1
|
Sequeira DP, Suomalainen M, Freitag PC, Plückthun A, Klingenbrunner M, Fischer L, Hemmi S, Münz C, Volle R, Greber UF. Activated blood-derived human primary T cells support replication of HAdV C5 and virus transmission to polarized human primary epithelial cells. J Virol 2025; 99:e0182524. [PMID: 40265914 PMCID: PMC12090788 DOI: 10.1128/jvi.01825-24] [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: 10/17/2024] [Accepted: 03/29/2025] [Indexed: 04/24/2025] Open
Abstract
Human adenoviruses (HAdVs) cause self-limiting disease but are life-threatening to immunocompromised individuals. HAdV-C5 infects epithelial cells of the airways and eyes through aerosols, contaminated hands, or medical instruments, as well as fecal-oral contacts, gives rise to viremia, persisting in lymphoid cells of the gastrointestinal tract. Here, we show that pre-activated human primary blood-derived T cells can be infected with HAdV-C5 in vitro, upon incubation of the virus with a mixture of three distinct homotrimeric adapter proteins that target the virus to T cells. Each of the adapter proteins can bind 1 of the 12 fiber knobs of the virion through a designed ankyrin repeat protein. Two of the adapters contained a single-chain antibody fragment to T cell surface proteins CD3 or CD28, and the third one contained the cytokine interleukin-2. These adapters mediated efficient infection of primary T cells by HAdV-C5 and infectious progeny release, albeit with donor-to-donor variability. Co-culture of well-polarized air-liquid interface human bronchial epithelial cells with infected CD3+ T cells gave rise to progressively increased viral titers from replicating but not from replication-defective E1-deleted HAdV-C5, notably with similar kinetics as cell-free virus infections, suggesting that progeny virus from T cells was further amplified in epithelial cells. This study provides a platform to explore interactions between epithelial and immune cells in acute and persistent HAdV-C5 infection settings.IMPORTANCEMany human adenoviruses (HAdV), including HAdV-C5, infect and propagate to high titers in epithelial cells of the airways. Virus ends up in lymphoid cells of the gastrointestinal and respiratory mucosa, where it can persist subclinically for years, restricted by memory T cells and humoral immune defense. In immunodeficient patients or newborns, however, HAdV can be fatal, coincident with lymphocytopenia and virus proliferation in epithelial cells. Here, we show that activated blood-derived human primary T lymphocytes can be productively infected with HAdV-C5 coated with trimerized adapter proteins targeting CD3, CD28, and the interleukin 2 receptors. A co-culture model of infected T cells and primary human bronchial epithelial cells in the absence of HAdV-specific immune cells showed that progeny virus from T cells was transferred to epithelial cells and led to increased progeny production compared to infected T cells alone, a situation potentially mimicking persistently infected mucosal lymphoid cells in immunosuppressed patients.
Collapse
Affiliation(s)
| | - Maarit Suomalainen
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | | | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Zürich, Switzerland
| | | | - Lucy Fischer
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Silvio Hemmi
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Romain Volle
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Urs F. Greber
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Ball J, Bradley A, Le A, Tisdale JF, Uchida N. Current and future treatments for sickle cell disease: From hematopoietic stem cell transplantation to in vivo gene therapy. Mol Ther 2025; 33:2172-2191. [PMID: 40083162 DOI: 10.1016/j.ymthe.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Sickle cell disease (SCD) is a single-gene disorder caused by a point mutation of the β-globin gene, resulting in hemolytic anemia, acute pain, multiorgan damage, and early mortality. Hydroxyurea is a first-line drug therapy that switches sickle-globin to non-pathogenic γ-globin; however, it requires lifelong oral administration. Allogeneic hematopoietic stem cell (HSC) transplantation allows for a one-time cure for SCD, albeit with histocompatibility limitations. Therefore, autologous HSC gene therapy was developed to cure SCD in a single treatment, without HSC donors. Current HSC gene therapy is based on the ex vivo culture of patients' HSCs with lentiviral gene addition and gene editing, followed by autologous transplantation back to the patient. However, the complexity of the treatment process and high costs hinder the universal application of ex vivo gene therapy. Therefore, the development of in vivo HSC gene therapy, where gene therapy tools are directly administered to patients, is desirable to provide a more accessible, cost-effective solution that can cure SCD worldwide. In this review, we discuss current treatments, including drug therapies, HSC transplantation, and ex vivo gene therapy; the development of gene therapy tools; and progress toward curative in vivo gene therapy in SCD.
Collapse
Affiliation(s)
- Julia Ball
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Avery Bradley
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anh Le
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Stewart G, Tazzyman S, Sun Y, Andrews RE, Harrison J, Lath D, Down J, Robinson G, Wang X, Muthana M, Chantry AD, Lawson MA. An oncolytic adenovirus targeting SLAMF7 demonstrates anti-myeloma efficacy. Leukemia 2025:10.1038/s41375-025-02617-3. [PMID: 40247106 DOI: 10.1038/s41375-025-02617-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
We investigated a novel SLAMF7-promoter driven oncolytic adenovirus (Ad[CE1A]) as a potential therapeutic for multiple myeloma, an incurable hematological malignancy. Ad[CE1A] infection, replication, and oncolysis were assessed in a panel of myeloma cell lines (n = 8) and ex vivo samples from myeloma patients (n = 17) and healthy donors (HDs) (n = 14). Ad[CE1A] efficiently infected, replicated, and induced oncolysis in myeloma cells, but not in control cell lines or HDs, demonstrating selective cytotoxicity. Mechanistic studies revealed Ad[CE1A]-induced cell death is caspase-independent, with a potential involvement of necroptosis. Ad[CE1A] also altered immunogenic cell death markers (calreticulin, CD47, extracellular ATP), enhanced antigen presentation via increased MHC class I and II receptor expression (HLA-ABC and HLA-DR), and stimulated bystander cytokine killing, indicating potential for direct and immune-mediated anti-myeloma responses. In vivo experiments with 5TGM1 syngeneic and U266 xenograft models showed Ad[CE1A] significantly reduced myeloma tumor burden compared to vehicle control. Combination therapy with anti-myeloma drugs, bortezomib, melphalan, panobinostat and pomalidomide, enhanced Ad[CE1A] efficacy, with melphalan upregulating SLAMF7, resulting in increased viral replication. In summary, these findings support Ad[CE1A] as a promising myeloma therapy.
Collapse
Affiliation(s)
- Georgia Stewart
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Simon Tazzyman
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Yidan Sun
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Rebecca E Andrews
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Jack Harrison
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Darren Lath
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Jenny Down
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Georgia Robinson
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Xue Wang
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Munitta Muthana
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Andrew D Chantry
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Michelle A Lawson
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK.
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK.
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK.
| |
Collapse
|
4
|
Murala MST, Gairola V, Sayedahmed EE, Mittal SK. Next-Generation Adenoviral Vector-Based Vaccines for Severe Acute Respiratory Syndrome Coronavirus-2. Vaccines (Basel) 2025; 13:406. [PMID: 40333307 PMCID: PMC12031563 DOI: 10.3390/vaccines13040406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
This review systematically revises adenovirus (Ad) biology, vector structure, immune responses, and currently available Ad vector COVID-19 vaccines. It analyzes the challenges associated with the Ad vector-based vaccines, including preexisting vector immunity and other side effects. Moreover, this review explores novel and innovative strategies to overcome these constraints for developing next-generation vaccines for broad protection to cover emerging SARS-CoV-2 variants. The future refinement of Ad vaccine platforms will be pivotal in achieving durable immunity against emerging variants for global preparedness.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907-2027, USA; (M.S.T.M.); (V.G.); (E.E.S.)
| |
Collapse
|
5
|
Dai Z, Si Y, Xiong S, Li Y, Ye J, Gao Q, Ma D, Jin X, Li F. Chimeric Ad5/35 oncolytic adenovirus overcome preexisting neutralizing antibodies and enhance tumor targeting efficiency. Cancer Gene Ther 2025; 32:418-436. [PMID: 40057574 DOI: 10.1038/s41417-025-00884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025]
Abstract
KD01, a third-generation conditionally replicating adenovirus serotype 5 developed by our team, has approved by the China Center for Drug Evaluation (CDE) for Phase I clinical trials (NCT06552598). However, 60% seroprevalence of anti-Ad5 neutralizing antibodies is a major hurdle for Ad5-based oncolytic viruses. To address this issue, we developed oAd5/35-HF, a fourth-generation oncolytic adenovirus vector designed to enhance infection efficiency and evade pre-existing neutralizing antibodies (NABs). To achieve this, we introduced targeted capsid modifications, replacing hexon hypervariable regions (HVRs) 1 and 5 with those from adenovirus serotype 35 (Ad35), along with alterations to the fiber region. These combined modifications significantly improved infection efficiency, maintained high viral titers, and enabled the virus to resist NABs. This is the first report of replacing both the Ad5 hexon HVRs and fiber regions with those from Ad35 in an oncolytic adenovirus, resulting in potent antitumor activity across multiple cancer types, even in the presence of high NAB levels. The oAd5/35-HF backbone provides a versatile platform for developing new chimera oncolytic adenovirus and adenovirus vector-based vaccine.
Collapse
Affiliation(s)
- Zhoutong Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Si
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengfeng Xiong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Ye
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Jin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fei Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Centre for Obstetrics and Gynecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Dermody TS, Sutherland DM. mGem: The complexity of viral entry-one virus, many receptors. mBio 2025; 16:e0296424. [PMID: 39932305 PMCID: PMC11898697 DOI: 10.1128/mbio.02964-24] [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] [Indexed: 03/14/2025] Open
Abstract
Binding to cellular receptors initiates viral replication and dictates sites in the host infected by the virus. As illustrated by mammalian orthoreovirus (reovirus), viruses can bind several types of receptors using distinct capsid components to facilitate the viral entry steps of attachment, internalization, and disassembly. The outer of the two concentric capsids of reovirus virions is formed by four viral proteins, three of which bind receptors. These capsid-receptor interactions mediate stepwise entry of reovirus, dictate viral tropism in infected animals, and expand the viral host range. Engagement of independent receptors by different capsid proteins is a property of many pathogenic viruses and illustrates common themes of receptor use in viral entry and disease.
Collapse
Affiliation(s)
- Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Danica M. Sutherland
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institute of Infection, Inflammation and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
A R, Ueki H, Nishioka S, Yamazaki R, Maekawa M, Kitagawa K, Miyake H, Shirakawa T. A conditionally replicative adenovirus vector containing the synNotch receptor gene for the treatment of muscle-invasive bladder cancer. Cancer Gene Ther 2025; 32:306-317. [PMID: 40011711 PMCID: PMC11946899 DOI: 10.1038/s41417-025-00879-8] [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: 08/19/2024] [Revised: 01/16/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
Muscle-invasive bladder cancer (MIBC), a highly heterogeneous disease, shows genomic instability and a high mutation rate, making it difficult to treat. Recent studies revealed that cancer stem cells (CSCs) play a critical role in MIBC frequent recurrence and high morbidity. Previous research has shown that Cyclooxygenases-2 (COX-2) is particularly highly expressed in bladder cancer cells. In recent years, the development of oncolytic adenoviruses and their use in clinical trials have gained increased attention. In this study, we composed a conditionally replicative adenovirus vector (CRAd-synNotch) that carries the COX-2 promotor driving adenoviral E1 gene, the synNotch receptor therapeutic gene, and the Ad5/35 fiber gene. Activation of the COX-2 promoter gene causes replication only within COX-2 expressing cancer cells, thereby leading to tumor oncolysis. Also, CD44 and HIF signals contribute to cancer stemness and maintaining CSCs in bladder cancer, and the transduced synNotch receptor inhibits both CD44 and HIF signals simultaneously. We performed an in vivo study using a mouse xenograft model of T24 human MIBC cells and confirmed the significant antitumor activity of CRAd-synNotch. Our findings in this study warrant the further development of CRAd-synNotch for treating patients with MIBC.
