1
|
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
|
2
|
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
|
3
|
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
|
4
|
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: 11] [Impact Index Per Article: 11.0] [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
|
5
|
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
|
6
|
Dong T, Shah JR, Phung AT, Larson C, Sanchez AB, Aisagbonhi O, Blair SL, Oronsky B, Trogler WC, Reid T, Kummel AC. A Local and Abscopal Effect Observed with Liposomal Encapsulation of Intratumorally Injected Oncolytic Adenoviral Therapy. Cancers (Basel) 2023; 15:3157. [PMID: 37370769 PMCID: PMC10296131 DOI: 10.3390/cancers15123157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
This study evaluated the in vivo therapeutic efficacy of oncolytic serotype 5 adenovirus TAV255 in CAR-deficient tumors. In vitro experiments were performed with cell lines that expressed different levels of CAR (HEK293, A549, CT26, 4T1, and MCF-7). Low CAR cells, such as CT26, were poorly transduced by Ad in vitro unless the adenovirus was encapsulated in liposomes. However, the CT26 tumor in an immune-competent mouse model responded to the unencapsulated TAV255; 33% of the tumors were induced into complete remission, and mice with complete remission rejected the rechallenge with cancer cell injection. Encapsulation of TAV255 improves its therapeutic efficacy by transducing more CT26 cells, as expected from in vitro results. In a bilateral tumor model, nonencapsulated TAV255 reduced the growth rate of the locally treated tumors but had no effect on the growth rate of the distant tumor site. Conversely, encapsulated TAV255-infected CT26 induced a delayed growth rate of both the primary injected tumor and the distant tumor, consistent with a robust immune response. In vivo, intratumorally injected unencapsulated adenoviruses infect CAR-negative cells with only limited efficiency. However, unencapsulated adenoviruses robustly inhibit the growth of CAR-deficient tumors, an effect that constitutes an 'in situ vaccination' by stimulating cytotoxic T cells.
Collapse
Affiliation(s)
- Tao Dong
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Jaimin R. Shah
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Materials Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Abraham T. Phung
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | - Omonigho Aisagbonhi
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Sarah L. Blair
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
| | | | - William C. Trogler
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony Reid
- EpicentRx, Inc., La Jolla, CA 92037, USA
| | - Andrew C. Kummel
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Matthaeus C, Jüttner R, Gotthardt M, Rathjen FG. The IgCAM CAR Regulates Gap Junction-Mediated Coupling on Embryonic Cardiomyocytes and Affects Their Beating Frequency. Life (Basel) 2022; 13:14. [PMID: 36675963 PMCID: PMC9866089 DOI: 10.3390/life13010014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The IgCAM coxsackie-adenovirus receptor (CAR) is essential for embryonic heart development and electrical conduction in the mature heart. However, it is not well-understood how CAR exerts these effects at the cellular level. To address this question, we analyzed the spontaneous beating of cultured embryonic hearts and cardiomyocytes from wild type and CAR knockout (KO) embryos. Surprisingly, in the absence of the CAR, cultured cardiomyocytes showed increased frequencies of beating and calcium cycling. Increased beatings of heart organ cultures were also induced by the application of reagents that bind to the extracellular region of the CAR, such as the adenovirus fiber knob. However, the calcium cycling machinery, including calcium extrusion via SERCA2 and NCX, was not disrupted in CAR KO cells. In contrast, CAR KO cardiomyocytes displayed size increases but decreased in the total numbers of membrane-localized Cx43 clusters. This was accompanied by improved cell-cell coupling between CAR KO cells, as demonstrated by increased intercellular dye diffusion. Our data indicate that the CAR may modulate the localization and oligomerization of Cx43 at the plasma membrane, which could in turn influence electrical propagation between cardiomyocytes via gap junctions.
