in predicting neoepitopes that trigger autologous CD4+ T cell responses, accurately identifying the antigen presentation by human leukocyte antigen (HLA) class II molecules remains a challenge. This identification is critical for developing vaccines and cancer immunotherapies. Current prediction methods are limited, primarily due to a lack of high-quality training epitope datasets and algorithmic constraints. To predict the exogenous HLA class II-restricted peptides across most of the human population, we utilized the mass spectrometry data to profile >223 000 eluted ligands over HLA-DR, -DQ, and -DP alleles. Here, by integrating these data with peptide processing and gene expression, we introduce HLAIImaster, an attention-based deep learning framework with adaptive domain knowledge for predicting neoepitope immunogenicity. Leveraging diverse biological characteristics and our enhanced deep learning framework, HLAIImaster is significantly improved against existing tools in terms of positive predictive value across various neoantigen studies. Robust domain knowledge learning accurately identifies neoepitope immunogenicity, bridging the gap between neoantigen biology and the clinical setting and paving the way for future neoantigen-based therapies to provide greater clinical benefit. In summary, we present a comprehensive exploitation of the immunogenic neoepitope repertoire of cancers, facilitating the effective development of "just-in-time" personalized vaccines.
Collapse
Affiliation(s)
- Qiang Yang
- School of Medicine and Health, Harbin Institute of Technology, Yikuang Street, Harbin 150000, China
| | - Long Xu
- School of Computer Science and Technology, Harbin Institute of Technology, West Dazhi Street, Harbin 150001, China
| | - Weihe Dong
- College of Computer and Control Engineering, Northeast Forestry University, Hexing Road, Harbin 150004, China
| | - Xiaokun Li
- School of Computer Science and Technology, Harbin Institute of Technology, West Dazhi Street, Harbin 150001, China
- School of Computer Science and Technology, Heilongjiang University, Xuefu Road, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Xuefu Road, Harbin 150090, China
- Shandong Hengxun Technology Co., Ltd., Miaoling Road, Qingdao 266100, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, West Dazhi Street, Harbin 150001, China
| | - Suyu Dong
- College of Computer and Control Engineering, Northeast Forestry University, Hexing Road, Harbin 150004, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road, Harbin 150081, China
| | - Tiansong Yang
- Department of Rehabilitation, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, and Traditional Chinese Medicine Informatics Key Laboratory of Heilongjiang Province, Heping Road, Harbin 150040, China
| | - Feng Jiang
- School of Medicine and Health, Harbin Institute of Technology, Yikuang Street, Harbin 150000, China
| | - Bin Zhang
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| | - Gongning Luo
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University, Hexing Road, Harbin 150004, China
| |
Collapse
|
7
|
Lee CK, Lee JB, Park SJ, Che J, Kwon WS, Kim HS, Jung M, Lee S, Park SR, Koo DH, Lee HW, Bae WK, Jeung HC, Hwang IG, Kim H, Nam CM, Chung HC, Rha SY. Second-line chemoimmunotherapy with nivolumab and paclitaxel in immune-related biomarker-enriched advanced gastric cancer: a multicenter phase Ib/II study. Gastric Cancer 2024; 27:118-130. [PMID: 37906316 DOI: 10.1007/s10120-023-01435-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/29/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND We conducted a trial to evaluate the efficacy and safety of nivolumab and paclitaxel as second-line therapy for immune-related biomarker-enriched advanced gastric cancer (AGC). METHODS This open-label, single-arm, phase Ib/II study was a part of multi-institutional, biomarker-integrated umbrella study conducted in Korea. In phase Ib, patients received nivolumab (3 mg/kg) on Days 1 and 15 and paclitaxel (dose level 1, 70 mg/m2 or dose level 2, 80 mg/m2) on Days 1, 8, 15 every four weeks. In phase II, patients with Epstein-Barr virus-related, deficient mismatch repair or programmed cell death-ligand-1-positive AGC were enrolled. The primary endpoints were recommended phase II dose (RP2D, phase Ib) and progression-free survival (PFS, phase II). Secondary endpoints included objective response rate (ORR), overall survival (OS), safety, and exploratory biomarker analysis. RESULTS Dose level 2 was selected as RP2D. In phase II, 48 patients were enrolled. The median PFS and OS were 3.9 and 11.2 months, respectively. The ORR was 23.3%, and the median response duration was 16.7 months. Grade 3 or higher treatment-related adverse events, mainly neutropenia, occurred in 20 patients (41.7%). Targeted sequencing revealed that patients with RTK/RAS pathway alterations or the HLA-A02 supertype had better survival. Patients with elevated baseline interleukin-1 receptor antagonist levels had worse survival. CONCLUSIONS Although the study did not meet its primary end point, nivolumab and paclitaxel for AGC demonstrated a durable response with manageable toxicity profiles. Genomic analysis or plasma cytokine analysis may provide information for the selection of patients who would benefit more from immunotherapy combined with chemotherapy.
Collapse
Affiliation(s)
- Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
| | - Se Jung Park
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Jingmin Che
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Sun Kwon
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyo Song Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Seulkee Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sook Ryun Park
- Division of Oncology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dong-Hoe Koo
- Division of Hematology/Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, South Korea
| | - Woo Kyun Bae
- Division of Hematology and Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, South Korea
| | - Hei-Cheul Jeung
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - In Gyu Hwang
- Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea.
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
8
|
Papadaki GF, Woodward CH, Young MC, Winters TJ, Burslem GM, Sgourakis NG. A Chicken Tapasin ortholog can chaperone empty HLA-B∗37:01 molecules independent of other peptide-loading components. J Biol Chem 2023; 299:105136. [PMID: 37543367 PMCID: PMC10534222 DOI: 10.1016/j.jbc.2023.105136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/07/2023] Open
Abstract
Human Tapasin (hTapasin) is the main chaperone of MHC-I molecules, enabling peptide loading and antigen repertoire optimization across HLA allotypes. However, it is restricted to the endoplasmic reticulum (ER) lumen as part of the protein loading complex (PLC), and therefore is highly unstable when expressed in recombinant form. Additional stabilizing co-factors such as ERp57 are required to catalyze peptide exchange in vitro, limiting uses for the generation of pMHC-I molecules of desired antigen specificities. Here, we show that the chicken Tapasin (chTapasin) ortholog can be expressed recombinantly at high yields in a stable form, independent of co-chaperones. chTapasin can bind the human HLA-B∗37:01 with low micromolar-range affinity to form a stable tertiary complex. Biophysical characterization by methyl-based NMR methods reveals that chTapasin recognizes a conserved β2m epitope on HLA-B∗37:01, consistent with previously solved X-ray structures of hTapasin. Finally, we provide evidence that the B∗37:01/chTapasin complex is peptide-receptive and can be dissociated upon binding of high-affinity peptides. Our results highlight the use of chTapasin as a stable scaffold for protein engineering applications aiming to expand the ligand exchange function on human MHC-I and MHC-like molecules.
Collapse
Affiliation(s)
- Georgia F Papadaki
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Claire H Woodward
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael C Young
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Trenton J Winters
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nikolaos G Sgourakis
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
9
|
Zucco AG, Bennedbæk M, Ekenberg C, Gabrielaite M, Leung P, Polizzotto MN, Kan V, Murray DD, Lundgren JD, MacPherson CR. Associations of functional human leucocyte antigen class I groups with HIV viral load in a heterogeneous cohort. AIDS 2023; 37:1643-1650. [PMID: 37534724 PMCID: PMC10399941 DOI: 10.1097/qad.0000000000003557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023]
Abstract
OBJECTIVE Human leucocyte antigen (HLA) class I alleles are the main host genetic factors involved in controlling HIV-1 viral load (VL). Nevertheless, HLA diversity has proven a significant challenge in association studies. We assessed how accounting for binding affinities of HLA class I alleles to HIV-1 peptides facilitate association testing of HLA with HIV-1 VL in a heterogeneous cohort. DESIGN Cohort from the Strategic Timing of AntiRetroviral Treatment (START) study. METHODS We imputed HLA class I alleles from host genetic data (2546 HIV+ participants) and sampled immunopeptidomes from 2079 host-paired viral genomes (targeted amplicon sequencing). We predicted HLA class I binding affinities to HIV-1 and unspecific peptides, grouping alleles into functional clusters through consensus clustering. These functional HLA class I clusters were used to test associations with HIV VL. RESULTS We identified four clades totaling 30 HLA alleles accounting for 11.4% variability in VL. We highlight HLA-B∗57:01 and B∗57:03 as functionally similar but yet overrepresented in distinct ethnic groups, showing when combined a protective association with HIV+ VL (log, β -0.25; adj. P-value < 0.05). We further demonstrate only a slight power reduction when using unspecific immunopeptidomes, facilitating the use of the inferred functional HLA groups in other studies. CONCLUSION The outlined computational approach provides a robust and efficient way to incorporate HLA function and peptide diversity, aiding clinical association studies in heterogeneous cohorts. To facilitate access to the proposed methods and results we provide an interactive application for exploring data.
Collapse
Affiliation(s)
| | - Marc Bennedbæk
- Virus Research and Development Laboratory, Virus and Microbiological Special Diagnostics, Statens Serum Institut
| | | | - Migle Gabrielaite
- Center for Genomic Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Mark N. Polizzotto
- Clinical Hub for Interventional Research, College of Health and Medicine, The Australian National University, Canberra, Australia
| | - Virginia Kan
- George Washington University, Veterans Affairs Medical Center, Washington, DC, USA
| | | | | | | |
Collapse
|
10
|
Papadaki GF, Woodward CH, Young MC, Winters TJ, Burslem GM, Sgourakis NG. A Chicken Tapasin ortholog can chaperone empty HLA molecules independently of other peptide-loading components. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.23.546255. [PMID: 37425753 PMCID: PMC10326978 DOI: 10.1101/2023.06.23.546255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Human Tapasin (hTapasin) is the main chaperone of MHC-I molecules, enabling peptide loading and antigen repertoire optimization across HLA allotypes. However, it is restricted to the endoplasmic reticulum (ER) lumen as part of the protein loading complex (PLC) and therefore is highly unstable when expressed in recombinant form. Additional stabilizing co-factors such as ERp57 are required to catalyze peptide exchange in vitro , limiting uses for the generation of pMHC-I molecules of desired antigen specificities. Here, we show that the chicken Tapasin (chTapasin) ortholog can be expressed recombinantly at high yields in stable form, independently of co-chaperones. chTapasin can bind the human HLA-B * 37:01 with low micromolar-range affinity to form a stable tertiary complex. Biophysical characterization by methyl-based NMR methods reveals that chTapasin recognizes a conserved β 2 m epitope on HLA-B * 37:01, consistent with previously solved X-ray structures of hTapasin. Finally, we provide evidence that the B * 37:01/chTapasin complex is peptide-receptive and can be dissociated upon binding of high-affinity peptides. Our results highlight the use of chTapasin as a stable scaffold for future protein engineering applications aiming to expand the ligand exchange function on human MHC-I and MHC-like molecules.