Collapse
Affiliation(s)
- Ruhan A
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Hideto Ueki
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shunya Nishioka
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Rion Yamazaki
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Marina Maekawa
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Koichi Kitagawa
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Hideaki Miyake
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiro Shirakawa
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan.
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
8
|
Yang Y, Yuan H, Zhang Y, Luan J, Wang H. Progress in African Swine Fever Vector Vaccine Development. Int J Mol Sci 2025; 26:921. [PMID: 39940691 PMCID: PMC11816837 DOI: 10.3390/ijms26030921] [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: 11/22/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
African swine fever (ASF) is a highly lethal, infectious, hemorrhagic fever disease, characterized by an acute mortality rate approaching 100%. It is highly contagious, and results in significant losses to the global hog industry as it spreads. Despite incremental progress in research on the African swine fever virus (ASFV), a safe and effective commercial vaccine has yet to be developed. Vector vaccines, a promising type of vaccine, offer unique advantages, and are a primary focus in ASFV vaccine research. This paper focuses on the characteristics of viral, bacterial, and yeast vector vaccines; elucidates the immunological mechanisms associated with antigens; lists the types of antigens that have significant potential; discusses the feasibility of using exogenously expressed cytokines to enhance the protective power of vector vaccines; and, finally, discusses the types of vectors that are commonly used and the latest advances in this field.
Collapse
Affiliation(s)
| | | | | | | | - Hailong Wang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, Shandong University, Qingdao 266237, China; yangyue-@mail.sdu.edu.cn (Y.Y.); (H.Y.); (Y.Z.); (J.L.)
| |
Collapse
|
9
|
Sosnovtseva AO, Le TH, Karpov DS, Vorobyev PO, Gumennaya YD, Alekseeva ON, Chumakov PM, Lipatova AV. Establishment of a Panel of Human Cell Lines to Identify Cellular Receptors Used by Enteroviruses to Infect Cells. Int J Mol Sci 2025; 26:923. [PMID: 39940693 PMCID: PMC11817244 DOI: 10.3390/ijms26030923] [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: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Non-pathogenic natural and recombinant strains of human Enteroviruses are the subject of ongoing study with some strains having been approved for use as anticancer agents. The efficacy of oncolytic virotherapy depends upon identifying the receptor utilized by a specific strain for cell entry, and the presence of this receptor on the surface of cancer cells. Accordingly, a rapid and straightforward approach to determining the enteroviral receptors is necessary for developing an effective patient-specific, virus-based cancer therapy. To this end, we created a panel of seven lines with double knockouts on the background of the HEK293T cell line, which lacks the IFNAR1 gene. In these lines, the main viral receptor genes, including PVR, CXADR, CD55, ITGA2, SCARB2, ICAM1, and FCGRT, were knocked out using the CRISPR/Cas9 system. The panel of lines was validated on twelve different Enteroviruses types, providing a basis for studying the molecular mechanisms of enterovirus entry into cells, and for developing new therapeutic strains.
Collapse
Affiliation(s)
- Anastasiia O. Sosnovtseva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.S.K.); (P.O.V.); (P.M.C.); (A.V.L.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| | - Thi Hoa Le
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Dmitry S. Karpov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.S.K.); (P.O.V.); (P.M.C.); (A.V.L.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| | - Pavel O. Vorobyev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.S.K.); (P.O.V.); (P.M.C.); (A.V.L.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| | - Yana D. Gumennaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| | - Olga N. Alekseeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.S.K.); (P.O.V.); (P.M.C.); (A.V.L.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| | - Anastasia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (D.S.K.); (P.O.V.); (P.M.C.); (A.V.L.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.D.G.); (O.N.A.)
| |
Collapse
|
10
|
Liaci AM, Chandra N, Vodnala SM, Strebl M, Kumar P, Pfenning V, Bachmann P, Caraballo R, Chai W, Johansson E, Elofsson M, Feizi T, Liu Y, Stehle T, Arnberg N. Extended receptor repertoire of an adenovirus associated with human obesity. PLoS Pathog 2025; 21:e1012892. [PMID: 39883726 PMCID: PMC11813153 DOI: 10.1371/journal.ppat.1012892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/11/2025] [Accepted: 01/07/2025] [Indexed: 02/01/2025] Open
Abstract
Human adenovirus type 36 (HAdV-D36) has been putatively linked to obesity in animals and has been associated with obesity in humans in some but not all studies. Despite extensive epidemiological research there is limited information about its receptor profile. We investigated the receptor portfolio of HAdV-D36 using a combined structural biology and virology approach. The HAdV-D36 fiber knob domain (FK), which mediates the primary attachment of many HAdVs to host cells, has a significantly elongated DG loop that alters known binding interfaces for established adenovirus receptors such as the coxsackie- and adenovirus receptor (CAR) and CD46. Our data suggest that HAdV-D36 attaches to host cells using a versatile receptor pool comprising sialic acid-containing glycans and CAR. Sialic acids are recognized at the same binding site used by other HAdVs of species D such as HAdV-D37. Using glycan microarrays, we demonstrate that HAdV-D36 displays a binding preference for glycans containing a rare sialic acid variant, 4-O,5-N-diacetylneuraminic acid, over the more common 5-N-acetylneuraminic acid. To date, this sialic acid variant has not been detected in humans, although it can be synthesized by various animal species, including a range of domestic and livestock animals. Taken together, our results indicate that HAdV-D36 has evolved to recognize a specialized set of primary attachment receptors that are different from known HAdV types and coincides with a unique host range and pathogenicity profile.
Collapse
Affiliation(s)
- A. Manuel Liaci
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Naresh Chandra
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | | | - Michael Strebl
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Pravin Kumar
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Vanessa Pfenning
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Paul Bachmann
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Rémi Caraballo
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Wengang Chai
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Emil Johansson
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Mikael Elofsson
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Ten Feizi
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Yan Liu
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Niklas Arnberg
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
11
|
Xie Y, Mei H, Wang W, Li X, Hu P, Tian X, Zhou R, Liu J, Qu J. ALCAM is an entry factor for severe community acquired Pneumonia-associated Human adenovirus species B. Nat Commun 2024; 15:10889. [PMID: 39738070 PMCID: PMC11686370 DOI: 10.1038/s41467-024-55261-3] [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: 03/26/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Human adenovirus (HAdV) is a widely spread respiratory pathogen that can cause infections in multiple tissues and organs. Previous studies have established an association between HAdV species B (HAdV-B) infection and severe community-acquired pneumonia (SCAP). However, the connection between SCAP-associated HAdV-B infection and host factor expression profile in patients has not been systematically investigated. Here, we perform a CRISPR genetic screen on HAdV-B using two generations of cell surface protein-focused CRISPR libraries and identify a series of host factors including the known receptor DSG-2 and an unknown factor, activated leukocyte cell adhesion molecule (ALCAM). Further investigation shows that ALCAM affects HAdV-B infection by participating in viral internalization. Transcriptomics data from human blood samples suggests that ALCAM expression is higher in SCAP patients with HAdV-B infection than in those with other infections. Chimeric and authentic virus experiments show that ALCAM is a widely used host factor across B1 and B2 genetic clusters of HAdV-B. The dissociation constant between the knob domain of HAdV-B fiber and ALCAM is 837 nM in average. In summary, our results suggest that ALCAM is an entry factor for SCAP-associated HAdV-B.
Collapse
Affiliation(s)
- Yusang Xie
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institutes of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Hong Mei
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiao Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Pengfei Hu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xingui Tian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Rong Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, Guangdong, China
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, Guangdong, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institutes of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China.
| |
Collapse
|
12
|
Wienen F, Nilson R, Allmendinger E, Peters S, Barth TF, Kochanek S, Krutzke L. An oncolytic HAdV-5 with reduced surface charge combines diminished toxicity and improved tumor targeting. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200909. [PMID: 39758252 PMCID: PMC11699628 DOI: 10.1016/j.omton.2024.200909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/12/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025]
Abstract
Human adenovirus type 5 (HAdV-5)-based oncolytic viruses hold significant promise for anti-cancer therapy. However, poor tumor-targeting and off-target organ transduction after systemic administration limit their therapeutic efficacy. In addition, the strong liver tropism of HAdV-5-based vectors poses the risk of hepatotoxicity. By genetic modification of the major capsid protein hexon we generated a HAdV-5-based oncolytic vector (HAdV-5-HexPos3) with reduced negative surface charge. Coxsackie and adenovirus receptor (CAR) binding-ablated (ΔCAR) HAdV-5-HexPos3_ΔCAR exhibited superior and CAR-independent transduction of various cancer cell lines in vitro, further enhanced in the presence of HAdV-5 naive murine plasma. Upon intravenous administration into tumor-bearing immunodeficient NSG mice, replication-deficient HAdV-5-HexPos3_ΔCAR vector particles showed significantly reduced off-target organ tropism in all tissues analyzed, including the liver. Moreover, we detected a significantly increased intratumoral vector load for HAdV-5-HexPos3_ΔCAR, leading to a 29-fold elevated tumor-to-liver ratio compared with a control vector with unmodified hexon. Intravenous injection of a conditionally replicating hexon-unmodified control vector induced severe hepatotoxicity in tumor-bearing NSG mice, while a conditionally replicating HAdV-5-HexPos3_ΔCAR vector was well tolerated and resulted in intratumoral vector presence for up to 56 days. HAdV-5-HexPos3_ΔCAR represents a promising vector platform for the generation of HAdV-5-based oncolytic viruses with reduced systemic toxicity and improved therapeutic efficacy.
Collapse
Affiliation(s)
- Frederik Wienen
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| | - Robin Nilson
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| | | | - Sarah Peters
- Department of Clinical Chemistry, Ulm University Medical Center, 89081 Ulm, Germany
| | - Thomas F.E. Barth
- Institute of Pathology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Stefan Kochanek
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| | - Lea Krutzke
- Department of Gene Therapy, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
13
|
Muravyeva A, Smirnikhina S. Adenoviral Vectors for Gene Therapy of Hereditary Diseases. BIOLOGY 2024; 13:1052. [PMID: 39765719 PMCID: PMC11673936 DOI: 10.3390/biology13121052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
Adenoviral vectors (AdVs) are effective vectors for gene therapy due to their broad tropism, high capacity, and high transduction efficiency, which makes them actively used as oncolytic vectors and for creating vector vaccines. However, despite their numerous advantages, AdVs have not yet found their place in gene therapy for hereditary diseases. This review provides an overview of AdVs, their features, and clinical trials using them for gene replacement therapy in monogenic diseases and analyzes the reasons for the failures of these studies. Additionally, current research on the modification of AdVs to reduce immune responses and target delivery is discussed.
Collapse
Affiliation(s)
| | - Svetlana Smirnikhina
- Laboratory of Genome Editing, Research Centre for Medical Genetics, Moskvorechye, 1, 115522 Moscow, Russia
| |
Collapse
|
14
|
Bastea LI, Liu X, Fleming AK, Pandey V, Döppler H, Edenfield BH, Krishna M, Zhang L, Thompson EA, Grandgenett PM, Hollingsworth MA, Fairweather D, Clemens D, Storz P. Coxsackievirus and adenovirus receptor expression facilitates enteroviral infections to drive the development of pancreatic cancer. Nat Commun 2024; 15:10547. [PMID: 39627248 PMCID: PMC11615305 DOI: 10.1038/s41467-024-55043-x] [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: 10/07/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
The development of pancreatic cancer requires both, acquisition of an oncogenic mutation in KRAS as well as an inflammatory insult. However, the physiological causes for pancreatic inflammation are less defined. We show here that oncogenic KRas-expressing pre-neoplastic lesion cells upregulate coxsackievirus (CVB) and adenovirus receptor (CAR). This facilitates infections from enteroviruses such as CVB3, which can be detected in approximately 50% of pancreatic cancer patients. Moreover, using an animal model we show that a one-time pancreatic infection with CVB3 in control mice is transient, but in the presence of oncogenic KRas drives chronic inflammation and rapid development of pancreatic cancer. We further demonstrate that a knockout of CAR in pancreatic lesion cells blocks these CVB3-induced effects. Our data demonstrate that KRas-caused lesions promote the development of pancreatic cancer by enabling certain viral infections.