Collapse
Affiliation(s)
- Claudia Matthaeus
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
- Laboratory of Cellular Biophysics, NHLBI, NIH, 50 South Drive, Building 50 RM 3312, Bethesda, MD 20892, USA
| | - René Jüttner
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
| | - Michael Gotthardt
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
| | - Fritz G. Rathjen
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
| |
Collapse
|
8
|
Gresele P, Momi S, Marcucci R, Ramundo F, De Stefano V, Tripodi A. Interactions of adenoviruses with platelets and coagulation and the vaccine-induced immune thrombotic thrombocytopenia syndrome. Haematologica 2021; 106:3034-3045. [PMID: 34407607 PMCID: PMC8634187 DOI: 10.3324/haematol.2021.279289] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
The COVID-19 pandemic has had a heavy impact on global health and economy and vaccination remains the primary way of controlling the infection. During the ongoing vaccination campaign some unexpected thrombotic events have emerged in subjects who had recently received the AstraZeneca (Vaxzevria) vaccine or the Johnson and Johnson (Janssen) vaccine, two adenovirus vector-based vaccines. Epidemiological studies confirm that the observed/expected ratio of these unusual thromboses is abnormally increased, especially in women in fertile age. The characteristics of this complication, with venous thromboses at unusual sites, most frequently in the cerebral vein sinuses but also in splanchnic vessels, often with multiple associated thromboses, thrombocytopenia, and sometimes disseminated intravascular coagulation, are unique and the time course and tumultuous evolution are suggestive of an acute immunological reaction. Indeed, plateletactivating anti-PF4 antibodies have been detected in a large proportion of the affected patients. Several data suggest that adenoviruses may interact with platelets, the endothelium and the blood coagulation system. Here we review interactions between adenoviral vectors and the hemostatic system that are of possible relevance in vaccine-associated thrombotic thrombocytopenia syndrome. We systematically analyze the clinical data on the reported thrombotic complications of adenovirus-based therapeutics and discuss all the current hypotheses on the mechanisms triggering this novel syndrome. Although, considering current evidence, the benefit of vaccination clearly outweighs the potential risks, it is of paramount importance to fully unravel the mechanisms leading to vaccineassociated thrombotic thrombocytopenia syndrome and to identify prognostic factors through further research.
Collapse
Affiliation(s)
- Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia.
| | - Stefania Momi
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence; Atherothrombotic Center, AOU Careggi, Florence
| | - Francesco Ramundo
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli - IRCCS - Rome
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli - IRCCS - Rome
| | - Armando Tripodi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thromboses Center, Milan
| |
Collapse
|
9
|
Meecham A, Marshall J. Harnessing the power of foot-and-mouth-disease virus for targeting integrin alpha-v beta-6 for the therapy of cancer. Expert Opin Drug Discov 2021; 16:737-744. [PMID: 33533659 DOI: 10.1080/17460441.2021.1878143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/15/2021] [Indexed: 10/22/2022]
Abstract
Introduction: The integrin αvβ6 is a promising therapeutic target due to its limited expression in healthy tissue and significant overexpression in cancer and fibrosis. The peptide A20FMDV2, derived from the foot and mouth disease virus, is highly selective for αvβ6, and can be used therapeutically to target αvβ6 expressing cells.Areas covered: In this review, the authors discuss the logic that led to the discovery of A20FMDV2, the importance of its stereochemistry in receptor-binding, and the strategies employed to use it as a molecular-specific drug delivery system. These strategies include creating A20FMDV2-drug conjugates, genetically modifying oncolytic viruses to express A20FMDV2 and thus redirect their tropism to predominantly αvβ6 expressing cells, creation of A20FMDV2 expressing CAR T-cells, and modifying antibody tropism by inserting A20FMDV2 into the CDR3 loop.Expert opinion: αvβ6 is one of the most promising therapeutic targets in cancer and fibrosis discovered in the last few decades. The potential use of A20FMDV2 as a molecular-specific αvβ6-targeting agent is extremely promising, particularly when considering the success of the peptide and its variants in clinical imaging.