Collapse
|
11
|
Stephan-Falkenau S, Streubel A, Mairinger T, Blum TG, Kollmeier J, Mairinger FD, Bauer T, Pfannschmidt J, Hollmann M, Wessolly M. Integrated Clinical, Molecular and Immunological Characterization of Pulmonary Sarcomatoid Carcinomas Reveals an Immune Escape Mechanism That May Influence Therapeutic Strategies. Int J Mol Sci 2023; 24:10558. [PMID: 37445733 DOI: 10.3390/ijms241310558] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Pulmonary sarcomatoid carcinoma (PSC) has highly aggressive biological behaviour and poor clinical outcomes, raising expectations for new therapeutic strategies. We characterized 179 PSC by immunohistochemistry, next-generation sequencing and in silico analysis using a deep learning algorithm with respect to clinical, immunological and molecular features. PSC was more common in men, older ages and smokers. Surgery was an independent factor (p < 0.01) of overall survival (OS). PD-L1 expression was detected in 82.1% of all patients. PSC patients displaying altered epitopes due to processing mutations showed another PD-L1-independent immune escape mechanism, which also significantly influenced OS (p < 0.02). The effect was also maintained when only advanced tumour stages were considered (p < 0.01). These patients also showed improved survival with a significant correlation for immunotherapy (p < 0.05) when few or no processing mutations were detected, although this should be interpreted with caution due to the small number of patients studied. Genomic alterations for which there are already approved drugs were present in 35.4% of patients. Met exon 14 skipping was found more frequently (13.7%) and EGFR mutations less frequently (1.7%) than in other NSCLC. In summary, in addition to the divergent genomic landscape of PSC, the specific immunological features of this prognostically poor subtype should be considered in therapy stratification.
Collapse
Affiliation(s)
- Susann Stephan-Falkenau
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Anna Streubel
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Thomas Mairinger
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Torsten-Gerriet Blum
- Department of Pneumology, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Jens Kollmeier
- Department of Pneumology, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Fabian D Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Torsten Bauer
- Department of Pneumology, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Joachim Pfannschmidt
- Department of Thoracic Surgery, Heckeshorn Lung Clinic, Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Manuel Hollmann
- Institute for Tissue Diagnostics, MVZ at Helios Klinikum Emil von Behring, 14165 Berlin, Germany
| | - Michael Wessolly
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| |
Collapse
|
12
|
Sun Y, Papadaki GF, Devlin CA, Danon JN, Young MC, Winters TJ, Burslem GM, Procko E, Sgourakis NG. Xeno interactions between MHC-I proteins and molecular chaperones enable ligand exchange on a broad repertoire of HLA allotypes. SCIENCE ADVANCES 2023; 9:eade7151. [PMID: 36827371 PMCID: PMC9956121 DOI: 10.1126/sciadv.ade7151] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/19/2023] [Indexed: 06/01/2023]
Abstract
Immunological chaperones tapasin and TAP binding protein, related (TAPBPR) play key roles in antigenic peptide optimization and quality control of nascent class I major histocompatibility complex (MHC-I) molecules. The polymorphic nature of MHC-I proteins leads to a range of allelic dependencies on chaperones for assembly and cell-surface expression, limiting chaperone-mediated peptide exchange to a restricted set of human leukocyte antigen (HLA) allotypes. Here, we demonstrate and characterize xeno interactions between a chicken TAPBPR ortholog and a complementary repertoire of HLA allotypes, relative to its human counterpart. We find that TAPBPR orthologs recognize empty MHC-I with broader allele specificity and facilitate peptide exchange by maintaining a reservoir of receptive molecules. Deep mutational scanning of human TAPBPR further identifies gain-of-function mutants, resembling the chicken sequence, which can enhance HLA-A*01:01 expression in situ and promote peptide exchange in vitro. These results highlight that polymorphic sites on MHC-I and chaperone surfaces can be engineered to manipulate their interactions, enabling chaperone-mediated peptide exchange on disease-relevant HLA alleles.
Collapse
Affiliation(s)
- Yi Sun
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Georgia F. Papadaki
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Christine A. Devlin
- Department of Biochemistry and Cancer Center at Illinois, University of Illinois, Urbana, IL 61820, USA
| | - Julia N. Danon
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Michael C. Young
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Trenton J. Winters
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - George M. Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erik Procko
- Department of Biochemistry and Cancer Center at Illinois, University of Illinois, Urbana, IL 61820, USA
| | - Nikolaos G. Sgourakis
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3501 Civic Center Blvd., Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Omoboyede V, Ibrahim O, Umar HI, Bello T, Adedeji AA, Khalid A, Fayojegbe ES, Ayomide AB, Chukwuemeka PO. Designing a vaccine-based therapy against Epstein-Barr virus-associated tumors using immunoinformatics approach. Comput Biol Med 2022; 150:106128. [PMID: 36179514 DOI: 10.1016/j.compbiomed.2022.106128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/05/2022] [Accepted: 09/18/2022] [Indexed: 11/26/2022]
Abstract
Epstein-Barr virus (EBV) is widely known due to its role in the etiology of infectious mononucleosis. However, it is the first oncovirus that was identified and has been implicated in the etiology of several types of cancers. Globally, EBV infection is associated with more than 200, 000 new cancer cases and 150, 000 deaths yearly. A prophylactic or therapeutic vaccine targeting tumors associated with EBV infection is currently lacking. Therefore, this study aimed to develop a multiepitope-based polyvalent vaccine against EBV-associated tumors using immunoinformatics approach. The latency-associated proteins (LAP) of three strains of the virus were used in this study. Potential epitopes predicted from the proteins were analyzed and selected based on several predicted properties. Thirty viable B-cell and T-cell epitopes were selected and conjugated using various linkers alongside beta-defensin 3 as an adjuvant and pan HLA DR-binding epitope (PADRE) sequence to improve the immunogenicity of the vaccine construct. Molecular docking studies of the vaccine construct against toll-like receptors (TLRs) showed it is capable of inducing immune response via recognition by TLRs while immune simulation studies showed it could induce both cellular and humoral immune responses. Furthermore, molecular dynamics study of the complex formed by the vaccine candidate and TLR-4 showed that the complex was stable. Ultimately, the designed vaccine showed desirable properties based on in silico evaluation; however, experimental studies are needed to validate the efficacy of the vaccine against EBV-associated tumors.
Collapse
Affiliation(s)
- Victor Omoboyede
- Department of Biochemistry, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Computer Aided Therapeutics Laboratory (CATL) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
| | - Ochapa Ibrahim
- Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
| | - Haruna Isiyaku Umar
- Department of Biochemistry, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
| | - Taye Bello
- Department of Medical Rehabilitation, College of Health Sciences, Obafemi Awolowo University, Nigeria.
| | - Ayodeji Adeola Adedeji
- Department of Biochemistry, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
| | - Aqsa Khalid
- Research Center for Modelling and Simulation (RCMS), National University of Science and Technology (NUST), Islamabad, Pakistan.
| | | | - Adunola Blessing Ayomide
- Computer Aided Therapeutics Laboratory (CATL) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Department of Biotechnology, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
| | - Prosper Obed Chukwuemeka
- Computer Aided Therapeutics Laboratory (CATL) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria; Department of Biotechnology, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
| |
Collapse
|
14
|
An In Silico Analysis of Malaria Pre-Erythrocytic-Stage Antigens Interpreting Worldwide Genetic Data to Suggest Vaccine Candidate Variants and Epitopes. Microorganisms 2022; 10:microorganisms10061090. [PMID: 35744609 PMCID: PMC9231253 DOI: 10.3390/microorganisms10061090] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
Failure to account for genetic diversity of antigens during vaccine design may lead to vaccine escape. To evaluate the vaccine escape potential of antigens used in vaccines currently in development or clinical testing, we surveyed the genetic diversity, measured population differentiation, and performed in silico prediction and analysis of T-cell epitopes of ten such Plasmodium falciparum pre-erythrocytic-stage antigens using whole-genome sequence data from 1010 field isolates. Of these, 699 were collected in Africa (Burkina Faso, Cameroon, Guinea, Kenya, Malawi, Mali, and Tanzania), 69 in South America (Brazil, Colombia, French Guiana, and Peru), 59 in Oceania (Papua New Guinea), and 183 in Asia (Cambodia, Myanmar, and Thailand). Antigens surveyed include cell-traversal protein for ookinetes and sporozoites, circumsporozoite protein, liver-stage antigens 1 and 3, sporozoite surface proteins P36 and P52, sporozoite asparagine-rich protein-1, sporozoite microneme protein essential for cell traversal-2, and upregulated-in-infectious-sporozoite 3 and 4 proteins. The analyses showed that a limited number of these protein variants, when combined, would be representative of worldwide parasite populations. Moreover, predicted T-cell epitopes were identified that could be further explored for immunogenicity and protective efficacy. Findings can inform the rational design of a multivalent malaria vaccine.
Collapse
|
15
|
Wessolly M, Mairinger FD, Herold T, Hadaschik B, Szarvas T, Reis H. Proteasomal Processing Immune Escape Mechanisms in Platinum-Treated Advanced Bladder Cancer. Genes (Basel) 2022; 13:genes13030422. [PMID: 35327977 PMCID: PMC8948673 DOI: 10.3390/genes13030422] [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: 12/30/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
In recent years, the number and type of treatment options in advanced bladder cancer (BC) have been rapidly evolving. To select an effective therapy and spare unnecessary side effects, predictive biomarkers are urgently needed. As the host’s anti-cancer immune response is by far the most effective system to impede malignant tumor growth, immune system-based biomarkers are promising. We have recently described altered proteasomal epitope processing as an effective immune escape mechanism to impair cytotoxic T-cell activity. By altering the neoantigens’ characteristics through different proteasomal peptide cleavage induced by non-synonymous somatic mutations, the ability for T-cell activation was decreased (“processing escapes”). In the present study, we analyzed primary chemo-naïve tissue samples of 26 adjuvant platinum-treated urothelial BC patients using a targeted next-generation sequencing panel followed by the epitope determination of affected genes, a machine-learning based prediction of epitope processing and proteasomal cleavage and of HLA-affinity as well as immune activation. Immune infiltration (immunohistochemistries for CD8, granzyme B, CD45/LCA) was digitally quantified by a pathologist and clinico-pathological and survival data were collected. We detected 145 epitopes with characteristics of a processing escape associated with a higher number of CD8-positive but lower number of granzyme B-positive cells and no association with PD-L1-expression. In addition, a high prevalence of processing escapes was associated with unfavorable overall survival. Our data indicate the presence of processing escapes in advanced BC, potentially creating a tumor-promoting pro-inflammatory environment with lowered anti-cancerous activity and independence from PD-L1-expression. The data also need to be prospectively validated in BC treated with immune therapy.
Collapse
Affiliation(s)
- Michael Wessolly
- Institute of Pathology, University Medicine Essen, University of Duisburg-Essen, 45147 Essen, Germany; (M.W.); (F.D.M.); (T.H.)
| | - Fabian D. Mairinger
- Institute of Pathology, University Medicine Essen, University of Duisburg-Essen, 45147 Essen, Germany; (M.W.); (F.D.M.); (T.H.)
| | - Thomas Herold
- Institute of Pathology, University Medicine Essen, University of Duisburg-Essen, 45147 Essen, Germany; (M.W.); (F.D.M.); (T.H.)
| | - Boris Hadaschik
- Department of Urology, University Medicine Essen, University of Duisburg-Essen, 45147 Essen, Germany; (B.H.); (T.S.)
| | - Tibor Szarvas
- Department of Urology, University Medicine Essen, University of Duisburg-Essen, 45147 Essen, Germany; (B.H.); (T.S.)
- Department of Urology, Semmelweis University Budapest, 1085 Budapest, Hungary
| | - Henning Reis
- Institute of Pathology, University Medicine Essen, University of Duisburg-Essen, 45147 Essen, Germany; (M.W.); (F.D.M.); (T.H.)