Collapse
Affiliation(s)
- Ligia I Bastea
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Xiang Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Alicia K Fleming
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Veethika Pandey
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Heike Döppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Murli Krishna
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Lizhi Zhang
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - DeLisa Fairweather
- Department of Cardiovascular Diseases, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Dahn Clemens
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
15
|
Yan HW, Feng YD, Tang N, Cao FC, Lei YF, Cao W, Li XQ. Viral myocarditis: From molecular mechanisms to therapeutic prospects. Eur J Pharmacol 2024; 982:176935. [PMID: 39182550 DOI: 10.1016/j.ejphar.2024.176935] [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: 06/19/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Myocarditis is characterized as local or diffuse inflammatory lesions in the myocardium, primarily caused by viruses and other infections. It is a common cause of sudden cardiac death and dilated cardiomyopathy. In recent years, the global prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the widespread vaccination have coincided with a notable increase in the number of reported cases of myocarditis. In light of the potential threat that myocarditis poses to global public health, numerous studies have sought to elucidate the pathogenesis of this condition. However, despite these efforts, effective treatment strategies remain elusive. To collate the current research advances in myocarditis, and thereby provide possible directions for further research, this review summarizes the mechanisms involved in viral invasion of the organism and primarily focuses on how viruses trigger excessive inflammatory responses and in result in different types of cell death. Furthermore, this article outlines existing therapeutic approaches and potential therapeutic targets for the acute phase of myocarditis. In particular, immunomodulatory treatments are emphasized and suggested as the most extensively studied and clinically promising therapeutic options.
Collapse
Affiliation(s)
- Han-Wei Yan
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Ying-Da Feng
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Na Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Feng-Chuan Cao
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Ying-Feng Lei
- Department of Microbiology, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
16
|
Naveed M, Batool Z, Aziz T, Javed K, Ali N, Rehman HM, Alharbi M, Alasmari AF, Alshammari A. An in silico approach uncovering the competency of oncolytic human adenovirus 52 for targeted breast cancer virotherapy. Sci Rep 2024; 14:26405. [PMID: 39488601 PMCID: PMC11531525 DOI: 10.1038/s41598-024-77664-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
Breast cancer remains a major health threat throughout the world specifically in women above 30 years of age however, it is rarely known to affect men as well. It is characterized by the abnormal division of cells in the breast tissue resulting in the development of breast malignancies. Various risk factors contributing to breast cancer include age, family history, genetic mutations (chiefly in BRCA1 and BRCA2 genes) along with hormonal imbalances (oestrogen, progesterone, HER2). Early detection which can be obtained through frequent rounds of self-examination, mammographic scanning, and clinical assessment plays a crucial role in the prevention of the disease. In addition, appropriate diagnosis assists in better therapeutic responses. This study highlights the considerable health risks associated with the conventional treatment procedures which arise and increased demand of advanced, secure, and risk-free treatment alternatives. Oncolytic viruses are potentially apparent for the aim of improving cancer therapeutics with reduced side effects. These viruses act as the fundamental therapeutic agent themselves that selectively target and kill malignant cells without harm to healthy tissues. The key objective of the research is to provide evidence that Human Adenovirus 52 is a potent oncolytic virus and to highlight its capacity to target and eliminate cancer cells with precision while causing the least amount of harm to healthy tissues. Validating the in-silico method entails evaluating the precision and dependability of the computational modelling by contrasting the in-silico predictions with the findings from the experiments rank as the secondary objective. The workflow of this research utilized in-silico computational drug designing approaches including retrieval of tertiary structures of both the target Breast Cancer Type 1 Susceptibility Protein (BRCA1) and the viral Human Adenovirus 52 protein, their validation generating Ramachandran Plots determining favoured amino acid residue angles and prediction of their active residues. Furthermore, the study focused on the molecular dynamics docking of proteins, interpretation of molecular interactions between the docked complex, as well as the assessment of the molecular dynamic simulations (MD) in addition to their MMGBSA binding energy calculations. A successful docking between BRCA1 and Adenovirus protein provided a significant score of 329.2 +/- 24.3, furthermore, MD simulations showed a high RMSD peak at 2.8 Å, RMSF were maximum at 3.5 Å with highest protein-protein interaction, the radius of gyration was stable throughout the simulation representing elastic stability along with a high energy interaction value of - 7882 kCal/mol. Moreover, the MMGBSA calculation results showed a notable release of binding free energy of - 68.96 kCal/mol demonstrating effective bond formation between the docked complex. These findings propose the effectiveness of Human Adenovirus 52 to treat cancer. The selected oncolytic Human Adenovirus 52 is a potential candidate for the target specific treatment of breast cancer through virotherapy. This computer-aided drug discovery presents significant potential in targeting cancer cells and would assist in the development of potent drug reagents for the cancer therapy.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan.
| | - Zainab Batool
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health Food Hygiene and Quality, University of Ioannina, Arta, Greece
| | - Khushbakht Javed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Nouman Ali
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | | | - Metab Alharbi
- Department of Pharmacology and Toxicology College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
17
|
Badder LM, Davies JA, Meniel VS, Marušková M, Salvador-Barbero B, Bayliss RJ, Phesse TJ, Hogan C, Parker AL. The αvβ6 integrin specific virotherapy, Ad5 NULL-A20.FCU1, selectively delivers potent "in-tumour" chemotherapy to pancreatic ductal adenocarcinoma. Br J Cancer 2024; 131:1694-1706. [PMID: 39369056 PMCID: PMC11555051 DOI: 10.1038/s41416-024-02869-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) represent an unmet clinical need. Approximately 90% of PDACs express high levels of αvβ6 integrin. We have previously described Ad5NULL-A20, an adenovirus vector with ablated native means of cell entry and retargeted to αvβ6 integrin by incorporation of an A20 peptide. METHODS Here, we incorporate suicide genes FCY1 and FCU1 encoding for cytosine deaminase (CDase) or a combination of CDase and UPRTase, capable of catalysing a non-toxic prodrug, 5-FC into the chemotherapeutic 5-FU and downstream metabolites, into replication-deficient Ad5 and Ad5NULL-A20. RESULTS We show that Ad5NULL-A20 enables the transfer of suicide genes to αvβ6 integrin-positive PDAC cells which, in combination with 5-FC, results in cell death in vitro which is further mediated by a bystander effect in non-transduced cells. Intratumoural delivery of Ad5NULL-A20.FCU1 in combination with intraperitoneal delivery of 5-FC further results in tumour growth inhibition in a cell line xenograft in vivo. Using clinically-relevant 3D organoid models, we show selective transduction and therapeutic efficacy of FCU1 transgenes in combination with 5-FC. CONCLUSION Taken together these data provide the preclinical rationale for combined Ad5NULL-A20.FCU1 plus 5-FC as a promising targeted therapy to mediate "in-tumour chemotherapy" and merits further investigation for the treatment of PDAC patients.
Collapse
Affiliation(s)
- Luned M Badder
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - James A Davies
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Valerie S Meniel
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
| | - Mahulena Marušková
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Beatriz Salvador-Barbero
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
| | - Rebecca J Bayliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Toby J Phesse
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Catherine Hogan
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ, UK
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
18
|
Fuchs J, Hübner J, Schmidt A, Irrgang P, Maier C, Vieira Antão A, Oltmanns F, Thirion C, Lapuente D, Tenbusch M. Evaluation of adenoviral vector Ad19a encoding RSV-F as novel vaccine against respiratory syncytial virus. NPJ Vaccines 2024; 9:205. [PMID: 39472590 PMCID: PMC11522487 DOI: 10.1038/s41541-024-01001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in infants and toddlers. Since natural infections do not induce persistent immunity, there is the need of vaccines providing long-term protection. Here, we evaluated a new adenoviral vector (rAd) vaccine based on the rare serotype rAd19a and compared the immunogenicity and efficacy to the highly immunogenic rAd5. Given as an intranasal boost in DNA primed mice, both vectors encoding the F protein provided efficient protection against a subsequent RSV infection. However, intramuscular immunization with rAd19a vectors provoked vaccine-enhanced disease after RSV infection compared to non-vaccinated animals. While mucosal IgA antibodies and tissue-resident memory T-cells in intranasally vaccinated mice rapidly control RSV replication, a strong anamnestic systemic T-cell response in absence of local immunity might be the reason for immune-mediated enhanced disease. Our study highlighted the potential benefits of developing effective mucosal against respiratory pathogens.
Collapse
Affiliation(s)
- Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Julian Hübner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Anna Schmidt
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Ana Vieira Antão
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany.
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054, Erlangen, Germany.
| |
Collapse
|
19
|
Koizumi N, Hirai T, Kano J, Sato A, Suzuki Y, Sasaki A, Nomura T, Utoguchi N. Utilizing Adenovirus Knob Proteins as Carriers in Cancer Gene Therapy Amidst the Presence of Anti-Knob Antibodies. Int J Mol Sci 2024; 25:10679. [PMID: 39409008 PMCID: PMC11476472 DOI: 10.3390/ijms251910679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Numerous gene therapy drugs for cancer have received global approval, yet their efficacy against solid tumors remains inadequate. Our previous research indicated that the fiber protein, a component of the adenovirus capsid, can propagate from infected cells to neighboring cells that express the adenovirus receptor. We hypothesize that merging this fiber protein with an anti-cancer protein could enable the anti-cancer protein to disseminate around the transfected cells, presenting a novel approach to cancer gene therapy. In our study, we discovered that the knob region of the adenovirus type 5 fiber protein is the smallest unit capable of spreading to adjacent cells in a receptor-specific manner. We also showed that the recombinant knob protein infiltrates cells after dispersing to surrounding cells. To assess the potential of the knob protein to augment gene therapy for solid tumors in mice, we expressed a fusion gene of the A subunit of cytotoxic cholera toxin and the knob region in mouse tumors. We found that this fusion protein only inhibited tumor growth in receptor-expressing mouse melanomas, and this inhibitory effect persisted even in mice with anti-knob antibodies. Our study's findings propose a novel cancer gene therapy strategy that enhances therapeutic effects by specifically delivering therapeutic proteins, expressed from in vivo administered genes, to target molecules. This outcome offers a fresh perspective on gene therapy for solid cancers, and we anticipate that knob proteins will serve as a platform for this method.
Collapse
Affiliation(s)
- Naoya Koizumi
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| | - Takamasa Hirai
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki 210-9501, Japan
| | - Junpei Kano
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| | - Anna Sato
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| | - Yurika Suzuki
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| | - Arisa Sasaki
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| | - Tetsuya Nomura
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| | - Naoki Utoguchi
- Laboratory of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo 194-8543, Japan; (T.H.); (T.N.); (N.U.)
| |
Collapse
|
20
|
Alekseeva ON, Hoa LT, Vorobyev PO, Kochetkov DV, Gumennaya YD, Naberezhnaya ER, Chuvashov DO, Ivanov AV, Chumakov PM, Lipatova AV. Receptors and Host Factors for Enterovirus Infection: Implications for Cancer Therapy. Cancers (Basel) 2024; 16:3139. [PMID: 39335111 PMCID: PMC11430599 DOI: 10.3390/cancers16183139] [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: 05/08/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Enteroviruses, with their diverse clinical manifestations ranging from mild or asymptomatic infections to severe diseases such as poliomyelitis and viral myocarditis, present a public health threat. However, they can also be used as oncolytic agents. This review shows the intricate relationship between enteroviruses and host cell factors. Enteroviruses utilize specific receptors and coreceptors for cell entry that are critical for infection and subsequent viral replication. These receptors, many of which are glycoproteins, facilitate virus binding, capsid destabilization, and internalization into cells, and their expression defines virus tropism towards various types of cells. Since enteroviruses can exploit different receptors, they have high oncolytic potential for personalized cancer therapy, as exemplified by the antitumor activity of certain enterovirus strains including the bioselected non-pathogenic Echovirus type 7/Rigvir, approved for melanoma treatment. Dissecting the roles of individual receptors in the entry of enteroviruses can provide valuable insights into their potential in cancer therapy. This review discusses the application of gene-targeting techniques such as CRISPR/Cas9 technology to investigate the impact of the loss of a particular receptor on the attachment of the virus and its subsequent internalization. It also summarizes the data on their expression in various types of cancer. By understanding how enteroviruses interact with specific cellular receptors, researchers can develop more effective regimens of treatment, offering hope for more targeted and efficient therapeutic strategies.
