1
|
Ahmed MZ, Rao T, Mutahir Z, Ahmed S, Ullah N, Ojha SC. Immunoinformatic-driven design and evaluation of multi-epitope mRNA vaccine targeting HIV-1 gp120. Front Immunol 2025; 16:1480025. [PMID: 40433366 PMCID: PMC12106336 DOI: 10.3389/fimmu.2025.1480025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
HIV (human immunodeficiency virus) presents a global health crisis, causing significant AIDS-related deaths and over one million new infections annually. The curbing of HIV is an intricate and continuously evolving domain, marked by numerous challenges, including drug resistance and the absence of a significant cure or vaccine because of its mutating ability and diverse antigens in its envelope, prompting research for functional cures and long-term remission strategies. The endeavor to devise an HIV vaccine capable of eliciting robust and broadly cross-reactive humoral and cellular immune responses is a formidable undertaking, primarily due to the pronounced genetic heterogeneity of HIV-1, the variances observed in virus subtypes (clades) across distinct geographic regions, and the polymorphic nature of human leukocyte antigens (HLA). The viral envelope protein (gp120) selectively interacts with CD4 and chemokine receptors on the surface of target cells. It serves as the key initiator in the intricate viral entry into host cells, rendering it a compelling candidate for vaccine development. This study used bioinformatic tools to design a safe, hypoallergenic, and non-toxic mRNA HIV-1 vaccine by assembling immunogenic B- and T-cell epitopes from the gp120 protein. We identified antigenic, non-toxic, and non-allergic B-cell epitopes (IEPLGIAPTRAKRRVVER) and T-cell epitopes (QQKVHALFY, ITIGPGQVF, WQGVGQAMY, APTRAKRRV, KQQKVHALFYRLDIV, QQKVHALFYRLDIVQ, QKVHALFYRLDIVQI, SLAEEEIIIRSENLT, and IRSENLTNNVKTIIV). For designing the mRNA vaccine against HIV-1 gp120, we assembled the epitopes with 5' m7G cap, 5' UTR (untranslated region), Kozak sequence, signal peptide (tPA), RpfE (resuscitation-promoting factor E) adjuvant at N-terminal and MITD (MHC class I trafficking domain) adjuvant, stop codon, 3' UTR, and 120-nucleotide long poly(A) tail at the C-terminal with immunogenic robustness linkers. The mRNA vaccine is translated into a protein-based vaccine by the host body's ribosomes. Their comprehensive computational findings, including physicochemical, structural, and 3D refinement analyses, substantiated the stability and quality of the translated vaccine. Molecular docking and simulation revealed a strong and stable binding affinity of vaccine immunization with immune cells' pattern recognition receptors (TLR4). Immune simulations demonstrated a potent primary immune response characterized by a gradual increase in immunoglobulins and a corresponding decline in antigen concentration. This bioinformatics-driven study presents a promising HIV-1 mRNA vaccine candidate, underscoring the need for further experimental validation through preclinical and clinical trials. At the same time, its methodologies hold the potential for addressing other challenging infectious diseases, thereby impacting vaccinology broadly.
Collapse
Affiliation(s)
- Muhammad Zeeshan Ahmed
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Tazeen Rao
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Zeeshan Mutahir
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Sarfraz Ahmed
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Najeeb Ullah
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Suvash Chandra Ojha
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Chih YC, Dietsch AC, Koopmann P, Ma X, Agardy DA, Zhao B, De Roia A, Kourtesakis A, Kilian M, Krämer C, Suwala AK, Stenzinger M, Boenig H, Blum A, Pienkowski VM, Aman K, Becker JP, Feldmann H, Bunse T, Harbottle R, Riemer AB, Liu HK, Etminan N, Sahm F, Ratliff M, Wick W, Platten M, Green EW, Bunse L. Vaccine-induced T cell receptor T cell therapy targeting a glioblastoma stemness antigen. Nat Commun 2025; 16:1262. [PMID: 39893177 PMCID: PMC11787355 DOI: 10.1038/s41467-025-56547-w] [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: 07/26/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
T cell receptor-engineered T cells (TCR-T) could be advantageous in glioblastoma by allowing safe and ubiquitous targeting of the glioblastoma-derived peptidome. Protein tyrosine phosphatase receptor type Z1 (PTPRZ1), is a clinically targetable glioblastoma antigen associated with glioblastoma cell stemness. Here, we identify a therapeutic HLA-A*02-restricted PTPRZ1-reactive TCR retrieved from a vaccinated glioblastoma patient. Single-cell sequencing of primary brain tumors shows PTPRZ1 overexpression in malignant cells, especially in glioblastoma stem cells (GSCs) and astrocyte-like cells. The validated vaccine-induced TCR recognizes the endogenously processed antigen without off-target cross-reactivity. PTPRZ1-specific TCR-T (PTPRZ1-TCR-T) kill target cells antigen-specifically, and in murine experimental brain tumors, their combined intravenous and intracerebroventricular administration is efficacious. PTPRZ1-TCR-T maintain stem cell memory phenotype in vitro and in vivo and lyse all examined HLA-A*02+ primary glioblastoma cell lines with a preference for GSCs and astrocyte-like cells. In summary, we demonstrate the proof of principle to employ TCR-T to treat glioblastoma.