Collapse
Affiliation(s)
- Amelia Meecham
- Centre for Tumour Biology, Barts Cancer Institute-Cancer Research UK Centre of Excellence, Queen Mary University of London, Charterhouse Square London, UK
| | - John Marshall
- Centre for Tumour Biology, Barts Cancer Institute-Cancer Research UK Centre of Excellence, Queen Mary University of London, Charterhouse Square London, UK
| |
Collapse
|
10
|
Hartman R, Eilers BJ, Bollschweiler D, Munson-McGee JH, Engelhardt H, Young MJ, Lawrence CM. The Molecular Mechanism of Cellular Attachment for an Archaeal Virus. Structure 2019; 27:1634-1646.e3. [PMID: 31587916 DOI: 10.1016/j.str.2019.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/21/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
Sulfolobus turreted icosahedral virus (STIV) is a model archaeal virus and member of the PRD1-adenovirus lineage. Although STIV employs pyramidal lysis structures to exit the host, knowledge of the viral entry process is lacking. We therefore initiated studies on STIV attachment and entry. Negative stain and cryoelectron micrographs showed virion attachment to pili-like structures emanating from the Sulfolobus host. Tomographic reconstruction and sub-tomogram averaging revealed pili recognition by the STIV C381 turret protein. Specifically, the triple jelly roll structure of C381 determined by X-ray crystallography shows that pilus recognition is mediated by conserved surface residues in the second and third domains. In addition, the STIV petal protein (C557), when present, occludes the pili binding site, suggesting that it functions as a maturation protein. Combined, these results demonstrate a role for the namesake STIV turrets in initial cellular attachment and provide the first molecular model for viral attachment in the archaeal domain of life.
Collapse
Affiliation(s)
- Ross Hartman
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Brian J Eilers
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Daniel Bollschweiler
- Department of Molecular Structural Biology, Max-Planck-Institute for Biochemistry, Martinsried, Germany
| | - Jacob H Munson-McGee
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - Harald Engelhardt
- Department of Molecular Structural Biology, Max-Planck-Institute for Biochemistry, Martinsried, Germany
| | - Mark J Young
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA; Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT 59717, USA; The Thermal Biology Institute, Montana State University, Bozeman, MT 59717, USA.
| | - C Martin Lawrence
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; The Thermal Biology Institute, Montana State University, Bozeman, MT 59717, USA.
| |
Collapse
|
11
|
Le TMD, Jung BK, Li Y, Duong HTT, Nguyen TL, Hong JW, Yun CO, Lee DS. Physically crosslinked injectable hydrogels for long-term delivery of oncolytic adenoviruses for cancer treatment. Biomater Sci 2019; 7:4195-4207. [PMID: 31386700 DOI: 10.1039/c9bm00992b] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
A dual pH- and temperature-responsive physically crosslinked and injectable hydrogel system was developed for efficient and long-term delivery of oncolytic adenoviruses (Ads). Three different types of physically crosslinked hydrogels with different chemical compositions and properties were prepared. These hydrogels with good biocompatibility can be injected at pH 9.0 and room temperature and rapidly form a gel under body or tumor microenvironment conditions. Ads encapsulated in hydrogels were released gradually without burst release. Moreover, these physically crosslinked hydrogels provided a protective environment for Ads and maintained their bioactivity for a long period of time. Compared to naked Ads, Ads protected by these physically crosslinked hydrogels showed strong cytotoxicity to cancer cells even after 11 days. The Ad-loaded hydrogel system also exhibited enhanced and long-term antitumor therapeutic effects in human xenograft tumor models. Due to these outstanding properties, Ad-loaded injectable hydrogels might have potential for long-term cancer treatment.