Collapse
Affiliation(s)
- Olga N. Alekseeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Le T. Hoa
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pavel O. Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Dmitriy V. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Yana D. Gumennaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Elizaveta R. Naberezhnaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Denis O. Chuvashov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| |
Collapse
|
21
|
Bliss CM, Hulin-Curtis SL, Williams M, Marušková M, Davies JA, Statkute E, Baker AT, Stack L, Kerstetter L, Kerr-Jones LE, Milward KF, Russell G, George SJ, Badder LM, Stanton RJ, Coughlan L, Humphreys IR, Parker AL. A pseudotyped adenovirus serotype 5 vector with serotype 49 fiber knob is an effective vector for vaccine and gene therapy applications. Mol Ther Methods Clin Dev 2024; 32:101308. [PMID: 39206304 PMCID: PMC11357811 DOI: 10.1016/j.omtm.2024.101308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Adenoviruses (Ads) have demonstrated significant success as replication-deficient (RD) viral vectored vaccines, as well as broad potential across gene therapy and cancer therapy. Ad vectors transduce human cells via direct interactions between the viral fiber knob and cell surface receptors, with secondary cellular integrin interactions. Ad receptor usage is diverse across the extensive phylogeny. Commonly studied human Ad serotype 5 (Ad5), and chimpanzee Ad-derived vector "ChAdOx1" in licensed ChAdOx1 nCoV-19 vaccine, both form primary interactions with the coxsackie and adenovirus receptor (CAR), which is expressed on human epithelial cells and erythrocytes. CAR usage is suboptimal for targeted gene delivery to cells with low/negative CAR expression, including human dendritic cells (DCs) and vascular smooth muscle cells (VSMCs). We evaluated the performance of an RD Ad5 vector pseudotyped with the fiber knob of human Ad serotype 49, termed Ad5/49K vector. Ad5/49K demonstrated superior transduction of murine and human DCs over Ad5, which translated into significantly increased T cell immunogenicity when evaluated in a mouse cancer vaccine model using 5T4 tumor-associated antigen. Additionally, Ad5/49K exhibited enhanced transduction of primary human VSMCs. These data highlight the potential of Ad5/49K vector for both vascular gene therapy applications and as a potent vaccine vector.
Collapse
Affiliation(s)
- Carly M. Bliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Sarah L. Hulin-Curtis
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Marta Williams
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Mahulena Marušková
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - James A. Davies
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Evelina Statkute
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alexander T. Baker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Louise Stack
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Lucas Kerstetter
- University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, MD 21201, USA
| | - Lauren E. Kerr-Jones
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Kate F. Milward
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Gabrielle Russell
- University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, MD 21201, USA
| | - Sarah J. George
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Luned M. Badder
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Richard J. Stanton
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Lynda Coughlan
- University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, MD 21201, USA
- University of Maryland School of Medicine, Center for Vaccine Development and Global Health, Baltimore, MD 21201, USA
| | - Ian R. Humphreys
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alan L. Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
22
|
Longo C, Saito M, Castro PT, Traina E, Werner H, Elito Júnior J, Araujo Júnior E. Coxsackievirus Group B Infections during Pregnancy: An Updated Literature Review. J Clin Med 2024; 13:4922. [PMID: 39201064 PMCID: PMC11355224 DOI: 10.3390/jcm13164922] [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: 06/22/2024] [Revised: 07/16/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
Coxsackievirus group B (CVB), a member of the Picornaviridae family and enterovirus genus, poses risks during pregnancy due to its potential to cause severe fetal and neonatal infections. Transmission primarily occurs through fecal-oral routes, with infections peaking mostly in warmer months. Vertical transmission to the fetus can lead to conditions such as myocarditis, encephalitis, and systemic neonatal disease, presenting clinically as severe myocardial syndromes and neurological deficits. Diagnostic challenges include detecting asymptomatic maternal infections and conducting in utero assessments using advanced techniques like RT-PCR from amniotic fluid samples. Morbidity and mortality associated with congenital CVB infections are notable, linked to preterm delivery, fetal growth restriction, and potential long-term health impacts such as type 1 diabetes mellitus and structural cardiac anomalies. Current treatments are limited to supportive care, with emerging therapies showing promise but requiring further study for efficacy in utero. Preventive measures focus on infection control and hygiene to mitigate transmission risks, which are crucial especially during pregnancy. Future research should aim to fill knowledge gaps in epidemiology, improve diagnostic capabilities, and develop targeted interventions to enhance maternal and fetal outcomes.
Collapse
Affiliation(s)
- Carolina Longo
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (C.L.); (E.T.); (J.E.J.)
| | - Mauricio Saito
- CONCEPTUS—Fetal Medicine Center, São Paulo 04001-084, SP, Brazil;
| | - Pedro Teixeira Castro
- Department of Fetal Medicine, Biodesign Laboratory DASA/PUC, Rio de Janeiro 22451-900, RJ, Brazil; (P.T.C.); (H.W.)
| | - Evelyn Traina
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (C.L.); (E.T.); (J.E.J.)
| | - Heron Werner
- Department of Fetal Medicine, Biodesign Laboratory DASA/PUC, Rio de Janeiro 22451-900, RJ, Brazil; (P.T.C.); (H.W.)
| | - Julio Elito Júnior
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (C.L.); (E.T.); (J.E.J.)
| | - Edward Araujo Júnior
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (C.L.); (E.T.); (J.E.J.)
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil
| |
Collapse
|
23
|
Mei S, Peng S, Vong EG, Zhan J. A PD-L1 tropism-expanded oncolytic adenovirus enhanced gene delivery efficiency and anti-tumor effects. Int Immunopharmacol 2024; 137:112393. [PMID: 38852522 DOI: 10.1016/j.intimp.2024.112393] [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: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Recombinant adenovirus serotype 5 (Ad5)-mediated virotherapy is a maturing technique in cancer treatment. However, the utility of adenovirus (Ad) has been limited by low expression of coxsackievirus and adenovirus receptor (CAR) in cancer cells resulting in poor infectivity of Ads. To overcome the problem, we aimed to develop a novel tropism-modified oncolytic adenovirus, ZD55-F-HI-sPD-1-EGFP, which contains the epitope of PD-1 (70-77aa) at the HI-loop of Ad fiber. Trimerization of Fiber-sPD-1 was confirmed by immunoblot analysis. ZD55-F-HI-sPD-1-EGFP shows a remarkable improvement in viral infection rate and gene transduction efficiency in the PD-L1-positive cancer cells. Competition assays with a PD-L1 protein reveals that cell internalization of ZD55-F-HI-sPD-1-EGFP is mediated by both CAR and PD-L1 at a high dose. The progeny virus production capacity showed that sPD-1 incorporated fiber-modified oncolytic Ad replication was not affected. Furthermore, treating with ZD55-F-HI-sPD-1-EGFP significantly increased viral infection rate and enhanced anti-tumor effect in vivo. This study demonstrates that the strategy to expand tropism of oncolytic Ad may significantly improve therapeutic profile for cancer treatment.
Collapse
Affiliation(s)
- Shengsheng Mei
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shanshan Peng
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Eu Gene Vong
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jinbiao Zhan
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
24
|
Dong Y, Shao E, Li S, Wang R, Wang D, Wang L, Yang H, He Y, Luan T, Chen Y, Wang Y, Lin L, Wang Y, Zhong Z, Zhao W. Baicalein suppresses Coxsackievirus B3 replication by inhibiting caspase-1 and viral protease 2A. Virol Sin 2024; 39:685-693. [PMID: 39025463 PMCID: PMC11401470 DOI: 10.1016/j.virs.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
Myocarditis is an inflammatory disease of the cardiac muscle and one of the primary causes of dilated cardiomyopathy. Group B coxsackievirus (CVB) is one of the leading causative pathogens of viral myocarditis, which primarily affects children and young adults. Due to the lack of vaccines, the development of antiviral medicines is crucial to controlling CVB infection and the progression of myocarditis. In this study, we investigated the antiviral effect of baicalein, a flavonoid extracted from Scutellaria baicaleinsis. Our results demonstrated that baicalein treatment significantly reduced cytopathic effect and increased cell viability in CVB3-infected cells. In addition, significant reductions in viral protein 3D, viral RNA, and viral particles were observed in CVB3-infected cells treated with baicalein. We found that baicalein exerted its inhibitory effect in the early stages of CVB3 infection. Baicalein also suppressed viral replication in the myocardium and effectively alleviated myocarditis induced by CVB3 infection. Our study revealed that baicalein exerts its antiviral effect by inhibiting the activity of caspase-1 and viral protease 2A. Taken together, our findings demonstrate that baicalein has antiviral activity against CVB3 infection and may serve as a potential therapeutic option for the myocarditis caused by enterovirus infection.
Collapse
Affiliation(s)
- Yanyan Dong
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Enze Shao
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Siwei Li
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Ruiqi Wang
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Dan Wang
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Lixin Wang
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Hong Yang
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Yingxia He
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Tian Luan
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Yang Chen
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Yao Wang
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Lexun Lin
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Yan Wang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Zhaohua Zhong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China.
| | - Wenran Zhao
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
25
|
Bouazzaoui A, Abdellatif AA. Vaccine delivery systems and administration routes: Advanced biotechnological techniques to improve the immunization efficacy. Vaccine X 2024; 19:100500. [PMID: 38873639 PMCID: PMC11170481 DOI: 10.1016/j.jvacx.2024.100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/21/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Since the first use of vaccine tell the last COVID-19 pandemic caused by spread of SARS-CoV-2 worldwide, the use of advanced biotechnological techniques has accelerated the development of different types and methods for immunization. The last pandemic showed that the nucleic acid-based vaccine, especially mRNA, has an advantage in terms of development time; however, it showed a very critical drawback namely, the higher costs when compared to other strategies, and its inability to protect against new variants. This showed the need of more improvement to reach a better delivery and efficacy. In this review we will describe different vaccine delivery systems including, the most used viral vector, and also variable strategies for delivering of nucleic acid-based vaccines especially lipid-based nanoparticles formulation, polymersomes, electroporation and also the new powerful tools for the delivery of mRNA, which is based on the use of cell-penetrating peptides (CPPs). Additionally, we will also discuss the main challenges associated with each system. Finlay, the efficacy and safety of the vaccines depends not only on the formulations and delivery systems, but also the dosage and route of administration are also important players, therefore we will see the different routes for the vaccine administration including traditionally routes (intramuscular, Transdermal, subcutaneous), oral inhalation or via nasal mucosa, and will describe the advantages and disadvantage of each administration route.