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-4514
| |
Collapse
|
16
|
Foix A, López D, Díez-Fuertes F, McConnell MJ, Martín-Galiano AJ. Predicted impact of the viral mutational landscape on the cytotoxic response against SARS-CoV-2. PLoS Comput Biol 2022; 18:e1009726. [PMID: 35143484 PMCID: PMC8830725 DOI: 10.1371/journal.pcbi.1009726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
The massive assessment of immune evasion due to viral mutations that increase COVID-19 susceptibility can be computationally facilitated. The adaptive cytotoxic T response is critical during primary infection and the generation of long-term protection. Here, potential HLA class I epitopes in the SARS-CoV-2 proteome were predicted for 2,915 human alleles of 71 families using the netMHCIpan EL algorithm. Allele families showed extreme epitopic differences, underscoring genetic variability of protective capacity between humans. Up to 1,222 epitopes were associated with any of the twelve supertypes, that is, allele clusters covering 90% population. Next, from all mutations identified in ~118,000 viral NCBI isolates, those causing significant epitope score reduction were considered epitope escape mutations. These mutations mainly involved non-conservative substitutions at the second and C-terminal position of the ligand core, or total ligand removal by large recurrent deletions. Escape mutations affected 47% of supertype epitopes, which in 21% of cases concerned isolates from two or more sub-continental areas. Some of these changes were coupled, but never surpassed 15% of evaded epitopes for the same supertype in the same isolate, except for B27. In contrast to most supertypes, eight allele families mostly contained alleles with few SARS-CoV-2 ligands. Isolates harboring cytotoxic escape mutations for these families co-existed geographically within sub-Saharan and Asian populations enriched in these alleles according to the Allele Frequency Net Database. Collectively, our findings indicate that escape mutation events have already occurred for half of HLA class I supertype epitopes. However, it is presently unlikely that, overall, it poses a threat to the global population. In contrast, single and double mutations for susceptible alleles may be associated with viral selective pressure and alarming local outbreaks. The integration of genomic, geographical and immunoinformatic information eases the surveillance of variants potentially affecting the global population, as well as minority subpopulations.
Collapse
Affiliation(s)
- Anna Foix
- European Bioinformatic Institute, European Molecular Biology Laboratory, Hinxton, United Kingdom
| | - Daniel López
- Presentation and Immune Regulation Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Francisco Díez-Fuertes
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Michael J. McConnell
- Intrahospital Infections Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Antonio J. Martín-Galiano
- Intrahospital Infections Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain
| |
Collapse
|
17
|
Joyce S, Ternette N. Know thy immune self and non-self: Proteomics informs on the expanse of self and non-self, and how and where they arise. Proteomics 2021; 21:e2000143. [PMID: 34310018 PMCID: PMC8865197 DOI: 10.1002/pmic.202000143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/30/2021] [Accepted: 07/19/2021] [Indexed: 12/30/2022]
Abstract
T cells play an important role in the adaptive immune response to a variety of infections and cancers. Initiation of a T cell mediated immune response requires antigen recognition in a process termed MHC (major histocompatibility complex) restri ction. A T cell antigen is a composite structure made up of a peptide fragment bound within the antigen-binding groove of an MHC-encoded class I or class II molecule. Insight into the precise composition and biology of self and non-self immunopeptidomes is essential to harness T cell mediated immunity to prevent, treat, or cure infectious diseases and cancers. T cell antigen discovery is an arduous task! The pioneering work in the early 1990s has made large-scale T cell antigen discovery possible. Thus, advancements in mass spectrometry coupled with proteomics and genomics technologies make possible T cell antigen discovery with ease, accuracy, and sensitivity. Yet we have only begun to understand the breadth and the depth of self and non-self immunopeptidomes because the molecular biology of the cell continues to surprise us with new secrets directly related to the source, and the processing and presentation of MHC ligands. Focused on MHC class I molecules, this review, therefore, provides a brief historic account of T cell antigen discovery and, against a backdrop of key advances in molecular cell biologic processes, elaborates on how proteogenomics approaches have revolutionised the field.
Collapse
Affiliation(s)
- Sebastian Joyce
- Department of Veterans AffairsTennessee Valley Healthcare System and the Department of PathologyMicrobiology and ImmunologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Nicola Ternette
- Centre for Cellular and Molecular PhysiologyNuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
18
|
Talarico L, Marta S, Rossi AR, Crescenzo S, Petrosino G, Martinoli M, Tancioni L. Balancing selection, genetic drift, and human-mediated introgression interplay to shape MHC (functional) diversity in Mediterranean brown trout. Ecol Evol 2021; 11:10026-10041. [PMID: 34367556 PMCID: PMC8328470 DOI: 10.1002/ece3.7760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022] Open
Abstract
The extraordinary polymorphism of major histocompatibility complex (MHC) genes is considered a paradigm of pathogen-mediated balancing selection, although empirical evidence is still scarce. Furthermore, the relative contribution of balancing selection to shape MHC population structure and diversity, compared to that of neutral forces, as well as its interaction with other evolutionary processes such as hybridization, remains largely unclear. To investigate these issues, we analyzed adaptive (MHC-DAB gene) and neutral (11 microsatellite loci) variation in 156 brown trout (Salmo trutta complex) from six wild populations in central Italy exposed to introgression from domestic hatchery lineages (assessed with the LDH gene). MHC diversity and structuring correlated with those at microsatellites, indicating the substantial role of neutral forces. However, individuals carrying locally rare MHC alleles/supertypes were in better body condition (a proxy of individual fitness/parasite load) regardless of the zygosity status and degree of sequence dissimilarity of MHC, hence supporting balancing selection under rare allele advantage, but not heterozygote advantage or divergent allele advantage. The association between specific MHC supertypes and body condition confirmed in part this finding. Across populations, MHC allelic richness increased with increasing admixture between native and domestic lineages, indicating introgression as a source of MHC variation. Furthermore, introgression across populations appeared more pronounced for MHC than microsatellites, possibly because initially rare MHC variants are expected to introgress more readily under rare allele advantage. Providing evidence for the complex interplay among neutral evolutionary forces, balancing selection, and human-mediated introgression in shaping the pattern of MHC (functional) variation, our findings contribute to a deeper understanding of the evolution of MHC genes in wild populations exposed to anthropogenic disturbance.
Collapse
Affiliation(s)
- Lorenzo Talarico
- Laboratory of Experimental Ecology and AquacultureDepartment of BiologyUniversity of Rome “Tor Vergata”RomeItaly
| | - Silvio Marta
- Department of Environmental Science and PolicyUniversity of MilanMilanItaly
| | - Anna Rita Rossi
- Department of Biology and Biotechnology C. DarwinUniversity of Rome “La Sapienza”RomeItaly
| | - Simone Crescenzo
- Department of Biology and Biotechnology C. DarwinUniversity of Rome “La Sapienza”RomeItaly
| | - Gerardo Petrosino
- Department of Biology and Biotechnology C. DarwinUniversity of Rome “La Sapienza”RomeItaly
| | - Marco Martinoli
- Laboratory of Experimental Ecology and AquacultureDepartment of BiologyUniversity of Rome “Tor Vergata”RomeItaly
- Consiglio per la Ricerca in Agricoltura e l'Analisi dell'Economia Agraria (CREA)Centro di Zootecnia e AcquacolturaMonterotondoItaly
| | - Lorenzo Tancioni
- Laboratory of Experimental Ecology and AquacultureDepartment of BiologyUniversity of Rome “Tor Vergata”RomeItaly
| |
Collapse
|
19
|
Prakash S, Srivastava R, Coulon PG, Dhanushkodi NR, Chentoufi AA, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, Buchmeier MJ, Bouziane M, Nesburn AB, Kuppermann BD, BenMohamed L. Genome-Wide B Cell, CD4 +, and CD8 + T Cell Epitopes That Are Highly Conserved between Human and Animal Coronaviruses, Identified from SARS-CoV-2 as Targets for Preemptive Pan-Coronavirus Vaccines. THE JOURNAL OF IMMUNOLOGY 2021; 206:2566-2582. [PMID: 33911008 DOI: 10.4049/jimmunol.2001438] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/16/2021] [Indexed: 02/06/2023]
Abstract
Over the last two decades, there have been three deadly human outbreaks of coronaviruses (CoVs) caused by SARS-CoV, MERS-CoV, and SARS-CoV-2, which has caused the current COVID-19 global pandemic. All three deadly CoVs originated from bats and transmitted to humans via various intermediate animal reservoirs. It remains highly possible that other global COVID pandemics will emerge in the coming years caused by yet another spillover of a bat-derived SARS-like coronavirus (SL-CoV) into humans. Determining the Ag and the human B cells, CD4+ and CD8+ T cell epitope landscapes that are conserved among human and animal coronaviruses should inform in the development of future pan-coronavirus vaccines. In the current study, using several immunoinformatics and sequence alignment approaches, we identified several human B cell and CD4+ and CD8+ T cell epitopes that are highly conserved in 1) greater than 81,000 SARS-CoV-2 genome sequences identified in 190 countries on six continents; 2) six circulating CoVs that caused previous human outbreaks of the common cold; 3) nine SL-CoVs isolated from bats; 4) nine SL-CoV isolated from pangolins; 5) three SL-CoVs isolated from civet cats; and 6) four MERS strains isolated from camels. Furthermore, the identified epitopes: 1) recalled B cells and CD4+ and CD8+ T cells from both COVID-19 patients and healthy individuals who were never exposed to SARS-CoV-2, and 2) induced strong B cell and T cell responses in humanized HLA-DR1/HLA-A*02:01 double-transgenic mice. The findings pave the way to develop a preemptive multiepitope pan-coronavirus vaccine to protect against past, current, and future outbreaks.
Collapse
Affiliation(s)
- Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Ruchi Srivastava
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Pierre-Gregoire Coulon
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Nisha R Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Aziz A Chentoufi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Delia F Tifrea
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA
| | - Robert A Edwards
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA
| | - Cesar J Figueroa
- Division of Trauma, Burns, Critical Care, and Acute Care Surgery, Department of Surgery, School of Medicine, University of California Irvine, Irvine, CA
| | - Sebastian D Schubl
- Division of Trauma, Burns, Critical Care, and Acute Care Surgery, Department of Surgery, School of Medicine, University of California Irvine, Irvine, CA
| | - Lanny Hsieh
- Division of Infectious Diseases and Hospitalist Program, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA
| | - Michael J Buchmeier
- Center for Virus Research, Division of Infectious Disease, School of Medicine, University of California Irvine, Irvine, CA
| | | | - Anthony B Nesburn
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Baruch D Kuppermann
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA; .,Center for Virus Research, Division of Infectious Disease, School of Medicine, University of California Irvine, Irvine, CA.,Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA
| |
Collapse
|
20
|
Evaluation of potential MHC-I allele-specific epitopes in Zika virus proteins and the effects of mutations on peptide-MHC-I interaction studied using in silico approaches. Comput Biol Chem 2021; 92:107459. [PMID: 33636637 DOI: 10.1016/j.compbiolchem.2021.107459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 11/24/2022]
Abstract
Zika virus (ZIKV) infection is a global health concern due to its association with microcephaly and neurological complications. The development of a T-cell vaccine is important to combat this disease. In this study, we propose ZIKV major histocompatibility complex I (MHC-I) epitopes based on in silico screening consensus followed by molecular docking, PRODIGY, and molecular dynamics (MD) simulation analyses. The effects of the reported mutations on peptide-MHC-I (pMHC-I) complexes were also evaluated. In general, our data indicate an allele-specific peptide-binding human leukocyte antigen (HLA) and potential epitopes. For HLA-B44, we showed that the absence of acidic residue Glu at P2, due to the loss of the electrostatic interaction with Lys45, has a negative impact on the pMHC-I complex stability and explains the low free energy estimated for the immunodominant peptide E-4 (IGVSNRDFV). Our MD data also suggest the deleterious effects of acidic residue Asp at P1 on the pMHC-I stability of HLA-B8 due to destabilization of the α-helix and β-strand. Free energy estimation further indicated that the mutation from Val to Ala at P9 of peptide E-247 (DAHAKRQTV), which was found exclusively in microcephaly samples, did not reduce HLA-B8 affinity. In contrast, the mutation from Thr to Pro at P2 of the peptide NS5-832 (VTKWTDIPY) decreased the interaction energy, number of intermolecular interactions, and adversely affected its binding mode with HLA-A1. Overall, our findings are important with regard to the design of T-cell peptide vaccines and for understanding how ZIKV escapes recognition by CD8 + T-cells.