Collapse
Affiliation(s)
- Thai Minh Duy Le
- School of Chemical Engineering and Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Bo-Kyeong Jung
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Yi Li
- School of Chemical Engineering and Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Huu Thuy Trang Duong
- School of Chemical Engineering and Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Thanh Loc Nguyen
- School of Chemical Engineering and Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Jin Woo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, 04763, Republic of Korea. and GeneMedicine Co., Ltd, Seoul, 04763, Republic of Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, 04763, Republic of Korea. and GeneMedicine Co., Ltd, Seoul, 04763, Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering and Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
12
|
Ongrádi J, Chatlynne LG, Tarcsai KR, Stercz B, Lakatos B, Pring-Åkerblom P, Gooss D, Nagy K, Ablashi DV. Adenovirus Isolated From a Cat Is Related to Human Adenovirus 1. Front Microbiol 2019; 10:1430. [PMID: 31293556 PMCID: PMC6603132 DOI: 10.3389/fmicb.2019.01430] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/06/2019] [Indexed: 01/03/2023] Open
Abstract
An adenovirus (AdV) has been isolated from the rectal swab of a domestic cat (Felis catus) and named feline adenovirus (FeAdV) isolate. It replicates and causes cytopathological effects in many human, feline, other mammalian cell lines that have both Coxsackie-adenovirus-receptor and integrins. Its antigens cross-react with anti-human adenovirus antibodies in immunofluorescence and immunocytochemistry assays. Electron microscopy revealed typical extracellular icosahedral particles and pseudo arrays inside cells. Sequence analysis of hexon and fiber genes indicates that this virus might belong to human adenovirus (HAdV) C species and might be a variant of type 1. In the fiber protein, three altered amino acids occur in the shaft; four altered residues are found in the knob region as compared to a European HAdV might be type 1 isolate (strain 1038, D11). One alteration affects amino acid 442 forming an RGS motif in an alanine rich region that might be an alternative way to bind integrins with subsequent internalization. Substitutions in the hexon sequence are silent. As compared to published HAdV sequences, the fiber is related to the original American prototype and recently described Taiwanese HAdV 1 isolates, but the hexon sequences are related to adenovirus isolates from France, Germany, Japan, and Taiwan. Serology carried out on FeAdV infected M426 cells indicates a prevalence of IgG in 80% of domestic cats in Delaware, United States. FeAdV isolate seems to be a recently recognized virus with possible pathogenic effects and, simultaneous human and feline infections are possible. Further molecular and biological characterization of this feline adenovirus isolate, as well as studies on both human and feline epidemiology and pathomechanisms, especially in endangered big cats, are warranted. FeAdV might have further practical advantages. Namely, it could be utilized in both human and feline AIDS research, developed into diagnostic tools, and gene therapy vectors in the near future.
Collapse
Affiliation(s)
- Joseph Ongrádi
- Department of Medical Microbiology, Semmelweis University, Budapest, Hungary.,National Institute of Dermato-Venereology, Budapest, Hungary
| | | | | | - Balázs Stercz
- Department of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | | | | | - Donald Gooss
- Selbyville Animal Hospital, Selbyville, DE, United States
| | - Károly Nagy
- Department of Medical Microbiology, Semmelweis University, Budapest, Hungary.,National Institute of Dermato-Venereology, Budapest, Hungary
| | | |
Collapse
|
13
|
Abstract
Synapse formation is mediated by a surprisingly large number and wide variety of genes encoding many different protein classes. One of the families increasingly implicated in synapse wiring is the immunoglobulin superfamily (IgSF). IgSF molecules are by definition any protein containing at least one Ig-like domain, making this family one of the most common protein classes encoded by the genome. Here, we review the emerging roles for IgSF molecules in synapse formation specifically in the vertebrate brain, focusing on examples from three classes of IgSF members: ( a) cell adhesion molecules, ( b) signaling molecules, and ( c) immune molecules expressed in the brain. The critical roles for IgSF members in regulating synapse formation may explain their extensive involvement in neuropsychiatric and neurodevelopmental disorders. Solving the IgSF code for synapse formation may reveal multiple new targets for rescuing IgSF-mediated deficits in synapse formation and, eventually, new treatments for psychiatric disorders caused by altered IgSF-induced synapse wiring.