Collapse
Affiliation(s)
- Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
- Science and Technology Unit, Umm Al Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Ahmed A.H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, 51452 Qassim, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, 71524 Assiut, Egypt
| |
Collapse
|
26
|
Ayyub M, Thomas JG, Hodeify R. An Overview of the Characteristics, Pathogenesis, Epidemiology, and Detection of Human Enterovirus in the Arabian Gulf Region. Viruses 2024; 16:1187. [PMID: 39205162 PMCID: PMC11359295 DOI: 10.3390/v16081187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Enteroviruses are RNA viruses that initiate infections through the gastrointestinal (GI) tract and are associated with enteric illness in individuals of all ages. Most serious infections of enteroviruses are in infants and young children where it is the common cause of aseptic meningitis and other systemic diseases, leading to a high mortality rate. Enteroviruses belong to the small non-enveloped family of the Picornaviridae family. The virus can spread mainly through fecal-oral and respiratory routes. In the Arabian Gulf countries, the incidence of enteroviral infections is only restricted to a few reports, and thus, knowledge of the epidemiology, characteristics, and pathogenesis of the virus in the gulf countries remains scarce. In this minireview, we sought to provide an overview of the characteristics of enterovirus and its pathogenesis, in addition to gathering the reports of enterovirus infection prevalence in Gulf Cooperation Council (GCC) countries. We also present a summary of the common methods used in its detection.
Collapse
Affiliation(s)
| | | | - Rawad Hodeify
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah 72603, United Arab Emirates; (M.A.); (J.G.T.)
| |
Collapse
|
27
|
Nikrad JA, Galvin RT, Sheehy MM, Novacek EL, Jacobsen KL, Corbière SMAS, Beckmann PJ, Jubenville TA, Yamamoto M, Largaespada DA. Conditionally replicative adenovirus as a therapy for malignant peripheral nerve sheath tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200783. [PMID: 38595983 PMCID: PMC10959710 DOI: 10.1016/j.omton.2024.200783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Oncolytic adenoviruses (Ads) stand out as a promising strategy for the targeted infection and lysis of tumor cells, with well-established clinical utility across various malignancies. This study delves into the therapeutic potential of oncolytic Ads in the context of neurofibromatosis type 1 (NF1)-associated malignant peripheral nerve sheath tumors (MPNSTs). Specifically, we evaluate conditionally replicative adenoviruses (CRAds) driven by the cyclooxygenase 2 (COX2) promoter, as selective agents against MPNSTs, demonstrating their preferential targeting of MPNST cells compared with non-malignant Schwann cell control. COX2-driven CRAds, particularly those with modified fiber-knobs exhibit superior binding affinity toward MPNST cells and demonstrate efficient and preferential replication and lysis of MPNST cells, with minimal impact on non-malignant control cells. In vivo experiments involving intratumoral CRAd injections in immunocompromised mice with human MPNST xenografts significantly extend survival and reduce tumor growth rate compared with controls. Moreover, in immunocompetent mouse models with MPNST-like allografts, CRAd injections induce a robust infiltration of CD8+ T cells into the tumor microenvironment (TME), indicating the potential to promote a pro-inflammatory response. These findings underscore oncolytic Ads as promising, selective, and minimally toxic agents for MPNST therapy, warranting further exploration.
Collapse
Affiliation(s)
- Julia A Nikrad
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Robert T Galvin
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Mackenzie M Sheehy
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Ethan L Novacek
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Kari L Jacobsen
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Stanislas M A S Corbière
- Institute for Research in Immunology and Cancer, Université de Montréal, 2950 Chemin de Polytechnique Marcelle-Coutu Pavilion, Montréal, QC H3T1J4, Canada
| | - Pauline J Beckmann
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Tyler A Jubenville
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
28
|
Wallace R, Bliss CM, Parker AL. The Immune System-A Double-Edged Sword for Adenovirus-Based Therapies. Viruses 2024; 16:973. [PMID: 38932265 PMCID: PMC11209478 DOI: 10.3390/v16060973] [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: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Pathogenic adenovirus (Ad) infections are widespread but typically mild and transient, except in the immunocompromised. As vectors for gene therapy, vaccine, and oncology applications, Ad-based platforms offer advantages, including ease of genetic manipulation, scale of production, and well-established safety profiles, making them attractive tools for therapeutic development. However, the immune system often poses a significant challenge that must be overcome for adenovirus-based therapies to be truly efficacious. Both pre-existing anti-Ad immunity in the population as well as the rapid development of an immune response against engineered adenoviral vectors can have detrimental effects on the downstream impact of an adenovirus-based therapeutic. This review focuses on the different challenges posed, including pre-existing natural immunity and anti-vector immunity induced by a therapeutic, in the context of innate and adaptive immune responses. We summarise different approaches developed with the aim of tackling these problems, as well as their outcomes and potential future applications.
Collapse
Affiliation(s)
- Rebecca Wallace
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
| | - Carly M. Bliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alan L. Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
29
|
Zhang Y, Qian L, Chen K, Gu S, Meng Z, Wang J, Li Y, Wang P. Oncolytic adenovirus in treating malignant ascites: A phase II trial and longitudinal single-cell study. Mol Ther 2024; 32:2000-2020. [PMID: 38659226 PMCID: PMC11184408 DOI: 10.1016/j.ymthe.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/24/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024] Open
Abstract
Malignant ascites is a common complication resulting from the peritoneal spread of malignancies, and currently lacks effective treatments. We conducted a phase II trial (NCT04771676) to investigate the efficacy and safety of oncolytic adenovirus H101 and virotherapy-induced immune response in 25 patients with malignant ascites. Oncolytic virotherapy achieved an increased median time to repeat paracentesis of 45 days (95% confidence interval 16.5-73.5 days), compared with the preset control value of 13 days. Therapy was well-tolerated, with pyrexia, fatigue, nausea, and abdominal pain as the most common toxicities. Longitudinal single-cell profiling identified marked oncolysis, early virus replication, and enhanced CD8+ T cells-macrophages immune checkpoint crosstalk, especially in responsive patients. H101 also triggered a proliferative burst of CXCR6+ and GZMK+CD8+ T cells with promoted tumor-specific cytotoxicity. Further establishment of oncolytic virus-induced T cell expansion signature (OiTE) implicated the potential benefits for H101-responsive patients from subsequent anti-PD(L)1 therapy. Patients with upregulated immune-signaling pathways in tumor cells and a higher proportion of CLEC10A+ dendritic cells and GZMK+CD8+ T cells at baseline showed a superior response to H101 treatment. Our study demonstrates promising clinical responses and tolerability of oncolytic adenovirus in treating malignant ascites and provides insights into the relevant cellular processes following oncolytic virotherapy.
Collapse
Affiliation(s)
- Yalei Zhang
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Qian
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kun Chen
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sijia Gu
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 300032, China.
| | - Ye Li
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Peng Wang
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Vicenzetto C, Giordani AS, Menghi C, Baritussio A, Peloso Cattini MG, Pontara E, Bison E, Rizzo S, De Gaspari M, Basso C, Thiene G, Iliceto S, Marcolongo R, Caforio ALP. The Role of the Immune System in Pathobiology and Therapy of Myocarditis: A Review. Biomedicines 2024; 12:1156. [PMID: 38927363 PMCID: PMC11200507 DOI: 10.3390/biomedicines12061156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/18/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The role of the immune system in myocarditis onset and progression involves a range of complex cellular and molecular pathways. Both innate and adaptive immunity contribute to myocarditis pathogenesis, regardless of its infectious or non-infectious nature and across different histological and clinical subtypes. The heterogeneity of myocarditis etiologies and molecular effectors is one of the determinants of its clinical variability, manifesting as a spectrum of disease phenotype and progression. This spectrum ranges from a fulminant presentation with spontaneous recovery to a slowly progressing, refractory heart failure with ventricular dysfunction, to arrhythmic storm and sudden cardiac death. In this review, we first examine the updated definition and classification of myocarditis at clinical, biomolecular and histopathological levels. We then discuss recent insights on the role of specific immune cell populations in myocarditis pathogenesis, with particular emphasis on established or potential therapeutic applications. Besides the well-known immunosuppressive agents, whose efficacy has been already demonstrated in human clinical trials, we discuss the immunomodulatory effects of other drugs commonly used in clinical practice for myocarditis management. The immunological complexity of myocarditis, while presenting a challenge to simplistic understanding, also represents an opportunity for the development of different therapeutic approaches with promising results.
Collapse
Affiliation(s)
- Cristina Vicenzetto
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Andrea Silvio Giordani
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Caterina Menghi
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Anna Baritussio
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Maria Grazia Peloso Cattini
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Elena Pontara
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Elisa Bison
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Stefania Rizzo
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Monica De Gaspari
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Cristina Basso
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Gaetano Thiene
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Sabino Iliceto
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Renzo Marcolongo
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Alida Linda Patrizia Caforio
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| |
Collapse
|
31
|
Nemerow GR. Integrin-Targeting Strategies for Adenovirus Gene Therapy. Viruses 2024; 16:770. [PMID: 38793651 PMCID: PMC11125847 DOI: 10.3390/v16050770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Numerous human adenovirus (AdV) types are endowed with arginine-glycine-aspartic acid (RGD) sequences that enable them to recognize vitronectin-binding (αv) integrins. These RGD-binding cell receptors mediate AdV entry into host cells, a crucial early step in virus infection. Integrin interactions with adenoviruses not only initiate receptor-mediated endocytosis but also facilitate AdV capsid disassembly, a prerequisite for membrane penetration by AdV protein VI. This review discusses fundamental aspects of AdV-host interactions mediated by integrins. Recent efforts to re-engineer AdV vectors and non-viral nanoparticles to target αv integrins for bioimaging and the eradication of cancer cells will also be discussed.
Collapse
Affiliation(s)
- Glen R Nemerow
- Department of Immunology, The Scripps Research Institute, 10666 North Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
32
|
Nguyen TP, Otani T, Tsutsumi M, Kinoshita N, Fujiwara S, Nemoto T, Fujimori T, Furuse M. Tight junction membrane proteins regulate the mechanical resistance of the apical junctional complex. J Cell Biol 2024; 223:e202307104. [PMID: 38517380 PMCID: PMC10959758 DOI: 10.1083/jcb.202307104] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Epithelia must be able to resist mechanical force to preserve tissue integrity. While intercellular junctions are known to be important for the mechanical resistance of epithelia, the roles of tight junctions (TJs) remain to be established. We previously demonstrated that epithelial cells devoid of the TJ membrane proteins claudins and JAM-A completely lack TJs and exhibit focal breakages of their apical junctions. Here, we demonstrate that apical junctions fracture when claudin/JAM-A-deficient cells undergo spontaneous cell stretching. The junction fracture was accompanied by actin disorganization, and actin polymerization was required for apical junction integrity in the claudin/JAM-A-deficient cells. Further deletion of CAR resulted in the disruption of ZO-1 molecule ordering at cell junctions, accompanied by severe defects in apical junction integrity. These results demonstrate that TJ membrane proteins regulate the mechanical resistance of the apical junctional complex in epithelial cells.
Collapse
Affiliation(s)
- Thanh Phuong Nguyen
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi, Japan
| | - Motosuke Tsutsumi
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Noriyuki Kinoshita
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Sachiko Fujiwara
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tomomi Nemoto
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
33
|
Marquez-Martinez S, Salisch N, Serroyen J, Zahn R, Khan S. Peak transgene expression after intramuscular immunization of mice with adenovirus 26-based vector vaccines correlates with transgene-specific adaptive immune responses. PLoS One 2024; 19:e0299215. [PMID: 38626093 PMCID: PMC11020485 DOI: 10.1371/journal.pone.0299215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/07/2024] [Indexed: 04/18/2024] Open
Abstract
Non-replicating adenovirus-based vectors have been broadly used for the development of prophylactic vaccines in humans and are licensed for COVID-19 and Ebola virus disease prevention. Adenovirus-based vectored vaccines encode for one or more disease specific transgenes with the aim to induce protective immunity against the target disease. The magnitude and duration of transgene expression of adenovirus 5- based vectors (human type C) in the host are key factors influencing antigen presentation and adaptive immune responses. Here we characterize the magnitude, duration, and organ biodistribution of transgene expression after single intramuscular administration of adenovirus 26-based vector vaccines in mice and evaluate the differences with adenovirus 5-based vector vaccine to understand if this is universally applicable across serotypes. We demonstrate a correlation between peak transgene expression early after adenovirus 26-based vaccination and transgene-specific cellular and humoral immune responses for a model antigen and SARS-CoV-2 spike protein, independent of innate immune activation. Notably, the memory immune response was similar in mice immunized with adenovirus 26-based vaccine and adenovirus 5-based vaccine, despite the latter inducing a higher peak of transgene expression early after immunization and a longer duration of transgene expression. Together these results provide further insights into the mode of action of adenovirus 26-based vector vaccines.