Collapse
|
21
|
Oyarzun P, Kashyap M, Fica V, Salas-Burgos A, Gonzalez-Galarza FF, McCabe A, Jones AR, Middleton D, Kobe B. A Proteome-Wide Immunoinformatics Tool to Accelerate T-Cell Epitope Discovery and Vaccine Design in the Context of Emerging Infectious Diseases: An Ethnicity-Oriented Approach. Front Immunol 2021; 12:598778. [PMID: 33717077 PMCID: PMC7952308 DOI: 10.3389/fimmu.2021.598778] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/11/2021] [Indexed: 01/06/2023] Open
Abstract
Emerging infectious diseases (EIDs) caused by viruses are increasing in frequency, causing a high disease burden and mortality world-wide. The COVID-19 pandemic caused by the novel SARS-like coronavirus (SARS-CoV-2) underscores the need to innovate and accelerate the development of effective vaccination strategies against EIDs. Human leukocyte antigen (HLA) molecules play a central role in the immune system by determining the peptide repertoire displayed to the T-cell compartment. Genetic polymorphisms of the HLA system thus confer a strong variability in vaccine-induced immune responses and may complicate the selection of vaccine candidates, because the distribution and frequencies of HLA alleles are highly variable among different ethnic groups. Herein, we build on the emerging paradigm of rational epitope-based vaccine design, by describing an immunoinformatics tool (Predivac-3.0) for proteome-wide T-cell epitope discovery that accounts for ethnic-level variations in immune responsiveness. Predivac-3.0 implements both CD8+ and CD4+ T-cell epitope predictions based on HLA allele frequencies retrieved from the Allele Frequency Net Database. The tool was thoroughly assessed, proving comparable performances (AUC ~0.9) against four state-of-the-art pan-specific immunoinformatics methods capable of population-level analysis (NetMHCPan-4.0, Pickpocket, PSSMHCPan and SMM), as well as a strong accuracy on proteome-wide T-cell epitope predictions for HIV-specific immune responses in the Japanese population. The utility of the method was investigated for the COVID-19 pandemic, by performing in silico T-cell epitope mapping of the SARS-CoV-2 spike glycoprotein according to the ethnic context of the countries where the ChAdOx1 vaccine is currently initiating phase III clinical trials. Potentially immunodominant CD8+ and CD4+ T-cell epitopes and population coverages were predicted for each population (the Epitope Discovery mode), along with optimized sets of broadly recognized (promiscuous) T-cell epitopes maximizing coverage in the target populations (the Epitope Optimization mode). Population-specific epitope-rich regions (T-cell epitope clusters) were further predicted in protein antigens based on combined criteria of epitope density and population coverage. Overall, we conclude that Predivac-3.0 holds potential to contribute in the understanding of ethnic-level variations of vaccine-induced immune responsiveness and to guide the development of epitope-based next-generation vaccines against emerging pathogens, whose geographic distributions and populations in need of vaccinations are often well-defined for regional epidemics.
Collapse
Affiliation(s)
- Patricio Oyarzun
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Manju Kashyap
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Victor Fica
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | | | - Faviel F Gonzalez-Galarza
- Center for Biomedical Research, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Antony McCabe
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Andrew R Jones
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Derek Middleton
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Smallbone W, Ellison A, Poulton S, van Oosterhout C, Cable J. Depletion of MHC supertype during domestication can compromise immunocompetence. Mol Ecol 2020; 30:736-746. [PMID: 33274493 PMCID: PMC7898906 DOI: 10.1111/mec.15763] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/27/2022]
Abstract
The major histocompatibility complex (MHC) plays an important role in infectious disease resistance. The presence of certain MHC alleles and functionally similar groups of MHC alleles (i.e., supertypes) has been associated with resistance to particular parasite species. Farmed and domesticated fish stocks are often depleted in their MHC alleles and supertype diversity, possibly as a consequence of artificial selection for desirable traits, inbreeding (loss of heterozygosity), genetic drift (loss of allelic diversity) and/or reduced parasite biodiversity. Here we quantify the effects of depletion of MHC class II genotype and supertype variation on resistance to the parasite Gyrodactylus turnbulli in guppies (Poecilia reticulata). Compared to the descendants of wild‐caught guppies, ornamental fish had a significantly reduced MHC variation (i.e., the numbers of MHC alleles and supertypes per individual, and per population). In addition, ornamental fish were significantly more susceptible to G. turnbulli infections, accumulating peak intensity 10 times higher than that of their wildtype counterparts. Four out of 13 supertypes were associated with a significantly reduced parasite load, and the presence of some supertypes had a dramatic effect on the intensity of infection. Remarkably, the ornamental and wildtype fish differed in the supertypes that were associated with parasite resistance. Analysis with a genetic algorithm showed that resistance‐conferring supertypes of the wildtype and ornamental fish shared two unique amino acids in the peptide‐binding region of the MHC that were not found in any other alleles. These data show that the supertype demarcation captures some, but not all, of the variation in the immune function of the alleles. This study highlights the importance of managing functional MHC diversity in livestock, and suggests there might be some immunological redundancy among MHC supertypes.
Collapse
Affiliation(s)
| | - Amy Ellison
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Simon Poulton
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Cock van Oosterhout
- School of Environmental Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Joanne Cable
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
23
|
Sidney J, Peters B, Sette A. Epitope prediction and identification- adaptive T cell responses in humans. Semin Immunol 2020; 50:101418. [PMID: 33131981 DOI: 10.1016/j.smim.2020.101418] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/24/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022]
Abstract
Epitopes, in the context of T cell recognition, are short peptides typically derived by antigen processing, and presented on the cell surface bound to MHC molecules (HLA molecules in humans) for TCR scrutiny. The identification of epitopes is a context-dependent process, with consideration given to, for example, the source pathogen and protein, the host organism, and state of the immune reaction (e.g., following natural infection, vaccination, etc.). In the following review, we consider the various approaches used to define T cell epitopes, including both bioinformatic and experimental approaches, and discuss the concepts of immunodominance and immunoprevalence. We also discuss HLA polymorphism and epitope restriction, and the resulting impact on the identification of, and potential population coverage afforded by, epitopes or epitope-based vaccines. Finally, some examples of the practical application of T cell epitope identification are provided, showing how epitopes have been valuable for deriving novel immunological insights in the context of the immune response to various pathogens and allergens.
Collapse
Affiliation(s)
- John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
24
|
Prakash S, Srivastava R, Coulon PG, Dhanushkodi NR, Chentoufi AA, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, Buchmeier MJ, Bouziane M, Nesburn AB, Kuppermann BD, BenMohamed L. Genome-Wide Asymptomatic B-Cell, CD4 + and CD8 + T-Cell Epitopes, that are Highly Conserved Between Human and Animal Coronaviruses, Identified from SARS-CoV-2 as Immune Targets for Pre-Emptive Pan-Coronavirus Vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33024971 DOI: 10.1101/2020.09.27.316018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Over the last two decades, there have been three deadly human outbreaks of Coronaviruses (CoVs) caused by emerging zoonotic CoVs: SARS-CoV, MERS-CoV, and the latest highly transmissible and deadly SARS-CoV-2, which has caused the current COVID-19 global pandemic. All three deadly CoVs originated from bats, the natural hosts, and transmitted to humans via various intermediate animal reservoirs. Because there is currently no universal pan-Coronavirus vaccine available, two worst-case scenarios remain highly possible: (1) SARS-CoV-2 mutates and transforms into a seasonal "flu-like" global pandemic; and/or (2) Other global COVID-like pandemics will emerge in the coming years, caused by yet another spillover of an unknown zoonotic bat-derived SARS-like Coronavirus (SL-CoV) into an unvaccinated human population. Determining the antigen and epitope landscapes that are conserved among human and animal Coronaviruses as well as the repertoire, phenotype and function of B cells and CD4 + and CD8 + T cells that correlate with resistance seen in asymptomatic COVID-19 patients should inform in the development of pan-Coronavirus vaccines 1 . In the present study, using several immuno-informatics and sequence alignment approaches, we identified several human B-cell, CD4 + and CD8 + T cell epitopes that are highly conserved in: ( i ) greater than 81,000 SARS-CoV-2 human strains identified to date in 190 countries on six continents; ( ii ) six circulating CoVs that caused previous human outbreaks of the "Common Cold"; ( iii ) five SL-CoVs isolated from bats; ( iv ) five SL-CoV isolated from pangolins; ( v ) three SL-CoVs isolated from Civet Cats; and ( vi ) four MERS strains isolated from camels. Furthermore, we identified cross-reactive asymptomatic epitopes that: ( i ) recalled B cell, CD4 + and CD8 + T cell responses from both asymptomatic COVID-19 patients and healthy individuals who were never exposed to SARS-CoV-2; and ( ii ) induced strong B cell and T cell responses in "humanized" Human Leukocyte Antigen (HLA)-DR/HLA-A*02:01 double transgenic mice. The findings herein pave the way to develop a pre-emptive multi-epitope pan-Coronavirus vaccine to protect against past, current, and potential future outbreaks.
Collapse
|
25
|
Wessolly M, Stephan-Falkenau S, Streubel A, Werner R, Borchert S, Griff S, Mairinger E, Walter RFH, Bauer T, Eberhardt WEE, Blum TG, Schmid KW, Kollmeier J, Mairinger T, Mairinger FD. A Novel Epitope Quality-Based Immune Escape Mechanism Reveals Patient's Suitability for Immune Checkpoint Inhibition. Cancer Manag Res 2020; 12:7881-7890. [PMID: 32922086 PMCID: PMC7457781 DOI: 10.2147/cmar.s258396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/16/2020] [Indexed: 12/31/2022] Open
Abstract
Background Immune checkpoint inhibition, especially the blockade of PD-1 and PD-L1, has become one of the most thriving therapeutic approaches in modern oncology. Immune evasion caused by altered tumor epitope processing (so-called processing escapes) may be one way to explain immune checkpoint inhibition therapy failure. In the present study, we aim to demonstrate the effects of processing escapes on immunotherapy outcome in NSCLC patients. Patients and Methods Whole exome sequencing data of 400 NSCLC patients (AdC and SCC) were extracted from the TCGA database. The ICB cohort was composed of primary tumor probes from 48 NSCLC patients treated with nivolumab. Mutations were identified by targeted amplicon-based sequencing including hotspots and whole exomes of 22 genes. The effect of mutations on proteasomal processing was evaluated by deep learning methods previously trained on 1260 known MHC-I ligands. Cox regression modelling was used to determine the influence on overall survival. Results In the TCGA cohort, processing escapes were associated with decreased overall survival (p= 0.0140). In the ICB cohort, patients showing processing escapes in combination with high levels of PD-L1 (n=8/48) also showed significantly decreased overall survival, independently of mutational load or PD-L1 status. Conclusion The concept of altered epitope processing may help to understand immunotherapy failure. Especially when combined with PD-L1 status, this method can be used as a biomarker to identify patients not suitable for immunotherapy.