Collapse
MESH Headings
- Glioblastoma/immunology
- Glioblastoma/therapy
- Glioblastoma/pathology
- Glioblastoma/genetics
- Humans
- Animals
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Mice
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Cancer Vaccines/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Antigens, Neoplasm/immunology
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/immunology
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
- HLA-A2 Antigen/immunology
- Immunotherapy, Adoptive/methods
- Xenograft Model Antitumor Assays
- Female
Collapse
Grants
- Swiss Cancer Foundation (Swiss Bridge Award), the Else Kröner Fresenius Foundation (2019_EKMS.49), the University Heidelberg Foundation (Hella Buühler Award), the DFG (German Research Foundation), project 404521405 (SFB1389 UNITE Glioblastoma B03), the DKFZ Hector institute (T-SIRE), the Hertie Foundation, the University of Heidelberg, ExploreTech! the DKTK Joint Funding AMI2GO, the Rolf Schwiete Foundation (2021-009), the HI-TRON strategy project PACESSETTING, the DKTK Joint Funding Program INNOVATION INVENT4GB.
- The DFG, project 404521405 (SFB1389 UNITE Glioblastoma B01) the DKTK Joint Funding AMI2GO, the Rolf Schwiete Foundation (2021-009), the HI-TRON strategy project PACESSETTING, the DKTK Joint Funding Program INNOVATION INVENT4GB.
Collapse
Affiliation(s)
- Yu-Chan Chih
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Amelie C Dietsch
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Philipp Koopmann
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Xiujian Ma
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Molecular Neurogenetics, DKFZ, DKFZ-ZMBH alliance, Heidelberg, Germany
| | - Dennis A Agardy
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Binghao Zhao
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Alice De Roia
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
- DNA Vector Laboratory, DKFZ, Heidelberg, Germany
| | - Alexandros Kourtesakis
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany
- CCU Neurooncology, DKFZ, Heidelberg, Germany
| | - Michael Kilian
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher Krämer
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Abigail K Suwala
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Institute for Pathology, Department of Neuropathology, Heidelberg University, Heidelberg, Germany
- CCU Neuropathology, DKFZ, Heidelberg, Germany
| | - Miriam Stenzinger
- Institute for Clinical Transfusion Medicine and Cell Therapy, Heidelberg, Germany
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Halvard Boenig
- Faculty of Medicine, Goethe University, Frankfurt a.M., Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Service Baden-Württemberg-Hessen, Frankfurt a.M., Frankfurt, Germany
| | | | | | - Kuralay Aman
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Jonas P Becker
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Immunotherapy and Immunoprevention, DKFZ, Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Henrike Feldmann
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Theresa Bunse
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Richard Harbottle
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- DNA Vector Laboratory, DKFZ, Heidelberg, Germany
| | - Angelika B Riemer
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Immunotherapy and Immunoprevention, DKFZ, Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Hai-Kun Liu
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Molecular Neurogenetics, DKFZ, DKFZ-ZMBH alliance, Heidelberg, Germany
| | - Nima Etminan
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - Felix Sahm
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Institute for Pathology, Department of Neuropathology, Heidelberg University, Heidelberg, Germany
- CCU Neuropathology, DKFZ, Heidelberg, Germany
| | - Miriam Ratliff
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- CCU Neurooncology, DKFZ, Heidelberg, Germany
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - Wolfgang Wick
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany
- CCU Neurooncology, DKFZ, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz) - A Helmholtz Institute of the DKFZ, Mainz, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Edward W Green
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Lukas Bunse
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| |
Collapse
|
3
|
Hare J, Nielsen M, Kiragga A, Ochiel D. Sustainable integration of artificial intelligence and machine learning approaches within the African infectious disease vaccine research and development ecosystem. Front Pharmacol 2024; 15:1499079. [PMID: 39741624 PMCID: PMC11685015 DOI: 10.3389/fphar.2024.1499079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025] Open
Abstract
Artificial Intelligence and Machine Learning (AI/ML) techniques, including reverse vaccinology and predictive models, have already been applied for developing vaccine candidates for COVID-19, HIV, and Hepatitis, streamlining the vaccine development lifecycle from discovery to deployment. The application of AI and ML technologies for improving heath interventions, including drug discovery and clinical development, are expanding across Africa, particularly in South Africa, Kenya, and Nigeria. Further initiatives are required however to expand AI/ML capabilities across the continent to ensure the development of a sustainable ecosystem including enhancing the requisite knowledge base, fostering collaboration between stakeholders, ensuring robust regulatory and ethical frameworks and investment in requisite infrastructure.