Collapse
Affiliation(s)
| | - Nadine Salisch
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| | - Jan Serroyen
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| | - Roland Zahn
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| | - Selina Khan
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| |
Collapse
|
34
|
Wang Y, Zou X, Guo X, Zhang Z, Wang M, Hung T, Lu Z. Redirect Tropism of Fowl Adenovirus 4 Vector by Modifying Fiber2 with Variable Domain of Heavy-Chain Antibody. Genes (Basel) 2024; 15:467. [PMID: 38674401 PMCID: PMC11049955 DOI: 10.3390/genes15040467] [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: 03/07/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The variable domain of a heavy-chain antibody (VHH) has the potential to be used to redirect the cell tropism of adenoviral vectors. Here, we attempted to establish platforms to simplify the screening of VHHs for their specific targeting function when being incorporated into the fiber of adenovirus. Both fowl adenovirus 4 (FAdV-4) and simian adenovirus 1 (SAdV-1) have two types of fiber, one of which is dispensable for virus propagation and is a proper site for VHH display. An intermediate plasmid, pMD-FAV4Fs, was constructed as the start plasmid for FAdV-4 fiber2 modification. Foldon from phage T4 fibritin, a trigger for trimerization, was employed to bridge the tail/shaft domain of fiber2 and VHHs against human CD16A, a key membrane marker of natural killer (NK) cells. Through one step of restriction-assembly, the modified fiber2 was transferred to the adenoviral plasmid, which was linearized and transfected to packaging cells. Five FAdV-4 viruses carrying the GFP gene were finally rescued and amplified, with three VHHs being displayed. One recombinant virus, FAdV4FC21-EG, could hardly transduce human 293 or Jurkat cells. In contrast, when it was used at a multiplicity of infection of 1000 viral particles per cell, the transduction efficiency reached 51% or 34% for 293 or Jurkat cells expressing exogenous CD16A. Such a strategy of fiber modification was transplanted to the SAdV-1 vector to construct SAdV1FC28H-EG, which moderately transduced primary human NK cells while the parental virus transduced none. Collectively, we reformed the strategy of integrating VHH to fiber and established novel platforms for screening VHHs to construct adenoviral vectors with a specific tropism.
Collapse
Affiliation(s)
- Yongjin Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
| | - Xiaohui Zou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
| | - Xiaojuan Guo
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
| | - Zhichao Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
- School of Public Health, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Min Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
| | - Tao Hung
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
| | - Zhuozhuang Lu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China
| |
Collapse
|
35
|
Cheng S, Li M, Li C, Dai Y, Zhuo J, Wang J, Qian J, Hao Z. JAML inhibits colorectal carcinogenesis by modulating the tumor immune microenvironment. In Vitro Cell Dev Biol Anim 2024; 60:382-396. [PMID: 38625487 DOI: 10.1007/s11626-024-00881-8] [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: 11/24/2023] [Accepted: 02/09/2024] [Indexed: 04/17/2024]
Abstract
It is necessary to explore new targets for the treatment of colon adenocarcinoma (COAD) according to the tumor microenvironment. The expression levels of JAML and CXADR were analyzed by bioinformatics analysis and validation of clinical samples. JAML over-expression CD8+ T cell line was constructed, and the proliferation activity was detected by MTT. The production of inflammatory factors was detected by ELISA. The expression of immune checkpoint PD-1 and TIM-3 was detected by Western blot. The apoptosis level was detected by flow cytometry and apoptosis markers. The AOM/DSS mouse model of colorectal cancer was constructed. The expression levels of JAML, CXADR and PD-1 were detected by PCR and Western blot, and the proportion of CD8+ T cells and exhausted T cells were detected by flow cytometry. The expression levels of JAML and CXADR were significantly decreased in colon cancer tissues. Overexpression of JAML can promote the proliferation of T cells, secrete a variety of inflammatory factors. Overexpression of CXADR can reduce the proliferation of colorectal cancer cells, promote apoptosis, and down-regulate the migration and invasion ability of tumor cells. Both JAML agonists and PD-L1 inhibitors can effectively treat colorectal cancer, and the combined use of JAML agonists and PD-L1 inhibitors can enhance the effect. JAML can promote the proliferation and toxicity of CD8+ T cells and down-regulate the expression of immune checkpoints in colon cancer. CXADR can inhibit the proliferation of cancer cells and promote the apoptosis. JAML agonist can effectively treat colorectal cancer by regulating CD8+ T cells.
Collapse
Affiliation(s)
- Shiliang Cheng
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China.
| | - Meng Li
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China
| | - Chunguang Li
- Emergency Medicine Department, Shandong Provincial Third Hospital, Shandong University, Jinan, People's Republic of China
| | - Yonggang Dai
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China
| | - Jinhua Zhuo
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China
| | - Jue Wang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China
| | - Jingrong Qian
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China
| | - Zhihao Hao
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, People's Republic of China
| |
Collapse
|
36
|
Padget RL, Zeitz MJ, Blair GA, Wu X, North MD, Tanenbaum MT, Stanley KE, Phillips CM, King DR, Lamouille S, Gourdie RG, Hoeker GS, Swanger SA, Poelzing S, Smyth JW. Acute Adenoviral Infection Elicits an Arrhythmogenic Substrate Prior to Myocarditis. Circ Res 2024; 134:892-912. [PMID: 38415360 PMCID: PMC11003857 DOI: 10.1161/circresaha.122.322437] [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: 12/21/2022] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Viral cardiac infection represents a significant clinical challenge encompassing several etiological agents, disease stages, complex presentation, and a resulting lack of mechanistic understanding. Myocarditis is a major cause of sudden cardiac death in young adults, where current knowledge in the field is dominated by later disease phases and pathological immune responses. However, little is known regarding how infection can acutely induce an arrhythmogenic substrate before significant immune responses. Adenovirus is a leading cause of myocarditis, but due to species specificity, models of infection are lacking, and it is not understood how adenoviral infection may underlie sudden cardiac arrest. Mouse adenovirus type-3 was previously reported as cardiotropic, yet it has not been utilized to understand the mechanisms of cardiac infection and pathology. METHODS We have developed mouse adenovirus type-3 infection as a model to investigate acute cardiac infection and molecular alterations to the infected heart before an appreciable immune response or gross cardiomyopathy. RESULTS Optical mapping of infected hearts exposes decreases in conduction velocity concomitant with increased Cx43Ser368 phosphorylation, a residue known to regulate gap junction function. Hearts from animals harboring a phospho-null mutation at Cx43Ser368 are protected against mouse adenovirus type-3-induced conduction velocity slowing. Additional to gap junction alterations, patch clamping of mouse adenovirus type-3-infected adult mouse ventricular cardiomyocytes reveals prolonged action potential duration as a result of decreased IK1 and IKs current density. Turning to human systems, we find human adenovirus type-5 increases phosphorylation of Cx43Ser368 and disrupts synchrony in human induced pluripotent stem cell-derived cardiomyocytes, indicating common mechanisms with our mouse whole heart and adult cardiomyocyte data. CONCLUSIONS Together, these findings demonstrate that adenoviral infection creates an arrhythmogenic substrate through direct targeting of gap junction and ion channel function in the heart. Such alterations are known to precipitate arrhythmias and likely contribute to sudden cardiac death in acutely infected patients.
Collapse
Affiliation(s)
- Rachel L. Padget
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Michael J. Zeitz
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Grace A. Blair
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Xiaobo Wu
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Michael D. North
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | | | - Kari E. Stanley
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Chelsea M. Phillips
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - D. Ryan King
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Samy Lamouille
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Robert G. Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Gregory S. Hoeker
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Sharon A. Swanger
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Steven Poelzing
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - James W. Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
37
|
Dhillon A, Persson BD, Volkov AN, Sülzen H, Kádek A, Pompach P, Kereïche S, Lepšík M, Danskog K, Uetrecht C, Arnberg N, Zoll S. Structural insights into the interaction between adenovirus C5 hexon and human lactoferrin. J Virol 2024; 98:e0157623. [PMID: 38323814 PMCID: PMC10949841 DOI: 10.1128/jvi.01576-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/13/2024] [Indexed: 02/08/2024] Open
Abstract
Adenovirus (AdV) infection of the respiratory epithelium is common but poorly understood. Human AdV species C types, such as HAdV-C5, utilize the Coxsackie-adenovirus receptor (CAR) for attachment and subsequently integrins for entry. CAR and integrins are however located deep within the tight junctions in the mucosa where they would not be easily accessible. Recently, a model for CAR-independent AdV entry was proposed. In this model, human lactoferrin (hLF), an innate immune protein, aids the viral uptake into epithelial cells by mediating interactions between the major capsid protein, hexon, and yet unknown host cellular receptor(s). However, a detailed understanding of the molecular interactions driving this mechanism is lacking. Here, we present a new cryo-EM structure of HAdV-5C hexon at high resolution alongside a hybrid structure of HAdV-5C hexon complexed with human lactoferrin (hLF). These structures reveal the molecular determinants of the interaction between hLF and HAdV-C5 hexon. hLF engages hexon primarily via its N-terminal lactoferricin (Lfcin) region, interacting with hexon's hypervariable region 1 (HVR-1). Mutational analyses pinpoint critical Lfcin contacts and also identify additional regions within hLF that critically contribute to hexon binding. Our study sheds more light on the intricate mechanism by which HAdV-C5 utilizes soluble hLF/Lfcin for cellular entry. These findings hold promise for advancing gene therapy applications and inform vaccine development. IMPORTANCE Our study delves into the structural aspects of adenovirus (AdV) infections, specifically HAdV-C5 in the respiratory epithelium. It uncovers the molecular details of a novel pathway where human lactoferrin (hLF) interacts with the major capsid protein, hexon, facilitating viral entry, and bypassing traditional receptors such as CAR and integrins. The study's cryo-EM structures reveal how hLF engages hexon, primarily through its N-terminal lactoferricin (Lfcin) region and hexon's hypervariable region 1 (HVR-1). Mutational analyses identify critical Lfcin contacts and other regions within hLF vital for hexon binding. This structural insight sheds light on HAdV-C5's mechanism of utilizing soluble hLF/Lfcin for cellular entry, holding promise for gene therapy and vaccine development advancements in adenovirus research.