Collapse
Affiliation(s)
- Michael Wessolly
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | | | - Anna Streubel
- Department of Tissue Diagnostics, Helios Klinikum Emil Von Behring, Berlin, Germany
| | - Robert Werner
- Department of Tissue Diagnostics, Helios Klinikum Emil Von Behring, Berlin, Germany
| | - Sabrina Borchert
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Sergej Griff
- Department of Tissue Diagnostics, Helios Klinikum Emil Von Behring, Berlin, Germany
| | - Elena Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert F H Walter
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Ruhrlandklinik, West German Lung Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Torsten Bauer
- Lungenklinik Heckeshorn, Helios Klinikum Emil Von Behring, Berlin, Germany
| | - Wilfried E E Eberhardt
- Ruhrlandklinik, West German Lung Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Department of Medical Oncology, West German Cancer Center, University, Hospital Essen, Essen, Germany
| | - Torsten G Blum
- Lungenklinik Heckeshorn, Helios Klinikum Emil Von Behring, Berlin, Germany
| | - Kurt W Schmid
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jens Kollmeier
- Lungenklinik Heckeshorn, Helios Klinikum Emil Von Behring, Berlin, Germany
| | - Thomas Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Fabian D Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
26
|
Kumar A, Suryadevara NC, Wolf KJ, Wilson JT, Di Paolo RJ, Brien JD, Joyce S. Heterotypic immunity against vaccinia virus in an HLA-B*07:02 transgenic mousepox infection model. Sci Rep 2020; 10:13167. [PMID: 32759969 PMCID: PMC7406653 DOI: 10.1038/s41598-020-69897-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
Vaccination with vaccinia virus (VACV) elicits heterotypic immunity to smallpox, monkeypox, and mousepox, the mechanistic basis for which is poorly understood. It is generally assumed that heterotypic immunity arises from the presentation of a wide array of VACV-derived, CD8+ T cell epitopes that share homology with other poxviruses. Herein this assumption was tested using a large panel of VACV-derived peptides presented by HLA-B*07:02 (B7.2) molecules in a mousepox/ectromelia virus (ECTV)-infection, B7.2 transgenic mouse model. Most dominant epitopes recognized by ECTV- and VACV-reactive CD8+ T cells overlapped significantly without altering immunodominance hierarchy. Further, several epitopes recognized by ECTV-reactive CD8+ T cells were not recognized by VACV-reactive CD8+ T cells, and vice versa. In one instance, the lack of recognition owed to a N72K variation in the ECTV C4R70-78 variant of the dominant VACV B8R70-78 epitope. C4R70-78 does not bind to B7.2 and, hence, it was neither immunogenic nor antigenic. These findings provide a mechanistic basis for VACV vaccination-induced heterotypic immunity which can protect against Variola and Monkeypox disease. The understanding of how cross-reactive responses develop is essential for the rational design of a subunit-based vaccine that would be safe, and effectively protect against heterologous infection.
Collapse
Affiliation(s)
- Amrendra Kumar
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Naveen Chandra Suryadevara
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Kyle J Wolf
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Richard J Di Paolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Sebastian Joyce
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Centre, Nashville, TN, USA.
| |
Collapse
|
27
|
Kardani K, Hashemi A, Bolhassani A. Comparative analysis of two HIV-1 multiepitope polypeptides for stimulation of immune responses in BALB/c mice. Mol Immunol 2020; 119:106-122. [DOI: 10.1016/j.molimm.2020.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/20/2022]
|
28
|
Satapornpong P, Jinda P, Jantararoungtong T, Koomdee N, Chaichan C, Pratoomwun J, Na Nakorn C, Aekplakorn W, Wilantho A, Ngamphiw C, Tongsima S, Sukasem C. Genetic Diversity of HLA Class I and Class II Alleles in Thai Populations: Contribution to Genotype-Guided Therapeutics. Front Pharmacol 2020; 11:78. [PMID: 32180714 PMCID: PMC7057685 DOI: 10.3389/fphar.2020.00078] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Human leukocyte antigen (HLA) class I and II are known to have association with severe cutaneous adverse reactions (SCARs) when exposing to certain drug treatment. Due to genetic differences at population level, drug hypersensitivity reactions are varied, and thus common pharmacogenetics markers for one country might be different from another country, for instance, HLA-A*31:01 is associated with carbamazepine (CBZ)-induced SCARs in European and Japanese while HLA-B*15:02 is associated with CBZ-induced Stevens–Johnson syndrome/toxic epidermal necrolysis (SJS/TEN) among Taiwanese and Southeast Asian. Such differences pose a major challenge to prevent drug hypersensitivity when pharmacogenetics cannot be ubiquitously and efficiently translated into clinic. Therefore, a population-wide study of the distribution of HLA-pharmacogenetics markers is needed. This work presents a study of Thai HLA alleles on both HLA class I and II genes from 470 unrelated Thai individuals by means of polymerase chain reaction sequence-specific oligonucleotide (PCR-SSO) in which oligonucleotide probes along the stretches of HLA-A, -B, -C, -DRB1, -DQA1, and -DQB1 genes were genotyped. These 470 individuals were selected according to their regional locations, which were from North, Northeast, South, Central, and a capital city, Bangkok. Top ranked HLA alleles in Thai population include HLA-A*11:01 (26.06%), -B*46:01 (14.04%), -C* 01:02 (17.13%), -DRB1*12:02 (15.32%), -DQA1*01:01 (24.89%), and -DQB1*05:02 (21.28%). The results revealed that the distribution of HLA-pharmacogenetics alleles from the South had more HLA-B75 family that a typical HLA-B*15:02 pharmacogenetics test for SJS/TEN screening would not cover. Besides the view across the nation, when compared HLA alleles from Thai population with HLA alleles from both European and Asian countries, the distribution landscape of HLA-associated drug hypersensitivity across many countries could be observed. Consequently, this pharmacogenetics database offers a comprehensive view of pharmacogenetics marker distribution in Thailand that could be used as a reference for other Southeast Asian countries to validate the feasibility of their future pharmacogenetics deployment.
Collapse
Affiliation(s)
- Patompong Satapornpong
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand.,Division of General Pharmacy Practice, Department of Pharmaceutical Care, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Pimonpan Jinda
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Thawinee Jantararoungtong
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Napatrupron Koomdee
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Chonlawat Chaichan
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Jirawat Pratoomwun
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Chalitpon Na Nakorn
- Graduate Program in Translational Medicine, Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wichai Aekplakorn
- Department of Community Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Alisa Wilantho
- National Biobank of Thailand, National Science and Technology Development Agency, Pathum Thani, Thailand.,National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Chumpol Ngamphiw
- National Biobank of Thailand, National Science and Technology Development Agency, Pathum Thani, Thailand.,National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sissades Tongsima
- National Biobank of Thailand, National Science and Technology Development Agency, Pathum Thani, Thailand.,National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand.,The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group, Bangkok, Thailand
| |
Collapse
|
29
|
Sarkizova S, Klaeger S, Le PM, Li LW, Oliveira G, Keshishian H, Hartigan CR, Zhang W, Braun DA, Ligon KL, Bachireddy P, Zervantonakis IK, Rosenbluth JM, Ouspenskaia T, Law T, Justesen S, Stevens J, Lane WJ, Eisenhaure T, Lan Zhang G, Clauser KR, Hacohen N, Carr SA, Wu CJ, Keskin DB. A large peptidome dataset improves HLA class I epitope prediction across most of the human population. Nat Biotechnol 2020; 38:199-209. [PMID: 31844290 PMCID: PMC7008090 DOI: 10.1038/s41587-019-0322-9] [Citation(s) in RCA: 317] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022]
Abstract
Prediction of HLA epitopes is important for the development of cancer immunotherapies and vaccines. However, current prediction algorithms have limited predictive power, in part because they were not trained on high-quality epitope datasets covering a broad range of HLA alleles. To enable prediction of endogenous HLA class I-associated peptides across a large fraction of the human population, we used mass spectrometry to profile >185,000 peptides eluted from 95 HLA-A, -B, -C and -G mono-allelic cell lines. We identified canonical peptide motifs per HLA allele, unique and shared binding submotifs across alleles and distinct motifs associated with different peptide lengths. By integrating these data with transcript abundance and peptide processing, we developed HLAthena, providing allele-and-length-specific and pan-allele-pan-length prediction models for endogenous peptide presentation. These models predicted endogenous HLA class I-associated ligands with 1.5-fold improvement in positive predictive value compared with existing tools and correctly identified >75% of HLA-bound peptides that were observed experimentally in 11 patient-derived tumor cell lines.
Collapse
Affiliation(s)
- Siranush Sarkizova
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Susan Klaeger
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Phuong M Le
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Letitia W Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Wandi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David A Braun
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Keith L Ligon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Patient Derived Models, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Neuropathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Pavan Bachireddy
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | - Travis Law
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jonathan Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - William J Lane
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Guang Lan Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
| | | | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Center for Cancer Immunology, Massachusetts General Hospital, Boston, MA, USA.
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Catherine J Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Derin B Keskin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA.
| |
Collapse
|
30
|
Vlček J, Štefka J. Association between louse abundance and MHC II supertypes in Galápagos mockingbirds. Parasitol Res 2020; 119:1597-1605. [PMID: 32006226 DOI: 10.1007/s00436-020-06617-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/22/2020] [Indexed: 01/30/2023]
Abstract
Major histocompatibility complex class II (MHC II) is an essential molecule triggering the adaptive immune response by the presentation of pathogens to helper T cells. The association between individual MHC II variants and various parasites has become a frequent finding in studies of vertebrate populations. However, although bird ectoparasites have a significant effect on their host's fitness, and the host's immune system can regulate ectoparasitic infections, no study has yet investigated the association between MHC II polymorphism and ectoparasite infection in the populations of free-living birds. Here, we test whether an association exists between the abundance of a chewing louse (Myrsidea nesomimi) and MHC II polymorphism of its hosts, the Galápagos mockingbirds (Mimus). We have found that the presence of two MHC II supertypes (functionally differentiated clusters) was significantly associated with louse abundance. This pattern supports the theory that a co-evolutionary interaction stands behind the maintenance of MHC polymorphism. Moreover, we have found a positive correlation between louse abundance and heterophil/lymphocyte ratio (an indicator of immunological stress) that serves as an additional piece of evidence that ectoparasite burden is affected by immunological state of Galápagos mockingbirds.