Collapse
Affiliation(s)
- Scott Cameron
- Center for Neuroscience, University of California, Davis, California 95618, USA; ,
| | | |
Collapse
|
14
|
Stepanenko AA, Chekhonin VP. Tropism and transduction of oncolytic adenovirus 5 vectors in cancer therapy: Focus on fiber chimerism and mosaicism, hexon and pIX. Virus Res 2018; 257:40-51. [PMID: 30125593 DOI: 10.1016/j.virusres.2018.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 02/09/2023]
Abstract
The cellular internalization (infection of cells) of adenovirus 5 (Ad5) is mediated by the initial attachment of the globular knob domain of the capsid fiber protein to the cell surface coxsackievirus and adenovirus receptor (CAR), then followed by the interaction of the virus penton base proteins with cellular integrins. In tumors, there is a substantial intra- and intertumoral variability in CAR expression. The CAR-negative cells generally exhibit very low infectability. Since the fiber knob is a primary mediator of Ad5 binding to the cell surface, improved infectivity of Ad5-based vectors as oncolytic agents may be achieved via genetic modifications of this domain. The strategies to modify or broaden tropism and increase transduction efficiency of Ad5-based vectors include: 1) an incorporation of a targeting peptide into the fiber knob domain (the HI loop and/or C-terminus); 2) fiber knob serotype switching, or pseudotyping, by constructing chimeric fibers consisting of the knob domain derived from an alternate serotype (e.g., Ad5/3 or Ad5/35 chimeras), which binds to receptor(s) other than CAR (e.g., desmoglein 2/DSG2 and/or CD46); 3) "fiber complex mosaicism", an approach of combining serotype chimerism with peptide ligand(s) incorporation (e.g., Ad5/3-RGD); 4) "dual fiber mosaicism" by expressing two separate fibers with distinct receptor-binding capabilities on the same viral particle (e.g., Ad5-5/3 or Ad5-5/σ1); 5) fiber xenotyping by replacing the knob and shaft domains of wild-type Ad5 fiber protein with fibritin trimerization domain of T4 bacteriophage or σ1 attachment protein of reovirus. Other genetic approaches to increase the CAR-independent transduction efficiency include insertion of a targeting peptide into the hypervariable region of the capsid protein hexon or fusion to the C-terminus of pIX. Finally, we consider a yet unsolved molecular mechanism of liver targeting by Ad5-based vectors (CAR-, integrin-, fiber shaft KKTK motif-, and hepatic heparan sulfate glycosaminoglycans-independent, but fiber-, hexon- and blood factor X-dependent).
Collapse
Affiliation(s)
- Aleksei A Stepanenko
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky lane 23, 119034 Moscow, Russia.
| | - Vladimir P Chekhonin
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky lane 23, 119034 Moscow, Russia; Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov str. 1, 117997 Moscow, Russia.
| |
Collapse
|
15
|
Abstract
Cross-species transmission of viral pathogens is becoming an increasing problem for captive-animal facilities. This study highlights how animals in captivity are vulnerable to novel opportunistic pathogens, many of which do not result in straightforward diagnosis from symptoms and histopathology. In this study, a novel pathogen was suspected to have contributed to the death of a juvenile polar bear. HTS techniques were employed, and a novel Mastadenovirus was isolated. The virus was present in both the tissue and blood samples. Phylogenetic analysis of the virus at both the gene and genome levels revealed that it is highly divergent to other known mastadenoviruses. Overall, this study shows that animals in isolated conditions still come into contact with novel pathogens, and for many of these pathogens, the host reservoir and mode of transmission are yet to be determined. Polar bears in captivity can be exposed to opportunistic pathogens not present in their natural environments. A 4-month-old polar bear (Ursus maritimus) living in an isolated enclosure with his mother in the Tierpark Berlin, Berlin, Germany, was suffering from severe abdominal pain, mild diarrhea, and loss of appetite and died in early 2017. Histopathology revealed severe hepatic degeneration and necrosis without evidence of inflammation or inclusion bodies, although a viral infection had been suspected on the basis of the clinical signs. We searched for nucleic acids of pathogens by shotgun high-throughput sequencing (HTS) from genomic DNA and cDNA extracted from tissue and blood. We identified a novel Mastadenovirus and assembled a nearly