Collapse
Affiliation(s)
- Arun Dhillon
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | | | - Alexander N. Volkov
- VIB-VUB Center for Structural Biology, Flemish Institute of Biotechnology (VIB), Brussels, Belgium
- Jean Jeener NMR Centre, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hagen Sülzen
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Alan Kádek
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
- Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Petr Pompach
- Biotechnology and Biomedical Center of the Academy of Sciences and Charles University in Vestec, Vestec, Czech Republic
| | - Sami Kereïche
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Lepšík
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Katarina Danskog
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Charlotte Uetrecht
- Department of Health Sciences and Biomedicine, Faculty V: School of Life Sciences, CSSB Centre for Structural Systems Biology, Deutsches Elektronen Synchrotron DESY and Leibniz Institute of Virology, Hamburg, University of Siegen, Siegen, Germany
| | - Niklas Arnberg
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Sebastian Zoll
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
38
|
Wen J, Ke Z, Wang Y, Li Y, Zhang D, Mo X, Yin J, Shi C, Zhou W, Zheng S, Wang Q. Coxsackievirus and adenovirus receptor inhibits tilapia lake virus infection via binding to viral segment 8 and 10 encoded protein. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109438. [PMID: 38341116 DOI: 10.1016/j.fsi.2024.109438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
The global aquaculture industry of tilapia (Oreochromis niloticus) has been significantly impacted by the emergence of tilapia lake virus (TiLV). However, effective prevention and control measures are still not available due to a lack of unclear pathogenesis of TiLV. Our previous transcriptome found that coxsackievirus and adenovirus receptor (CAR) was in response to TiLV infection in tilapia. To explore the potential function of OnCAR, the effect of OnCAR on TiLV proliferation was analyzed in this study. The OnCAR open reading frame (ORF) sequence of tilapia was 516 bp in length that encoded 171 amino acids with an Ig-like domain and transmembrane region. The OnCAR gene showed widespread expression in all investigated tissues, with the highest levels in the heart. Moreover, the OnCAR gene in the liver and muscle of tilapia exhibited dynamic expression levels upon TiLV challenge. Subcellular localization analysis indicated that OnCAR protein was mainly localized on the membrane of tilapia brain (TiB) cells. Importantly, the gene transcripts, genome copy number, S8-encoded protein, cytopathic effect, and internalization of TiLV were obviously decreased in the TiB cells overexpressed with OnCAR, indicating that OnCAR could inhibit TiLV replication. Mechanically, OnCAR could interact with viral S8 and S10-encoded protein. To the best of our knowledge, OnCAR is the first potential anti-TiLV cellular surface molecular receptor discovered for inhibiting TiLV infection. This finding is beneficial for better understanding the antiviral mechanism of tilapia and lays a foundation for establishing effective prevention and control strategies against tilapia lake virus disease (TiLVD).
Collapse
Affiliation(s)
- Jing Wen
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China; College of Fisheries, Tianjin Agricultural University, Tianjin, China
| | - Zishan Ke
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Yingying Wang
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Yingying Li
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Defeng Zhang
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Xubing Mo
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Jiyuan Yin
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Cunbin Shi
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China
| | - Wenli Zhou
- College of Fisheries, Tianjin Agricultural University, Tianjin, China
| | - Shucheng Zheng
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China; State Key Lab of Marine Pollution, Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong, China.
| | - Qing Wang
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, Guangdong, China.
| |
Collapse
|
39
|
Scarsella L, Ehrke-Schulz E, Paulussen M, Thal SC, Ehrhardt A, Aydin M. Advances of Recombinant Adenoviral Vectors in Preclinical and Clinical Applications. Viruses 2024; 16:377. [PMID: 38543743 PMCID: PMC10974029 DOI: 10.3390/v16030377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 05/23/2024] Open
Abstract
Adenoviruses (Ad) have the potential to induce severe infections in vulnerable patient groups. Therefore, understanding Ad biology and antiviral processes is important to comprehend the signaling cascades during an infection and to initiate appropriate diagnostic and therapeutic interventions. In addition, Ad vector-based vaccines have revealed significant potential in generating robust immune protection and recombinant Ad vectors facilitate efficient gene transfer to treat genetic diseases and are used as oncolytic viruses to treat cancer. Continuous improvements in gene delivery capacity, coupled with advancements in production methods, have enabled widespread application in cancer therapy, vaccine development, and gene therapy on a large scale. This review provides a comprehensive overview of the virus biology, and several aspects of recombinant Ad vectors, as well as the development of Ad vector, are discussed. Moreover, we focus on those Ads that were used in preclinical and clinical applications including regenerative medicine, vaccine development, genome engineering, treatment of genetic diseases, and virotherapy in tumor treatment.
Collapse
Affiliation(s)
- Luca Scarsella
- Department of Anesthesiology, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Science (ZBAF), Department of Human Medicine, Faculty of Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Eric Ehrke-Schulz
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
| | - Michael Paulussen
- Chair of Pediatrics, University Children’s Hospital, Vestische Kinder- und Jugendklinik Datteln, Witten/Herdecke University, 45711 Datteln, Germany;
| | - Serge C. Thal
- Department of Anesthesiology, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
| | - Malik Aydin
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Science (ZBAF), Department of Human Medicine, Faculty of Medicine, Witten/Herdecke University, 58453 Witten, Germany
- Chair of Pediatrics, University Children’s Hospital, Vestische Kinder- und Jugendklinik Datteln, Witten/Herdecke University, 45711 Datteln, Germany;
- Institute for Medical Laboratory Diagnostics, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| |
Collapse
|
40
|
de Oliveira KG, Bång-Rudenstam A, Beyer S, Boukredine A, Talbot H, Governa V, Johansson MC, Månsson AS, Forsberg-Nilsson K, Bengzon J, Malmström J, Welinder C, Belting M. Decoding of the surfaceome and endocytome in primary glioblastoma cells identifies potential target antigens in the hypoxic tumor niche. Acta Neuropathol Commun 2024; 12:35. [PMID: 38414005 PMCID: PMC10898066 DOI: 10.1186/s40478-024-01740-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
Immunotherapies with antibody-drug-conjugates (ADC) and CAR-T cells, targeted at tumor surface antigens (surfaceome), currently revolutionize clinical oncology. However, target identification warrants a better understanding of the surfaceome and how it is modulated by the tumor microenvironment. Here, we decode the surfaceome and endocytome and its remodeling by hypoxic stress in glioblastoma (GBM), the most common and aggressive brain tumor in adults. We employed a comprehensive approach for global and dynamic profiling of the surfaceome and endocytosed (endocytome) proteins and their regulation by hypoxia in patient-derived GBM cultures. We found a heterogeneous surface-endocytome profile and a divergent response to hypoxia across GBM cultures. We provide a quantitative ranking of more than 600 surface resident and endocytosed proteins, and their regulation by hypoxia, serving as a resource to the cancer research community. As proof-of-concept, the established target antigen CD44 was identified as a commonly and abundantly expressed surface protein with high endocytic activity. Among hypoxia induced proteins, we reveal CXADR, CD47, CD81, BSG, and FXYD6 as potential targets of the stressed GBM niche. We could validate these findings by immunofluorescence analyses in patient tumors and by increased expression in the hypoxic core of GBM spheroids. Selected candidates were finally confronted by treatment studies, showing their high capacity for internalization and ADC delivery. Importantly, we highlight the limited correlation between transcriptomics and proteomics, emphasizing the critical role of membrane protein enrichment strategies and quantitative mass spectrometry. Our findings provide a comprehensive understanding of the surface-endocytome and its remodeling by hypoxia in GBM as a resource for exploration of targets for immunotherapeutic approaches in GBM.
Collapse
Affiliation(s)
- Kelin Gonçalves de Oliveira
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Anna Bång-Rudenstam
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Sarah Beyer
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Axel Boukredine
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Hugo Talbot
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Valeria Governa
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Maria C Johansson
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Ann-Sofie Månsson
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Division of Cancer and Stem Cells, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Johan Bengzon
- Department of Clinical Sciences, Section of Neurosurgery, Lund University, Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Charlotte Welinder
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden
| | - Mattias Belting
- Department of Clinical Sciences, Lund, Section of Oncology, Lund University, Barngatan 4, 221 85, Lund, Sweden.
- Department of Hematology, Oncology and Radiophysics, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
41
|
Słońska A, Miedzińska A, Chodkowski M, Bąska P, Mielnikow A, Bartak M, Bańbura MW, Cymerys J. Human Adenovirus Entry and Early Events during Infection of Primary Murine Neurons: Immunofluorescence Studies In Vitro. Pathogens 2024; 13:158. [PMID: 38392896 PMCID: PMC10892902 DOI: 10.3390/pathogens13020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Human adenovirus (HAdV) is a common pathogen, which can lead to various clinical symptoms and-in some cases-central nervous system (CNS) dysfunctions, such as encephalitis and meningitis. Although the initial events of virus entry have already been identified in various cell types, the mechanism of neuronal uptake of adenoviruses is relatively little understood. The aim of this study was to investigate early events during adenoviral infection, in particular to determine the connection between cellular coxsackievirus and adenovirus receptor (CAR), clathrin, caveolin, and early endosomal proteins (EEA1 and Rab5) with the entry of HAdVs into primary murine neurons in vitro. An immunofluorescence assay and confocal microscopy analysis were carried out to determine HAdV4, 5, and 7 correlation with CAR, clathrin, caveolin, and early endosomal proteins in neurons. The quantification of Pearson's coefficient between CAR and HAdVs indicated that the HAdV4 and HAdV5 types correlated with CAR and that the correlation was more substantial for HAdV5. Inhibition of clathrin-mediated endocytosis using chlorpromazine limited the infection with HAdV, whereas inhibition of caveolin-mediated endocytosis did not affect virus entry. Thus, the entry of tested HAdV types into neurons was most likely associated with clathrin but not caveolin. It was also demonstrated that HAdVs correlate with the Rab proteins (EEA1, Rab5) present in early vesicles, and the observed differences in the manner of correlation depended on the serotype of the virus. With our research, we strove to expand knowledge regarding the mechanism of HAdV entry into neurons, which may be beneficial for developing potential therapeutics in the future.
Collapse
Affiliation(s)
- Anna Słońska
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (A.M.); (A.M.); (M.B.); (M.W.B.); (J.C.)
| | - Aleksandra Miedzińska
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (A.M.); (A.M.); (M.B.); (M.W.B.); (J.C.)
| | - Marcin Chodkowski
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, 02-786 Warsaw, Poland;
| | - Aleksandra Mielnikow
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (A.M.); (A.M.); (M.B.); (M.W.B.); (J.C.)
| | - Michalina Bartak
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (A.M.); (A.M.); (M.B.); (M.W.B.); (J.C.)
| | - Marcin W. Bańbura
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (A.M.); (A.M.); (M.B.); (M.W.B.); (J.C.)
| | - Joanna Cymerys
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (A.M.); (A.M.); (M.B.); (M.W.B.); (J.C.)
| |
Collapse
|
42
|
Kolb AW, Chau VQ, Miller DL, Yannuzzi NA, Brandt CR. Phylogenetic and Recombination Analysis of Clinical Vitreous Humor-Derived Adenovirus Isolates Reveals Discordance Between Serotype and Phylogeny. Invest Ophthalmol Vis Sci 2024; 65:12. [PMID: 38319669 PMCID: PMC10854415 DOI: 10.1167/iovs.65.2.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/19/2024] [Indexed: 02/07/2024] Open
Abstract
Purpose To sequence, identify, and perform phylogenetic and recombination analysis on three clinical adenovirus samples taken from the vitreous humor at the Bascom Palmer Eye Institute. Methods The PacBio Sequel II was used to sequence the genomes of the three clinical adenovirus isolates. To identify the isolates, a full genome-based multiple sequence alignment (MSA) of 722 mastadenoviruses was generated using multiple alignment using fast Fourier transform (MAFFT). MAFFT was also used to generate genome-based human adenovirus B (HAdV-B) MSAs, as well as HAdV-B fiber, hexon, and penton protein-based MSAs. To examine recombination within HAdV-B, RF-Net 2 and Bootscan software programs were used. Results In the course of classifying three new atypical ocular adenovirus samples, taken from the vitreous humor, we found that all three isolates were HAdV-B species. The three Bascom Palmer HAdV-B genomes were then combined with over 300 HAdV-B genome sequences, including nine ocular HAdV-B genome sequences. Attempts to categorize the penton, hexon, and fiber serotypes using phylogeny of the three Bascom Palmer samples were inconclusive due to incongruence between serotype and phylogeny in the dataset. Recombination analysis using a subset of HAdV-B strains to generate a hybridization network detected recombination between nonhuman primate and human-derived strains, recombination between one HAdV-B strain and the HAdV-E outgroup, and limited recombination between the B1 and B2 clades. Conclusions The discordance between serotype and phylogeny detected in this study suggests that the current classification system does not accurately describe the natural history and phylogenetic relationships among adenoviruses.