Collapse
Affiliation(s)
- Jakub Vlček
- Institute of Parasitology, Biology Centre CAS, Branišovská 1160/31, 37005, České Budějovice, Czech Republic. .,Faculty of Science, University of South Bohemia, Branišovská, 1760, České Budějovice, Czech Republic.
| | - Jan Štefka
- Institute of Parasitology, Biology Centre CAS, Branišovská 1160/31, 37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská, 1760, České Budějovice, Czech Republic
| |
Collapse
|
31
|
Rekdal SL, Anmarkrud JA, Lifjeld JT, Johnsen A. Extra‐pair mating in a passerine bird with highly duplicated major histocompatibility complex class II: Preference for the golden mean. Mol Ecol 2019; 28:5133-5144. [DOI: 10.1111/mec.15273] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 12/26/2022]
Affiliation(s)
| | | | | | - Arild Johnsen
- Natural History Museum University of Oslo Oslo Norway
| |
Collapse
|
32
|
In silico Designed Ebola Virus T-Cell Multi-Epitope DNA Vaccine Constructions Are Immunogenic in Mice. Vaccines (Basel) 2019; 7:vaccines7020034. [PMID: 30934980 PMCID: PMC6630745 DOI: 10.3390/vaccines7020034] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 11/16/2022] Open
Abstract
Background: The lack of effective vaccines against Ebola virus initiates a search for new approaches to overcoming this problem. The aim of the study was to design artificial polyepitope T-cell immunogens⁻⁻candidate DNA vaccines against Ebola virus and to evaluate their capacity to induce a specific immune response in a laboratory animal model. Method: Design of two artificial polyepitope T-cell immunogens, one of which (EV.CTL) includes cytotoxic and the other (EV.Th)⁻⁻T-helper epitopes of Ebola virus proteins was carried out using original TEpredict/PolyCTLDesigner software. Synthesized genes were cloned in pcDNA3.1 plasmid vector. Target gene expression was estimated by synthesis of specific mRNAs and proteins in cells transfected with recombinant plasmids. Immunogenicity of obtained DNA vaccine constructs was evaluated according to their capacity to induce T-cell response in BALB/c mice using IFNγ ELISpot and ICS. Results: We show that recombinant plasmids pEV.CTL and pEV.Th encoding artificial antigens provide synthesis of corresponding mRNAs and proteins in transfected cells, as well as induce specific responses both to CD4+ and CD8+ T-lymphocytes in immunized animals. Conclusions: The obtained recombinant plasmids can be regarded as promising DNA vaccine candidates in future studies of their capacity to induce cytotoxic and protective responses against Ebola virus.
Collapse
|
33
|
Heide J, Vaughan KC, Sette A, Jacobs T, Schulze Zur Wiesch J. Comprehensive Review of Human Plasmodium falciparum-Specific CD8+ T Cell Epitopes. Front Immunol 2019; 10:397. [PMID: 30949162 PMCID: PMC6438266 DOI: 10.3389/fimmu.2019.00397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
Control of malaria is an important global health issue and there is still an urgent need for the development of an effective prophylactic vaccine. Multiple studies have provided strong evidence that Plasmodium falciparum-specific MHC class I-restricted CD8+ T cells are important for sterile protection against Plasmodium falciparum infection. Here, we present an interactive epitope map of all P. falciparum-specific CD8+ T cell epitopes published to date, based on a comprehensive data base (IEDB), and literature search. The majority of the described P. falciparum-specific CD8+ T cells were directed against the antigens CSP, TRAP, AMA1, and LSA1. Notably, most of the epitopes were discovered in vaccine trials conducted with malaria-naïve volunteers. Only few immunological studies of P. falciparum-specific CD8+ T cell epitopes detected in patients suffering from acute malaria or in people living in malaria endemic areas have been published. Further detailed immunological mappings of P. falciparum-specific epitopes of a broader range of P. falciparum proteins in different settings and with different disease status are needed to gain a more comprehensive understanding of the role of CD8+ T cell responses for protection, and to better guide vaccine design and to study their efficacy.
Collapse
Affiliation(s)
- Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Kerrie C Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute of Tropical Medicine, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
34
|
Brown SD, Holt RA. Neoantigen characteristics in the context of the complete predicted MHC class I self-immunopeptidome. Oncoimmunology 2018; 8:1556080. [PMID: 30723589 PMCID: PMC6350689 DOI: 10.1080/2162402x.2018.1556080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 01/21/2023] Open
Abstract
The self-immunopeptidome is the repertoire of all self-peptides that can be presented by the combination of MHC variants carried by an individual, defined by their HLA genotype. Each MHC variant presents a distinct set of self-peptides, and the number of peptides in a set is variable. Subjects carrying MHC variants that present fewer self-peptides should also present fewer mutated peptides, resulting in decreased immune pressure on tumor cells. To explore this, we predicted peptide-MHC binding values using all unique 8-11mer human peptides in the human proteome and all available HLA class I allelic variants, for a total of 134 billion unique peptide--MHC binding predictions. From these predictions, we observe that most peptides are able to be presented by relatively few (< 250) MHC, while some can be presented by upwards of 1,500 different MHC. There is substantial overlap among the repertoires of peptides presented by different MHC and no relationship between the number of peptides presented and HLA population frequency. Nearly 30% of self-peptides are presentable by at least one MHC, leaving 70% of the human peptidome unsurveyed by T cells. We observed similar distributions of predicted self-immunopeptidome sizes in cancer subjects compared to controls, and within the pan-cancer population, predicted self-immunopeptidome size combined with mutational load to predict survival. Self-immunopeptidome analysis revealed evidence for tumor immunoediting and identified specific peptide positions that most influence immunogenicity. Because self-immunopeptidome size is defined by HLA genotypes and approximates neoantigen load, HLA genotyping could offer a rapid predictive biomarker for response to immunotherapy.
Collapse
Affiliation(s)
- Scott D Brown
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.,Genome Science and Technology Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert A Holt
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.,Genome Science and Technology Program, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
35
|
Tian Y, da Silva Antunes R, Sidney J, Lindestam Arlehamn CS, Grifoni A, Dhanda SK, Paul S, Peters B, Weiskopf D, Sette A. A Review on T Cell Epitopes Identified Using Prediction and Cell-Mediated Immune Models for Mycobacterium tuberculosis and Bordetella pertussis. Front Immunol 2018; 9:2778. [PMID: 30555469 PMCID: PMC6281829 DOI: 10.3389/fimmu.2018.02778] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/12/2018] [Indexed: 01/01/2023] Open
Abstract
In the present review, we summarize work from our as well as other groups related to the characterization of bacterial T cell epitopes, with a specific focus on two important pathogens, namely, Mycobacterium tuberculosis (Mtb), the bacterium that causes tuberculosis (TB), and Bordetella pertussis (BP), the bacterium that causes whooping cough. Both bacteria and their associated diseases are of large societal significance. Although vaccines exist for both pathogens, their efficacy is incomplete. It is widely thought that defects and/or alteration in T cell compartments are associated with limited vaccine effectiveness. As discussed below, a full genome-wide map was performed in the case of Mtb. For BP, our focus has thus far been on the antigens contained in the acellular vaccine; a full genome-wide screen is in the planning stage. Nevertheless, the sum-total of the results in the two different bacterial systems allows us to exemplify approaches and techniques that we believe are generally applicable to the mapping and characterization of human immune responses to bacterial pathogens. Finally, we add, as a disclaimer, that this review by design is focused on the work produced by our laboratory as an illustration of approaches to the study of T cell responses to Mtb and BP, and is not meant to be comprehensive, nor to detract from the excellent work performed by many other groups.
Collapse
Affiliation(s)
- Yuan Tian
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Sandeep Kumar Dhanda
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Sinu Paul
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
36
|
Coppola M, Ottenhoff TH. Genome wide approaches discover novel Mycobacterium tuberculosis antigens as correlates of infection, disease, immunity and targets for vaccination. Semin Immunol 2018; 39:88-101. [PMID: 30327124 DOI: 10.1016/j.smim.2018.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 01/15/2023]
Abstract
Every day approximately six thousand people die of Tuberculosis (TB). Its causative agent, Mycobacterium tuberculosis (Mtb), is an ancient pathogen that through its evolution developed complex mechanisms to evade immune surveillance and acquire the ability to establish persistent infection in its hosts. Currently, it is estimated that one-fourth of the human population is latently infected with Mtb and among those infected 3-10% are at risk of developing active TB disease during their lifetime. The currently available diagnostics are not able to detect this risk group for prophylactic treatment to prevent transmission. Anti-TB drugs are available but only as long regimens with considerable side effects, which could both be reduced if adequate tests were available to monitor the response of TB to treatment. New vaccines are also urgently needed to substitute or boost Bacille Calmette-Guérin (BCG), the only approved TB vaccine: although BCG prevents disseminated TB in infants, it fails to impact the incidence of pulmonary TB in adults, and therefore has little effect on TB transmission. To achieve TB eradication, the discovery of Mtb antigens that effectively correlate with the human response to infection, with the curative host response following TB treatment, and with natural as well as vaccine induced protection will be critical. Over the last decade, many new Mtb antigens have been found and proposed as TB biomarkers and vaccine candidates, but only a very small number of these is being used in commercial diagnostic tests or is being assessed as candidate TB vaccine antigens in human clinical trials, aiming to prevent infection, disease or disease recurrence following treatment. Most of these antigens were discovered decades ago, before the complete Mtb genome sequence became available, and thus did not harness the latest insights from post-genomic antigen discovery strategies and genome wide approaches. These have, for example, revealed critical phase variation in Mtb replication and accompanying gene -and therefore antigen- expression patterns. In this review, we present a brief overview of past methodologies, and subsequently focus on the most important recent Mtb antigen discovery studies which have mined the Mtb antigenome through "unbiased" genome wide approaches. We compare the results for these approaches -as far as we know for the first time-, highlight Mtb antigens that have been identified independently by different strategies and present a comprehensive overview of the Mtb antigens thus discovered.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Dept. Infectious Diseases, LUMC, PO Box 9600, 2300RC Leiden, The Netherlands.
| | - Tom Hm Ottenhoff
- Dept. Infectious Diseases, LUMC, PO Box 9600, 2300RC Leiden, The Netherlands
| |
Collapse
|
37
|
Chew WL. Immunity to CRISPR Cas9 and Cas12a therapeutics. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10. [PMID: 29083112 DOI: 10.1002/wsbm.1408] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 12/27/2022]
Abstract
Genome-editing therapeutics are poised to treat human diseases. As we enter clinical trials with the most promising CRISPR-Cas9 and CRISPR-Cas12a (Cpf1) modalities, the risks associated with administering these foreign biomolecules into human patients become increasingly salient. Preclinical discovery with CRISPR-Cas9 and CRISPR-Cas12a systems and foundational gene therapy studies indicate that the host immune system can mount undesired responses against the administered proteins and nucleic acids, the gene-edited cells, and the host itself. These host defenses include inflammation via activation of innate immunity, antibody induction in humoral immunity, and cell death by T-cell-mediated cytotoxicity. If left unchecked, these immunological reactions can curtail therapeutic benefits and potentially lead to mortality. Ways to assay and reduce the immunogenicity of Cas9 and Cas12a proteins are therefore critical for ensuring patient safety and treatment efficacy, and for bringing us closer to realizing the vision of permanent genetic cures. WIREs Syst Biol Med 2018, 10:e1408. doi: 10.1002/wsbm.1408 This article is categorized under: Laboratory Methods and Technologies > Genetic/Genomic Methods Translational, Genomic, and Systems Medicine > Translational Medicine Translational, Genomic, and Systems Medicine > Therapeutic Methods.