Collapse
Affiliation(s)
| | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Agnes Kiragga
- Data Science Program, Africa Population Health Centre, Nairobi, Kenya
| | - Daniel Ochiel
- Henry Jackson Foundation Medical Research International, Nairobi, Kenya
| |
Collapse
|
4
|
Asadi Sarabi P, Shabanpouremam M, Eghtedari AR, Barat M, Moshiri B, Zarrabi A, Vosough M. AI-Based solutions for current challenges in regenerative medicine. Eur J Pharmacol 2024; 984:177067. [PMID: 39454850 DOI: 10.1016/j.ejphar.2024.177067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
The emergence of Artificial Intelligence (AI) and its usage in regenerative medicine represents a significant opportunity that holds the promise of tackling critical challenges and improving therapeutic outcomes. This article examines the ways in which AI, including machine learning and data fusion techniques, can contribute to regenerative medicine, particularly in gene therapy, stem cell therapy, and tissue engineering. In gene therapy, AI tools can boost the accuracy and safety of treatments by analyzing extensive genomic datasets to target and modify genetic material in a precise manner. In cell therapy, AI improves the characterization and optimization of cell products like mesenchymal stem cells (MSCs) by predicting their function and potency. Additionally, AI enhances advanced microscopy techniques, enabling accurate, non-invasive and quantitative analyses of live cell cultures. AI enhances tissue engineering by optimizing biomaterial and scaffold designs, predicting interactions with tissues, and streamlining development. This leads to faster and more cost-effective innovations by decreasing trial and error. The convergence of AI and regenerative medicine holds great transformative potential, promising effective treatments and innovative therapeutic strategies. This review highlights the importance of interdisciplinary collaboration and the continued integration of AI-based technologies, such as data fusion methods, to overcome current challenges and advance regenerative medicine.
Collapse
Affiliation(s)
- Pedram Asadi Sarabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahshid Shabanpouremam
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Amir Reza Eghtedari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Mahsa Barat
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Behzad Moshiri
- School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
5
|
Murcia Pienkowski V, Skoczylas P, Zaremba A, Kłęk S, Balawejder M, Biernat P, Czarnocka W, Gniewek O, Grochowalski Ł, Kamuda M, Król-Józaga B, Marczyńska-Grzelak J, Mazzocco G, Szatanek R, Widawski J, Welanyk J, Orzeszko Z, Szura M, Torbicz G, Borys M, Wohadlo Ł, Wysocki M, Karczewski M, Markowska B, Kucharczyk T, Piatek MJ, Jasiński M, Warchoł M, Kaczmarczyk J, Blum A, Sanecka-Duin A. Harnessing the power of AI in precision medicine: NGS-based therapeutic insights for colorectal cancer cohort. Front Oncol 2024; 14:1407465. [PMID: 39435285 PMCID: PMC11491396 DOI: 10.3389/fonc.2024.1407465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Purpose Developing innovative precision and personalized cancer therapeutics is essential to enhance cancer survivability, particularly for prevalent cancer types such as colorectal cancer. This study aims to demonstrate various approaches for discovering new targets for precision therapies using artificial intelligence (AI) on a Polish cohort of colorectal cancer patients. Methods We analyzed 71 patients with histopathologically confirmed advanced resectional colorectal adenocarcinoma. Whole exome sequencing was performed on tumor and peripheral blood samples, while RNA sequencing (RNAseq) was conducted on tumor samples. We employed three approaches to identify potential targets for personalized and precision therapies. First, using our in-house neoantigen calling pipeline, ARDentify, combined with an AI-based model trained on immunopeptidomics mass spectrometry data (ARDisplay), we identified neoepitopes in the cohort. Second, based on recurrent mutations found in our patient cohort, we selected corresponding cancer cell lines and utilized knock-out gene dependency scores to identify synthetic lethality genes. Third, an AI-based model trained on cancer cell line data was employed to identify cell lines with genomic profiles similar to selected patients. Copy number variants and recurrent single nucleotide variants in these cell lines, along with gene dependency data, were