Collapse
Affiliation(s)
- Aaron W. Kolb
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Viet Q. Chau
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, United States
| | - Darlene L. Miller
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, United States
| | - Nicolas A. Yannuzzi
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, United States
| | - Curtis R. Brandt
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
43
|
Yao J, Atasheva S, Wagner N, Di Paolo NC, Stewart PL, Shayakhmetov DM. Targeted, safe, and efficient gene delivery to human hematopoietic stem and progenitor cells in vivo using the engineered AVID adenovirus vector platform. Mol Ther 2024; 32:103-123. [PMID: 37919899 PMCID: PMC10787117 DOI: 10.1016/j.ymthe.2023.10.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
Targeted delivery and cell-type-specific expression of gene-editing proteins in various cell types in vivo represent major challenges for all viral and non-viral delivery platforms developed to date. Here, we describe the development and analysis of artificial vectors for intravascular delivery (AVIDs), an engineered adenovirus-based gene delivery platform that allows for highly targeted, safe, and efficient gene delivery to human hematopoietic stem and progenitor cells (HSPCs) in vivo after intravenous vector administration. Due to a set of refined structural modifications, intravenous administration of AVIDs did not trigger cytokine storm, hepatotoxicity, or thrombocytopenia. Single intravenous administration of AVIDs to humanized mice, grafted with human CD34+ cells, led to up to 20% transduction of CD34+CD38-CD45RA- HSPC subsets in the bone marrow. Importantly, targeted in vivo transduction of CD34+CD38-CD45RA-CD90-CD49f+ subsets, highly enriched for human hematopoietic stem cells (HSCs), reached up to 19%, which represented a 1,900-fold selectivity in gene delivery to HSC-enriched over lineage-committed CD34-negative cell populations. Because the AVID platform allows for regulated, cell-type-specific expression of gene-editing technologies as well as expression of immunomodulatory proteins to ensure persistence of corrected HSCs in vivo, the HSC-targeted AVID platform may enable development of curative therapies through in vivo gene correction in human HSCs after a single intravenous administration.
Collapse
Affiliation(s)
- Jia Yao
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Svetlana Atasheva
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nicole Wagner
- Cleveland Center for Membrane and Structural Biology, Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nelson C Di Paolo
- AdCure Bio, LLC, Century Spring West, 6000 Lake Forrest Drive, Atlanta, GA 30328, USA
| | - Phoebe L Stewart
- Cleveland Center for Membrane and Structural Biology, Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA; Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
44
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
45
|
Casasnovas JM. Virus-Receptor Interactions and Receptor-Mediated Virus Entry into Host Cells. Subcell Biochem 2024; 105:533-566. [PMID: 39738957 DOI: 10.1007/978-3-031-65187-8_15] [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] [Indexed: 01/02/2025]
Abstract
The virus particles described in the previous chapters of this book are vehicles that transmit the viral genome and the infection from cell to cell. To initiate the infective cycle, the viral genome must therefore translocate from the viral particle to the cell cytoplasm. Via distinct proteins or motifs in their outermost shell, the particles of animal viruses or bacteriophages attach initially to specific receptors on the host cell surface. These viral receptors thus mediate penetration of the viral genome inside the cell, where the intracellular infective cycle starts. The presence of these receptors on the cell surface is a principal determinant of virus-host tropism. Viruses can use diverse types of molecules to attach to and enter into cells. In addition, virus-receptor recognition can evolve over the course of an infection, and viral variants with distinct receptor-binding specificities and tropism can appear. The identification of viral receptors and the characterization of virus-receptor interactions have been major research goals in virology. In this chapter, we will describe, from a structural perspective, several virus-receptor interactions and the active role of receptor molecules in virus cell entry.
Collapse
Affiliation(s)
- José M Casasnovas
- Department of Macromolecular Structure, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
46
|
Trivedi PD, Byrne BJ, Corti M. Evolving Horizons: Adenovirus Vectors' Timeless Influence on Cancer, Gene Therapy and Vaccines. Viruses 2023; 15:2378. [PMID: 38140619 PMCID: PMC10747483 DOI: 10.3390/v15122378] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Efficient and targeted delivery of a DNA payload is vital for developing safe gene therapy. Owing to the recent success of commercial oncolytic vector and multiple COVID-19 vaccines, adenovirus vectors are back in the spotlight. Adenovirus vectors can be used in gene therapy by altering the wild-type virus and making it replication-defective; specific viral genes can be removed and replaced with a segment that holds a therapeutic gene, and this vector can be used as delivery vehicle for tissue specific gene delivery. Modified conditionally replicative-oncolytic adenoviruses target tumors exclusively and have been studied in clinical trials extensively. This comprehensive review seeks to offer a summary of adenovirus vectors, exploring their characteristics, genetic enhancements, and diverse applications in clinical and preclinical settings. A significant emphasis is placed on their crucial role in advancing cancer therapy and the latest breakthroughs in vaccine clinical trials for various diseases. Additionally, we tackle current challenges and future avenues for optimizing adenovirus vectors, promising to open new frontiers in the fields of cell and gene therapies.
Collapse
Affiliation(s)
| | | | - Manuela Corti
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA; (P.D.T.); (B.J.B.)
| |
Collapse
|
47
|
Dettmeyer R. [Lethal lymphocytic myocarditis-an underestimated diagnosis in infancy and childhood?]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:198-203. [PMID: 37987819 DOI: 10.1007/s00292-023-01279-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
In the past, histological diagnosis of (post-)viral myocarditis was based on the so-called Dallas criteria, which have been criticized because of high interobserver variability and sampling error. Immunohistochemical qualification and quantification of interstitial intramyocardial leucocytes was established and standard values concerning adults were published. Fatal casualties due to a viral myocarditis are rare as far as babies and children are concerned (sudden unexpected death in infancy; SUDI). Cases of sudden unexpected death in the first year of life are frequently regarded as sudden infant death syndrome (SIDS). To diagnose myocarditis when there are only single focal lymphocytic infiltrates in the myocardium, the number of samples taken by autopsy is relevant. But even in babies, immunohistochemical qualification and quantification of interstitial lymphocytes and macrophages can lead to standard values allowing diagnosis of myocarditis. Depending on the course of a viral infection, molecular pathological detection of viral genome in the myocardium is possible to support the diagnosis. Using the mentioned methods gradually, there are more cases of suspected SIDS, which are in fact cases of virus-induced myocarditis as cause of death. Primary enteroviruses (coxsackie viruses) and adenoviruses were found but also Epstein-Barr virus and PVB-19.
Collapse
Affiliation(s)
- R Dettmeyer
- Universitätsklinikum Gießen & Marburg, Justus-Liebig-Universität Gießen, Frankfurter Str. 58, 35392, Gießen, Deutschland.
| |
Collapse
|
48
|
Grand RJ. Pathogenicity and virulence of human adenovirus F41: Possible links to severe hepatitis in children. Virulence 2023; 14:2242544. [PMID: 37543996 PMCID: PMC10405776 DOI: 10.1080/21505594.2023.2242544] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023] Open
Abstract
Over 100 human adenoviruses (HAdVs) have been isolated and allocated to seven species, A-G. Species F comprises two members-HAdV-F40 and HAdV-F41. As their primary site of infection is the gastrointestinal tract they have been termed, with species A, enteric adenoviruses. HAdV-F40 and HAdV-F41 are a common cause of gastroenteritis and diarrhoea in children. Partly because of difficulties in propagating the viruses in the laboratory, due to their restrictions on growth in many cell lines, our knowledge of the properties of individual viral proteins is limited. However, the structure of HAdV-F41 has recently been determined by cryo-electron microscopy. The overall structure is similar to those of HAdV-C5 and HAdV-D26 although with some differences. The sequence and arrangement of the hexon hypervariable region 1 (HVR1) and the arrangement of the C-terminal region of protein IX differ. Variations in the penton base and hexon HVR1 may play a role in facilitating infection of intestinal cells by HAdV-F41. A unique feature of HAdV-F40 and F41, among human adenoviruses, is the presence and expression of two fibre genes, giving long and short fibre proteins. This may also contribute to the tropism of these viruses. HAdV-F41 has been linked to a recent outbreak of severe acute hepatitis "of unknown origin" in young children. Further investigation has shown a very high prevalence of adeno-associated virus-2 in the liver and/or plasma of some cohorts of patients. These observations have proved controversial as HAdV-F41 had not been reported to infect the liver and AAV-2 has generally been considered harmless.
Collapse
Affiliation(s)
- Roger J. Grand
- Institute for Cancer and Genomic Science, the Medical School, University of Birmingham, Birmingham, UK
| |
Collapse
|
49
|
Jetzer T, Studer L, Bieri M, Greber UF, Hemmi S. Engineered Human Adenoviruses of Species B and C Report Early, Intermediate Early, and Late Viral Gene Expression. Hum Gene Ther 2023; 34:1230-1247. [PMID: 37725579 DOI: 10.1089/hum.2023.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
Adenoviruses (AdVs) are being developed for oncolytic or vaccination therapy against existing and emerging conditions. Well-characterized replication-competent human and human primate AdVs expressing multiple payloads are desirable, but their replication in rodent models is limited. To score the timing of adenoviral gene expression in cell cultures, we developed fully replication-competent transcriptional reporter viruses for HAdV-C5, -B3, and -B35. The picornavirus-derived 2A sequence, which induces cotranslational peptide splitting and reinitiation (skipping), was linked to GFP and the fused sequence was inserted C-terminal of the early gene E1A, the intermediate early gene protein IX and the late fiber gene. The 2A peptide induced ribosomal skipping during translation of the messenger RNA (mRNA) and gave rise to GFP from the corresponding viral promoters, as shown by immunoblotting and flow cytometry analyses of human and rodent cells. In human cells, both species B and C AdV exhibited highest reporter expression for fiber, followed by protein IX and lowest for E1A. Inoculation with either HAdV-C5 or -B3/35 viruses encoding protein IX- or fiber-GFP gave rise to higher GFP levels in hamster than mouse cells. Remarkably, despite rather low 2A ribosomal skipping efficiency of ∼50% for E1A-2A-GFP, protein IX-2A-GFP, and fiber-2A-GFP, unprocessed protein IX-2A-GFP and fiber-2A-GFP fusion proteins were efficiently incorporated into HAdV-B3 virions, respectively. These data indicate that the B3 C-termini of protein IX and fiber can be considered for retargeting engineered oncolytic or vaccination vectors, or for antigen display. The variable expression levels of transgenes from different subviral promoters may be used to improve oncolytic AdV vectors expressing therapeutic genes.
Collapse
Affiliation(s)
- Tania Jetzer
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Lukas Studer
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Manuela Bieri
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Molecular Life Science Program, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Silvio Hemmi
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
50
|
Effantin G, Hograindleur MA, Fenel D, Fender P, Vassal-Stermann E. Toward the understanding of DSG2 and CD46 interaction with HAdV-11 fiber, a super-complex analysis. J Virol 2023; 97:e0091023. [PMID: 37921471 PMCID: PMC10688334 DOI: 10.1128/jvi.00910-23] [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: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 11/04/2023] Open
Abstract
IMPORTANCE The main limitation of oncolytic vectors is neutralization by blood components, which prevents intratumoral administration to patients. Enadenotucirev, a chimeric HAdV-11p/HAdV-3 adenovirus identified by bio-selection, is a low seroprevalence vector active against a broad range of human carcinoma cell lines. At this stage, there's still some uncertainty about tropism and primary receptor utilization by HAdV-11. However, this information is very important, as it has a direct influence on the effectiveness of HAdV-11-based vectors. The aim of this work is to determine which of the two receptors, DSG2 and CD46, is involved in the attachment of the virus to the host, and what role they play in the early stages of infection.
Collapse
Affiliation(s)
| | | | - Daphna Fenel
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Pascal Fender
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | |
Collapse
|