Collapse
Affiliation(s)
- Wei Leong Chew
- Synthetic Biology, Genome Institute of Singapore, Singapore, Singapore
| |
Collapse
|
38
|
Fundamentals and Methods for T- and B-Cell Epitope Prediction. J Immunol Res 2017; 2017:2680160. [PMID: 29445754 PMCID: PMC5763123 DOI: 10.1155/2017/2680160] [Citation(s) in RCA: 354] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 12/25/2022] Open
Abstract
Adaptive immunity is mediated by T- and B-cells, which are immune cells capable of developing pathogen-specific memory that confers immunological protection. Memory and effector functions of B- and T-cells are predicated on the recognition through specialized receptors of specific targets (antigens) in pathogens. More specifically, B- and T-cells recognize portions within their cognate antigens known as epitopes. There is great interest in identifying epitopes in antigens for a number of practical reasons, including understanding disease etiology, immune monitoring, developing diagnosis assays, and designing epitope-based vaccines. Epitope identification is costly and time-consuming as it requires experimental screening of large arrays of potential epitope candidates. Fortunately, researchers have developed in silico prediction methods that dramatically reduce the burden associated with epitope mapping by decreasing the list of potential epitope candidates for experimental testing. Here, we analyze aspects of antigen recognition by T- and B-cells that are relevant for epitope prediction. Subsequently, we provide a systematic and inclusive review of the most relevant B- and T-cell epitope prediction methods and tools, paying particular attention to their foundations.
Collapse
|
39
|
Hegde NR, Gauthami S, Sampath Kumar HM, Bayry J. The use of databases, data mining and immunoinformatics in vaccinology: where are we? Expert Opin Drug Discov 2017; 13:117-130. [PMID: 29226722 DOI: 10.1080/17460441.2018.1413088] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Vaccinology has evolved from a sub-discipline focussed on simplistic vaccine development based on antibody-mediated protection to a separate discipline involving epidemiology, host and pathogen biology, immunology, genomics, proteomics, structure biology, protein engineering, chemical biology, and delivery systems. Data mining in combination with bioinformatics has provided a scaffold linking all these disciplines to the design of vaccines and vaccine adjuvants. Areas covered: This review provides background knowledge on immunological aspects which have been exploited with informatics for the in silico analysis of immune responses and the design of vaccine antigens. Furthermore, the article presents various databases and bioinformatics tools, and discusses B and T cell epitope predictions, antigen design, adjuvant research and systems immunology, highlighting some important examples, and challenges for the future. Expert opinion: Informatics and data mining have not only reduced the time required for experimental immunology, but also contributed to the identification and design of novel vaccine candidates and the determination of biomarkers and pathways of vaccine response. However, more experimental data is required for benchmarking immunoinformatic tools. Nevertheless, developments in immunoinformatics and reverse vaccinology, which are nascent fields, are likely to hasten vaccine discovery, although the path to regulatory approval is likely to remain a necessary impediment.
Collapse
Affiliation(s)
| | - S Gauthami
- b Ella Foundation, Turkapally , Hyderabad , India
| | - H M Sampath Kumar
- c Council of Scientific and Industrial Research - Indian Institute of Chemical Technology , Hyderabad , India
| | - Jagadeesh Bayry
- d Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1138 , Centre de Recherche des Cordeliers, Paris , France
| |
Collapse
|
40
|
Kalyanaraman N. In silico prediction of potential vaccine candidates on capsid protein of human bocavirus 1. Mol Immunol 2017; 93:193-205. [PMID: 29207326 DOI: 10.1016/j.molimm.2017.11.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/18/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Human bocavirus 1 (HBoV1) is a newly identified parvovirus that causes serious respiratory infection among children across the globe. Aim of the present study was to predict immunogenic residues located on the VP2 protein of HBoV1 towards development of epitope based vaccines. Several computational tools were employed to predict epitopes (bothT and B cell restricted) with stringent regulation for the improvement of confidence. After meticulous analysis, the peptide "TTPWTYFNFNQY" was identified as potential candidate for development of preventive vaccine. Of note, the epitope "TTPWTYFNFNQY" was found to be recognized by fifteen different alleles belonging to seven HLA supertypes (A1, A3, A24, A26, B7, B58 and B62). Further, mutational variability analysis pointed that most of the amino acids were well conserved. Docking scores obtained from ClusPro and Autodock Vina for selected epitopes displayed energetically favorable and stable interaction of peptide-HLA-I complexes. The core peptide "LLYQMPFFL" was found to recognize by wide range of HLA class II allele recognition thereby qualified as candidate for therapeutic vaccine. Five distinct linear peptides (withT cell epitope superimposition) belonging to B cells were identified in the VP2 protein. Further attention on the enlisted epitopes may shed light on the path for development of diagnostic, therapeutic and preventive tools against HBoV1 infection. Additionally, the predicted epitopes may help us to address the original antigenic sin phenomena observed during consecutive HBoV2-4 infection.
Collapse
Affiliation(s)
- Narayanan Kalyanaraman
- Viral Research Diagnostic Laboratory (VRDL), Department of Microbiology, Govt Theni Medical College, Theni, Tamil Nadu, India.
| |
Collapse
|
41
|
Oyarzún P, Kobe B. Recombinant and epitope-based vaccines on the road to the market and implications for vaccine design and production. Hum Vaccin Immunother 2017; 12:763-7. [PMID: 26430814 DOI: 10.1080/21645515.2015.1094595] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Novel vaccination approaches based on rational design of B- and T-cell epitopes - epitope-based vaccines - are making progress in the clinical trial pipeline. The epitope-focused recombinant protein-based malaria vaccine (termed RTS,S) is a next-generation approach that successfully reached phase-III trials, and will potentially become the first commercial vaccine against a human parasitic disease. Progress made on methods such as recombinant DNA technology, advanced cell-culture techniques, immunoinformatics and rational design of immunogens are driving the development of these novel concepts. Synthetic recombinant proteins comprising both B- and T-cell epitopes can be efficiently produced through modern biotechnology and bioprocessing methods, and can enable the induction of large repertoires of immune specificities. In particular, the inclusion of appropriate CD4+ T-cell epitopes is increasingly considered a key vaccine component to elicit robust immune responses, as suggested by results coming from HIV-1 clinical trials. In silico strategies for vaccine design are under active development to address genetic variation in pathogens and several broadly protective "universal" influenza and HIV-1 vaccines are currently at different stages of clinical trials. Other methods focus on improving population coverage in target populations by rationally considering specificity and prevalence of the HLA proteins, though a proof-of-concept in humans has not been demonstrated yet. Overall, we expect immunoinformatics and bioprocessing methods to become a central part of the next-generation epitope-based vaccine development and production process.
Collapse
Affiliation(s)
- Patricio Oyarzún
- a Biotechnology Center, Facultad de Ingeniería y Tecnología, Universidad San Sebastián , Concepción , Chile
| | - Bostjan Kobe
- b School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Center, University of Queensland , Brisbane , Australia
| |
Collapse
|
42
|
Sankar S, Ramamurthy M, Nandagopal B, Sridharan G. Short peptide epitope design from hantaviruses causing HFRS. Bioinformation 2017; 13:231-236. [PMID: 28943728 PMCID: PMC5602290 DOI: 10.6026/97320630013231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Several genotypes of the hantavirus cause hemorrhagic fever with renal syndrome (HFRS) and is an important public health problem
worldwide. There is now growing interest to develop subunit vaccines especially focused to elicit cytotoxic T lymphocyte responses
which are important against viral infection. We identified candidate T-cell epitopes that bind to Class I HLA supertypes towards
identifying potential subunit vaccine entity. These epitopes are conserved in all 5 hantavirus genotypes of HFRS (Hantaan, Dobrava-
Belgrade, Seoul, Gou virus and Amur). The epitopes identified from S and M segment genomes were analyzed for human proteasome
cleavage, transporter associated antigen processing (TAP) efficiency and antigenicity using bioinformatic approaches. The epitope
MRNTIMASK which had the two characteristics of high proteasomal cleavage score and TAP score, also had high antigenicity score.
Our results indicate that this epitope from the nucleocapsid protein may be considered the most favorable moiety for the development
of subunit peptide vaccine.
Collapse
Affiliation(s)
- Sathish Sankar
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore 632 055, Tamil Nadu, India
| | - Mageshbabu Ramamurthy
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore 632 055, Tamil Nadu, India
| | - Balaji Nandagopal
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore 632 055, Tamil Nadu, India
| | - Gopalan Sridharan
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore 632 055, Tamil Nadu, India
| |
Collapse
|
43
|
Rosendahl Huber SK, Luimstra JJ, van Beek J, Hoppes R, Jacobi RHJ, Hendriks M, Kapteijn K, Ouwerkerk C, Rodenko B, Ovaa H, de Jonge J. Chemical Modification of Influenza CD8+ T-Cell Epitopes Enhances Their Immunogenicity Regardless of Immunodominance. PLoS One 2016; 11:e0156462. [PMID: 27333291 PMCID: PMC4917206 DOI: 10.1371/journal.pone.0156462] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/13/2016] [Indexed: 11/19/2022] Open
Abstract
T cells are essential players in the defense against infection. By targeting the MHC class I antigen-presenting pathway with peptide-based vaccines, antigen-specific T cells can be induced. However, low immunogenicity of peptides poses a challenge. Here, we set out to increase immunogenicity of influenza-specific CD8+ T cell epitopes. By substituting amino acids in wild type sequences with non-proteogenic amino acids, affinity for MHC can be increased, which may ultimately enhance cytotoxic CD8+ T cell responses. Since preventive vaccines against viruses should induce a broad immune response, we used this method to optimize influenza-specific epitopes of varying dominance. For this purpose, HLA-A*0201 epitopes GILGFVFTL, FMYSDFHFI and NMLSTVLGV were selected in order of decreasing MHC-affinity and dominance. For all epitopes, we designed chemically enhanced altered peptide ligands (CPLs) that exhibited greater binding affinity than their WT counterparts; even binding scores of the high affinity GILGFVFTL epitope could be improved. When HLA-A*0201 transgenic mice were vaccinated with selected CPLs, at least 2 out of 4 CPLs of each epitope showed an increase in IFN-γ responses of splenocytes. Moreover, modification of the low affinity epitope NMLSTVLGV led to an increase in the number of mice that responded. By optimizing three additional influenza epitopes specific for HLA-A*0301, we show that this strategy can be extended to other alleles. Thus, enhancing binding affinity of peptides provides a valuable tool to improve the immunogenicity and range of preventive T cell-targeted peptide vaccines.