used to find personalized synthetic lethality pairs. Results We identified approximately 8,700 unique neoepitopes, but none were shared by more than two patients, indicating limited potential for shared neoantigenic targets across our cohort. Additionally, we identified three synthetic lethality pairs: the well-known APC-CTNNB1 and BRAF-DUSP4 pairs, along with the recently described APC-TCF7L2 pair, which could be significant for patients with APC and BRAF variants. Furthermore, by leveraging the identification of similar cancer cell lines, we uncovered a potential gene pair, VPS4A and VPS4B, with therapeutic implications. Conclusion Our study highlights three distinct approaches for identifying potential therapeutic targets in cancer patients. Each approach yielded valuable insights into our cohort, underscoring the relevance and utility of these methodologies in the development of precision and personalized cancer therapies. Importantly, we developed a novel AI model that aligns tumors with representative cell lines using RNAseq and methylation data. This model enables us to identify cell lines closely resembling patient tumors, facilitating accurate selection of models needed for in vitro validation.
Collapse
Affiliation(s)
| | | | | | - Stanisław Kłęk
- Surgical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow, Poland
| | | | | | | | | | | | | | | | | | | | | | | | - Joanna Welanyk
- Surgical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow, Poland
| | - Zofia Orzeszko
- Department of Surgery, Faculty of Health Sciences, Jagiellonian University Medical College, Cracow, Poland
| | - Mirosław Szura
- Department of Surgery, Faculty of Health Sciences, Jagiellonian University Medical College, Cracow, Poland
| | - Grzegorz Torbicz
- Department of General Surgery and Surgical Oncology, Ludwik Rydygier Memorial Hospital, Cracow, Poland
| | - Maciej Borys
- Department of General Surgery and Surgical Oncology, Ludwik Rydygier Memorial Hospital, Cracow, Poland
| | - Łukasz Wohadlo
- Department of Oncological and General Surgery, Andrzej Frycz Modrzewski Krakow University, Cracow, Poland
| | - Michał Wysocki
- Department of General Surgery and Surgical Oncology, Ludwik Rydygier Memorial Hospital, Cracow, Poland
| | - Marek Karczewski
- Department of General and Transplant Surgery, Poznan University of Medical Sciences, University Hospital, Poznan, Poland
| | - Beata Markowska
- Department of Surgery, Faculty of Health Sciences, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz Kucharczyk
- Holy Cross Cancer Center Clinic of Clinical Oncology, Kielce, Poland
| | | | | | | | | | | | | |
Collapse
|
6
|
Olawade DB, Teke J, Fapohunda O, Weerasinghe K, Usman SO, Ige AO, Clement David-Olawade A. Leveraging artificial intelligence in vaccine development: A narrative review. J Microbiol Methods 2024; 224:106998. [PMID: 39019262 DOI: 10.1016/j.mimet.2024.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Vaccine development stands as a cornerstone of public health efforts, pivotal in curbing infectious diseases and reducing global morbidity and mortality. However, traditional vaccine development methods are often time-consuming, costly, and inefficient. The advent of artificial intelligence (AI) has ushered in a new era in vaccine design, offering unprecedented opportunities to expedite the process. This narrative review explores the role of AI in vaccine development, focusing on antigen selection, epitope prediction, adjuvant identification, and optimization strategies. AI algorithms, including machine learning and deep learning, leverage genomic data, protein structures, and immune system interactions to predict antigenic epitopes, assess immunogenicity, and prioritize antigens for experimentation. Furthermore, AI-driven approaches facilitate the rational design of immunogens and the identification of novel adjuvant candidates with optimal safety and efficacy profiles. Challenges such as data heterogeneity, model interpretability, and regulatory considerations must be addressed to realize the full potential of AI in vaccine development. Integrating emerging technologies, such as single-cell omics and synthetic biology, promises to enhance vaccine design precision and scalability. This review underscores the transformative impact of AI on vaccine development and highlights the need for interdisciplinary collaborations and regulatory harmonization to accelerate the delivery of safe and effective vaccines against infectious diseases.
Collapse
Affiliation(s)
- David B Olawade
- Department of Allied and Public Health, School of Health, Sport and Bioscience, University of East London, London, United Kingdom; Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, United Kingdom.
| | - Jennifer Teke
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, United Kingdom; Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, United Kingdom
| | | | - Kusal Weerasinghe
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, United Kingdom
| | - Sunday O Usman
- Department of Systems and Industrial Engineering, University of Arizona, USA
| | - Abimbola O Ige
- Department of Chemistry, Faculty of Science, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
7
|
Ananya, Panchariya DC, Karthic A, Singh SP, Mani A, Chawade A, Kushwaha S. Vaccine design and development: Exploring the interface with computational biology and AI. Int Rev Immunol 2024; 43:361-380. [PMID: 38982912 DOI: 10.1080/08830185.2024.2374546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/29/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
Computational biology involves applying computer science and informatics techniques in biology to understand complex biological data. It allows us to collect, connect, and analyze biological data at a large scale and build predictive models. In the twenty first century, computational resources along with Artificial Intelligence (AI) have been widely used in various fields of biological sciences such as biochemistry, structural biology, immunology, microbiology, and genomics to handle massive data for decision-making, including in applications such as drug design and vaccine development, one of the major areas of focus for human and animal welfare. The knowledge of available computational resources and AI-enabled tools in vaccine design and development can improve our ability to conduct cutting-edge research. Therefore, this review article aims to summarize important computational resources and AI-based tools. Further, the article discusses the various applications and limitations of AI tools in vaccine development.
Collapse
Affiliation(s)
- Ananya
- National Institute of Animal Biotechnology, Hyderabad, India
| | | | | | | | - Ashutosh Mani
- Motilal Nehru National Institute of Technology, Prayagraj, India
| | - Aakash Chawade
- Swedish University of Agricultural Sciences, Alnarp, Sweden
| | | |
Collapse
|
8
|
Akinsulie OC, Idris I, Aliyu VA, Shahzad S, Banwo OG, Ogunleye SC, Olorunshola M, Okedoyin DO, Ugwu C, Oladapo IP, Gbadegoye JO, Akande QA, Babawale P, Rostami S, Soetan KO. The potential application of artificial intelligence in veterinary clinical practice and biomedical research. Front Vet Sci 2024; 11:1347550. [PMID: 38356661 PMCID: PMC10864457 DOI: 10.3389/fvets.2024.1347550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Artificial intelligence (AI) is a fast-paced technological advancement in terms of its application to various fields of science and technology. In particular, AI has the potential to play various roles in veterinary clinical practice, enhancing the way veterinary care is delivered, improving outcomes for animals and ultimately humans. Also, in recent years, the emergence of AI has led to a new direction in biomedical research, especially in translational research with great potential, promising to revolutionize science. AI is applicable in antimicrobial resistance (AMR) research, cancer research, drug design and vaccine development, epidemiology, disease surveillance, and genomics. Here, we highlighted and discussed the potential impact of various aspects of AI in veterinary clinical practice and biomedical research, proposing this technology as a key tool for addressing pressing global health challenges across various domains.