Collapse
Affiliation(s)
- Sietske K. Rosendahl Huber
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Jolien J. Luimstra
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Institute for Chemical Immunology (ICI), Utrecht, the Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Rieuwert Hoppes
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ronald H. J. Jacobi
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Marion Hendriks
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Kim Kapteijn
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Casper Ouwerkerk
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Boris Rodenko
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Huib Ovaa
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Institute for Chemical Immunology (ICI), Utrecht, the Netherlands
| | - Jørgen de Jonge
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- * E-mail:
| |
Collapse
|
44
|
Epitope prediction for MSP119 protein in Plasmodium yeolii using computational approaches. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s13721-016-0127-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
45
|
Xiang K, Ying G, Yan Z, Shanshan Y, Lei Z, Hongjun L, Maosheng S. Progress on adenovirus-vectored universal influenza vaccines. Hum Vaccin Immunother 2016; 11:1209-22. [PMID: 25876176 DOI: 10.1080/21645515.2015.1016674] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Influenza virus (IFV) infection causes serious health problems and heavy financial burdens each year worldwide. The classical inactivated influenza virus vaccine (IIVV) and live attenuated influenza vaccine (LAIV) must be updated regularly to match the new strains that evolve due to antigenic drift and antigenic shift. However, with the discovery of broadly neutralizing antibodies that recognize conserved antigens, and the CD8(+) T cell responses targeting viral internal proteins nucleoprotein (NP), matrix protein 1 (M1) and polymerase basic 1 (PB1), it is possible to develop a universal influenza vaccine based on the conserved hemagglutinin (HA) stem, NP, and matrix proteins. Recombinant adenovirus (rAd) is an ideal influenza vaccine vector because it has an ideal stability and safety profile, induces balanced humoral and cell-mediated immune responses due to activation of innate immunity, provides 'self-adjuvanting' activity, can mimic natural IFV infection, and confers seamless protection against mucosal pathogens. Moreover, this vector can be developed as a low-cost, rapid-response vaccine that can be quickly manufactured. Therefore, an adenovirus vector encoding conserved influenza antigens holds promise in the development of a universal influenza vaccine. This review will summarize the progress in adenovirus-vectored universal flu vaccines and discuss future novel approaches.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- APC, antigen-presenting cell
- Ad: adenovirus
- CAR, Coxsackie-Adenovirus Receptor
- CTLs, cytotoxic T lymphocytes
- DC, lung dendritic cells
- DVD, drug–vaccine duo
- FcγRs, Fc receptors for IgG
- HA, hemagglutinin
- HDAd, helper-dependent adenoviral
- HEK293, human embryonic kidney 293 cell
- HI, hemagglutination inhibition
- HLA, human leukocyte antigen
- IF-γ, interferon-γ
- IFV, Influenza virus
- IIVV, inactivated influenza virus vaccine
- IL-2, interleukin-2
- ITRs, inverted terminal repeats
- LAIV, live attenuated influenza vaccine
- M1, matrix protein 1
- M2, matrix protein 2
- MHC-I, major histocompatibility complex class I
- NA, neuraminidase
- NP, nucleoprotein
- RCA, replication competent adenovirus
- VAERD, vaccine-associated enhanced respiratory disease
- adenovirus vector
- broadly neutralizing antibodies
- cellular immunity
- flu, influenza
- hemagglutinin
- humoral immunity
- influenza
- mAbs, monoclonal antibodies
- mucosal immunity
- rAd, recombinant adenovirus
- universal vaccine
Collapse
Affiliation(s)
- Kui Xiang
- a Department of Molecular Biology; Institute of Medical Biology; Chinese Academy of Medical Sciences; Peking Union Medical College ; Kunming , Yunnan , PR China
| | | | | | | | | | | | | |
Collapse
|
46
|
Molero-Abraham M, Glutting JP, Flower DR, Lafuente EM, Reche PA. EPIPOX: Immunoinformatic Characterization of the Shared T-Cell Epitome between Variola Virus and Related Pathogenic Orthopoxviruses. J Immunol Res 2015; 2015:738020. [PMID: 26605344 PMCID: PMC4641182 DOI: 10.1155/2015/738020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/08/2015] [Accepted: 10/01/2015] [Indexed: 11/26/2022] Open
Abstract
Concerns that variola viruses might be used as bioweapons have renewed the interest in developing new and safer smallpox vaccines. Variola virus genomes are now widely available, allowing computational characterization of the entire T-cell epitome and the use of such information to develop safe and yet effective vaccines. To this end, we identified 124 proteins shared between various species of pathogenic orthopoxviruses including variola minor and major, monkeypox, cowpox, and vaccinia viruses, and we targeted them for T-cell epitope prediction. We recognized 8,106, and 8,483 unique class I and class II MHC-restricted T-cell epitopes that are shared by all mentioned orthopoxviruses. Subsequently, we developed an immunological resource, EPIPOX, upon the predicted T-cell epitome. EPIPOX is freely available online and it has been designed to facilitate reverse vaccinology. Thus, EPIPOX includes key epitope-focused protein annotations: time point expression, presence of leader and transmembrane signals, and known location on outer membrane structures of the infective viruses. These features can be used to select specific T-cell epitopes suitable for experimental validation restricted by single MHC alleles, as combinations thereof, or by MHC supertypes.
Collapse
Affiliation(s)
- Magdalena Molero-Abraham
- School of Medicine, Unit of Immunology, Complutense University of Madrid, Pza. Ramón y Cajal, s/n, 28040 Madrid, Spain
| | - John-Paul Glutting
- School of Medicine, Unit of Immunology, Complutense University of Madrid, Pza. Ramón y Cajal, s/n, 28040 Madrid, Spain
| | - Darren R. Flower
- School of Life and Health Sciences, University of Aston, Aston Triangle, Birmingham B4 7ET, UK
| | - Esther M. Lafuente
- School of Medicine, Unit of Immunology, Complutense University of Madrid, Pza. Ramón y Cajal, s/n, 28040 Madrid, Spain
| | - Pedro A. Reche
- School of Medicine, Unit of Immunology, Complutense University of Madrid, Pza. Ramón y Cajal, s/n, 28040 Madrid, Spain
| |
Collapse
|
47
|
Oyarzun P, Kobe B. Computer-aided design of T-cell epitope-based vaccines: addressing population coverage. Int J Immunogenet 2015. [PMID: 26211755 DOI: 10.1111/iji.12214] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Epitope-based vaccines (EVs) make use of short antigen-derived peptides corresponding to immune epitopes, which are administered to trigger a protective humoral and/or cellular immune response. EVs potentially allow for precise control over the immune response activation by focusing on the most relevant - immunogenic and conserved - antigen regions. Experimental screening of large sets of peptides is time-consuming and costly; therefore, in silico methods that facilitate T-cell epitope mapping of protein antigens are paramount for EV development. The prediction of T-cell epitopes focuses on the peptide presentation process by proteins encoded by the major histocompatibility complex (MHC). Because different MHCs have different specificities and T-cell epitope repertoires, individuals are likely to respond to a different set of peptides from a given pathogen in genetically heterogeneous human populations. In addition, protective immune responses are only expected if T-cell epitopes are restricted by MHC proteins expressed at high frequencies in the target population. Therefore, without careful consideration of the specificity and prevalence of the MHC proteins, EVs could fail to adequately cover the target population. This article reviews state-of-the-art algorithms and computational tools to guide EV design through all the stages of the process: epitope prediction, epitope selection and vaccine assembly, while optimizing vaccine immunogenicity and coping with genetic variation in humans and pathogens.
Collapse
Affiliation(s)
- P Oyarzun
- Biotechnology Centre, Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Concepción, Chile
| | - B Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
48
|
Singh SP, Verma V, Mishra BN. Characterization of Plasmodium falciparum Proteome at Asexual Blood Stages for Screening of Effective Vaccine Candidates: An Immunoinformatics Approach. ACTA ACUST UNITED AC 2015. [DOI: 10.4137/iii.s24755] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Malaria is a complex parasitic disease that is currently causing great concerns globally owing to the resistance to antimalarial drugs and lack of an effective vaccine. The present study involves the characterization of extracellular secretory proteins as vaccine candidates derived from proteome analysis of Plasmodium falciparum at asexual blood stages of malaria. Among the screened 32 proteins, 31 were predicted as antigens by the VaxiJen program, and 26 proteins had less than two transmembrane spanning regions predicted using the THMMM program. Moreover, 10 and 5 proteins were predicted to contain secretory signals by SignalP and TargetP, respectively. T-cell epitope prediction using MULTIPRED2 and NetCTL programs revealed that most of the predicted antigens are immunogenic and contain more than 10% supertype and 5% promiscuous epitopes of HLA-A, -B, or -DR. We anticipate that T-cell immune responses against asexual blood stages of Plasmodium are dispersed on a relatively large number of parasite antigens. This is the first report, to the best of our knowledge, offering new insights, at the proteome level, for the putative screening of effective vaccine candidates against the malaria pathogen. The findings also suggest new ways forward for the modern omics-guided vaccine target discovery using reverse vaccinology.
Collapse
Affiliation(s)
- Satarudra Prakash Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, India
| | - Vishal Verma
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, India
| | | |
Collapse
|
49
|
Castiblanco J, Anaya JM. Genetics and vaccines in the era of personalized medicine. Curr Genomics 2015; 16:47-59. [PMID: 25937813 PMCID: PMC4412964 DOI: 10.2174/1389202916666141223220551] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 12/17/2022] Open
Abstract
Vaccines represent the most successful and sustainable tactic to prevent and counteract infection. A vaccine generally improves immunity to a particular disease upon administration by inducing specific protective and efficient immune responses in all of the receiving population. The main known factors influencing the observed heterogeneity for immune re-sponses induced by vaccines are gender, age, co-morbidity, immune system, and genetic background. This review is mainly focused on the genetic status effect to vaccine immune responses and how this could contribute to the development of novel vaccine candidates that could be better directed and predicted relative to the genetic history of an individual and/or population. The text offers a brief history of vaccinology as a field, a description of the genetic status of the most relevant and studied genes and their functionality and correlation with exposure to specific vaccines; followed by an inside look into autoimmunity as a concern when designing vaccines as well as perspectives and conclusions looking towards an era of personalized and predictive vaccinology instead of a one size fits all approach.
Collapse
Affiliation(s)
- John Castiblanco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 #63-C-69, Bogota, Colombia ; Doctoral Program in Biomedical Sciences, Universidad del Rosario, Bogotá,Colombia
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 #63-C-69, Bogota, Colombia
| |
Collapse
|
50
|
Reguzova A, Antonets D, Karpenko L, Ilyichev A, Maksyutov R, Bazhan S. Design and evaluation of optimized artificial HIV-1 poly-T cell-epitope immunogens. PLoS One 2015; 10:e0116412. [PMID: 25786238 PMCID: PMC4364888 DOI: 10.1371/journal.pone.0116412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 12/09/2014] [Indexed: 11/18/2022] Open
Abstract
A successful HIV vaccine in addition to induction of antibody responses should elicit effective T cell responses. Here we described possible strategies for rational design of T-cell vaccine capable to induce high levels of both CD4+ and CD8+ T- cell responses. We developed artificial HIV-1 polyepitope T-cell immunogens based on the conserved natural CD8+ and CD4+ T cell epitopes from different HIV-1 strains and restricted by the most frequent major human leukocyte antigen (HLA) alleles. Designed immunogens contain optimized core polyepitope sequence and additional "signal" sequences which increase epitope processing and presentation to CD8+ and CD4+ T-lymphocytes: N-terminal ubiquitin, N-terminal signal peptide and C-terminal tyrosine motif of LAMP-1 protein. As a result we engineered three T cell immunogens - TCI-N, TCI-N2, and TCI-N3, with different combinations of signal sequences. All designed immunogens were able to elicit HIV-specific CD4+ and CD8+ T cell responses following immunization. Attachment of either ubiquitin or ER-signal/LAMP-1 sequences increased both CD4+ and CD8+ mediated HIV-specific T cell responses in comparison with polyepitope immunogen without any additional signal sequences. Moreover, TCI-N3 polyepitope immunogen with ubiquitin generated highest magnitude of HIV-specific CD4+ and CD8+ T cell responses in our study. Obtained data suggests that attachment of signal sequences targeting polyepitope immunogens to either MHC class I or MHC class II presentation pathways may improve immunogenicity of T-cell vaccines. These results support the strategy of the rational T cell immunogen design and contribute to the development of effective HIV-1 vaccine.
Collapse
Affiliation(s)
- Alena Reguzova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Denis Antonets
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Larisa Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Alexander Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Rinat Maksyutov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| | - Sergei Bazhan
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk region, 630559, Russia
| |
Collapse
|