Collapse
Affiliation(s)
- Olalekan Chris Akinsulie
- Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
- College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Ibrahim Idris
- Faculty of Veterinary Medicine, Usman Danfodiyo University, Sokoto, Nigeria
| | | | - Sammuel Shahzad
- College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | | | - Seto Charles Ogunleye
- Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Population Medicine and Pathobiology, College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Mercy Olorunshola
- Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
| | - Deborah O. Okedoyin
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, United States
| | - Charles Ugwu
- College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | | | - Joy Olaoluwa Gbadegoye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Qudus Afolabi Akande
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | - Pius Babawale
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Sahar Rostami
- Department of Population Medicine and Pathobiology, College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | | |
Collapse
|
9
|
Bujak J, Kłęk S, Balawejder M, Kociniak A, Wilkus K, Szatanek R, Orzeszko Z, Welanyk J, Torbicz G, Jęckowski M, Kucharczyk T, Wohadlo Ł, Borys M, Stadnik H, Wysocki M, Kayser M, Słomka ME, Kosmowska A, Horbacka K, Gach T, Markowska B, Kowalczyk T, Karoń J, Karczewski M, Szura M, Sanecka-Duin A, Blum A. Creating an Innovative Artificial Intelligence-Based Technology (TCRact) for Designing and Optimizing T Cell Receptors for Use in Cancer Immunotherapies: Protocol for an Observational Trial. JMIR Res Protoc 2023; 12:e45872. [PMID: 37440307 PMCID: PMC10375398 DOI: 10.2196/45872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Cancer continues to be the leading cause of mortality in high-income countries, necessitating the development of more precise and effective treatment modalities. Immunotherapy, specifically adoptive cell transfer of T cell receptor (TCR)-engineered T cells (TCR-T therapy), has shown promise in engaging the immune system for cancer treatment. One of the biggest challenges in the development of TCR-T therapies is the proper prediction of the pairing between TCRs and peptide-human leukocyte antigen (pHLAs). Modern computational immunology, using artificial intelligence (AI)-based platforms, provides the means to optimize the speed and accuracy of TCR screening and discovery. OBJECTIVE This study proposes an observational clinical trial protocol to collect patient samples and generate a database of pHLA:TCR sequences to aid the development of an AI-based platform for efficient selection of specific TCRs. METHODS The multicenter observational study, involving 8 participating hospitals, aims to enroll patients diagnosed with stage II, III, or IV colorectal cancer adenocarcinoma. RESULTS Patient recruitment has recently been completed, with 100 participants enrolled. Primary tumor tissue and peripheral blood samples have been obtained, and peripheral blood mononuclear cells have been isolated and cryopreserved. Nucleic acid extraction (DNA and RNA) has been performed in 86 cases. Additionally, 57 samples underwent whole exome sequencing to determine the presence of somatic mutations and RNA sequencing for gene expression profiling. CONCLUSIONS The results of this study may have a significant impact on the treatment of patients with colorectal cancer. The comprehensive database of pHLA:TCR sequences generated through this observational clinical trial will facilitate the development of the AI-based platform for TCR selection. The results obtained thus far demonstrate successful patient recruitment and sample collection, laying the foundation for further analysis and the development of an innovative tool to expedite and enhance TCR selection for precision cancer treatments. TRIAL REGISTRATION ClinicalTrials.gov NCT04994093; https://clinicaltrials.gov/ct2/show/NCT04994093. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/45872.
Collapse
Affiliation(s)
- Joanna Bujak
- Ardigen SA, Cracow, Poland
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland
| | - Stanisław Kłęk
- Surgical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow, Poland
| | | | | | | | | | - Zofia Orzeszko
- Department of General and Oncological Surgery, Brothers Hospitallers Hospital, Cracow, Poland
| | - Joanna Welanyk
- Surgical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow, Poland
| | - Grzegorz Torbicz
- Department of General Surgery and Surgical Oncology, Ludwik Rydygier Memorial Hospital, Cracow, Poland
| | - Mateusz Jęckowski
- Colon Cancer Unit, Department of Oncological Surgery, Voivodeship Multi-Specialist Center for Oncology and Traumatology, Lodz, Poland
| | - Tomasz Kucharczyk
- Holy Cross Cancer Center Clinic of Clinical Oncology, Cracow, Poland
| | - Łukasz Wohadlo
- Department of General Surgery, Andrzej Frycz Modrzewski Krakow University, Cracow, Poland
| | - Maciej Borys
- Department of General Surgery and Surgical Oncology, Ludwik Rydygier Memorial Hospital, Cracow, Poland
| | - Honorata Stadnik
- Department of General and Transplant Surgery, Poznan University of Medical Sciences, University Hospital, Poznan, Poland
| | - Michał Wysocki
- Department of General Surgery and Surgical Oncology, Ludwik Rydygier Memorial Hospital, Cracow, Poland
| | - Magdalena Kayser
- General and Colorectal Surgery Department, J Struś Multispecialist Municipal Hospital, Poznan, Poland
| | - Marta Ewa Słomka
- Colon Cancer Unit, Department of Oncological Surgery, Voivodeship Multi-Specialist Center for Oncology and Traumatology, Lodz, Poland
| | - Anna Kosmowska
- General and Colorectal Surgery Department, J Struś Multispecialist Municipal Hospital, Poznan, Poland
| | - Karolina Horbacka
- General and Colorectal Surgery Department, J Struś Multispecialist Municipal Hospital, Poznan, Poland
| | - Tomasz Gach
- Surgical Clinic Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, Cracow, Poland
| | - Beata Markowska
- Surgical Clinic Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz Kowalczyk
- Department of General Surgery, Andrzej Frycz Modrzewski Krakow University, Cracow, Poland
| | - Jacek Karoń
- General and Colorectal Surgery Department, J Struś Multispecialist Municipal Hospital, Poznan, Poland
| | - Marek Karczewski
- Department of General and Transplant Surgery, Poznan University of Medical Sciences, University Hospital, Poznan, Poland
| | - Mirosław Szura
- Surgical Clinic Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, Cracow, Poland
| | | | | |
Collapse
|
10
|
Fu W, Xie Z, Bai M, Zhang Z, Zhao Y, Tian J. Proteomics analysis of methionine enkephalin upregulated macrophages against infection by the influenza-A virus. Proteome Sci 2023; 21:4. [PMID: 37041527 PMCID: PMC10088144 DOI: 10.1186/s12953-023-00205-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/03/2023] [Indexed: 04/13/2023] Open
Abstract
Macrophages have a vital role in phagocytosis and antiviral effect against invading influenza viruses. Previously, we found that methionine enkephalin (MENK) inhibited influenza virus infection by upregulating the "antiviral state" of macrophages. To investigate the immunoregulatory mechanism of action of MENK on macrophages, we employed proteomic analysis to identify differentially expressed proteins (DEPs) between macrophages infected with the influenza-A virus and cells infected with the influenza-A virus after pretreatment with MENK. A total of 215 DEPs were identified: 164 proteins had upregulated expression and 51 proteins had downregulated expression. Proteomics analysis showed that DEPs were highly enriched in "cytokine-cytokine receptor interaction", "phagosome", and "complement and coagulation cascades pathway". Proteomics analysis revealed that MENK could be an immune modulator or prophylactic for the prevention and treatment of influenza. MENK promoted the polarization of M1 macrophages, activated inflammatory responses, and enhanced phagocytosis and killing function by upregulating opsonizing receptors.
Collapse
Affiliation(s)
- Wenrui Fu
- Graduate College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Zifeng Xie
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Mei Bai
- Department of Microbiology, Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, China
| | - Zhen Zhang
- Department of Microbiology, Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, China
| | - Yuanlong Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Jing Tian
- Department of Immunology, School of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
11
|
State of the art in epitope mapping and opportunities in COVID-19. Future Sci OA 2023; 16:FSO832. [PMID: 36897962 PMCID: PMC9987558 DOI: 10.2144/fsoa-2022-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
The understanding of any disease calls for studying specific biological structures called epitopes. One important tool recently drawing attention and proving efficiency in both diagnosis and vaccine development is epitope mapping. Several techniques have been developed with the urge to provide precise epitope mapping for use in designing sensitive diagnostic tools and developing rpitope-based vaccines (EBVs) as well as therapeutics. In this review, we will discuss the state of the art in epitope mapping with a special emphasis on accomplishments and opportunities in combating COVID-19. These comprise SARS-CoV-2 variant analysis versus the currently available immune-based diagnostic tools and vaccines, immunological profile-based patient stratification, and finally, exploring novel epitope targets for potential prophylactic, therapeutic or diagnostic agents for COVID-19.
Collapse
|
12
|
Liu CH, Lu CH, Lin LT. Pandemic strategies with computational and structural biology against COVID-19: A retrospective. Comput Struct Biotechnol J 2021; 20:187-192. [PMID: 34900126 PMCID: PMC8650801 DOI: 10.1016/j.csbj.2021.11.040] [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: 07/06/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022] Open
Abstract
The emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), which is the etiologic agent of the coronavirus disease 2019 (COVID-19) pandemic, has dominated all aspects of life since of 2020. Research studies on the virus and exploration of therapeutic and preventive strategies has been moving at rapid rates to control the pandemic. In the field of bioinformatics or computational and structural biology, recent research strategies have used multiple disciplines to compile large datasets to uncover statistical correlations and significance, visualize and model proteins, perform molecular dynamics simulations, and employ the help of artificial intelligence and machine learning to harness computational processing power to further the research on COVID-19, including drug screening, drug design, vaccine development, prognosis prediction, and outbreak prediction. These recent developments should help us better understand the viral disease and develop the much-needed therapies and strategies for the management of COVID-19.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Cheng-Hua Lu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Liang-Tzung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|