complete genome from the shotgun sequences. Quantitative PCR (qPCR) revealed that viral DNA was present in various concentrations in all tissues examined and that the highest concentrations were found in blood. Viral culture did not yield cytopathic effects, but qPCR suggested that virus replication was sustained for up to three passages. Positive immunofluorescence staining confirmed that the virus was able to replicate in the cells during early passage. Phylogenetic analysis demonstrated that the virus is highly divergent compared to other previously identified Mastadenovirus members and basal to most known viral clades. The virus was found only in the 4-month-old bear and not in other captive polar bears tested. We surmised, therefore, that the polar bear was infected from an unknown reservoir, illustrating that adenoviral diversity remains underestimated and that cross-species transmission of viruses can occur even under conditions of relative isolation. IMPORTANCE Cross-species transmission of viral pathogens is becoming an increasing problem for captive-animal facilities. This study highlights how animals in captivity are vulnerable to novel opportunistic pathogens, many of which do not result in straightforward diagnosis from symptoms and histopathology. In this study, a novel pathogen was suspected to have contributed to the death of a juvenile polar bear. HTS techniques were employed, and a novel Mastadenovirus was isolated. The virus was present in both the tissue and blood samples. Phylogenetic analysis of the virus at both the gene and genome levels revealed that it is highly divergent to other known mastadenoviruses. Overall, this study shows that animals in isolated conditions still come into contact with novel pathogens, and for many of these pathogens, the host reservoir and mode of transmission are yet to be determined.
Collapse
|
16
|
Baker AT, Aguirre-Hernández C, Halldén G, Parker AL. Designer Oncolytic Adenovirus: Coming of Age. Cancers (Basel) 2018; 10:E201. [PMID: 29904022 PMCID: PMC6025169 DOI: 10.3390/cancers10060201] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
The licensing of talimogene laherparepvec (T-Vec) represented a landmark moment for oncolytic virotherapy, since it provided unequivocal evidence for the long-touted potential of genetically modified replicating viruses as anti-cancer agents. Whilst T-Vec is promising as a locally delivered virotherapy, especially in combination with immune-checkpoint inhibitors, the quest continues for a virus capable of specific tumour cell killing via systemic administration. One candidate is oncolytic adenovirus (Ad); it’s double stranded DNA genome is easily manipulated and a wide range of strategies and technologies have been employed to empower the vector with improved pharmacokinetics and tumour targeting ability. As well characterised clinical and experimental agents, we have detailed knowledge of adenoviruses’ mechanisms of pathogenicity, supported by detailed virological studies and in vivo interactions. In this review we highlight the strides made in the engineering of bespoke adenoviral vectors to specifically infect, replicate within, and destroy tumour cells. We discuss how mutations in genes regulating adenoviral replication after cell entry can be used to restrict replication to the tumour, and summarise how detailed knowledge of viral capsid interactions enable rational modification to eliminate native tropisms, and simultaneously promote active uptake by cancerous tissues. We argue that these designer-viruses, exploiting the viruses natural mechanisms and regulated at every level of replication, represent the ideal platforms for local overexpression of therapeutic transgenes such as immunomodulatory agents. Where T-Vec has paved the way, Ad-based vectors now follow. The era of designer oncolytic virotherapies looks decidedly as though it will soon become a reality.
Collapse
Affiliation(s)
- Alexander T Baker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Carmen Aguirre-Hernández
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| |
Collapse
|
17
|
Native and engineered tropism of vectors derived from a rare species D adenovirus serotype 43. Oncotarget 2018; 7:53414-53429. [PMID: 27462785 PMCID: PMC5288196 DOI: 10.18632/oncotarget.10800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/13/2016] [Indexed: 02/03/2023] Open
Abstract
Unique molecular properties of species D adenoviruses (Ads)—the most diverse yet underexplored group of Ads—have been used to develop improved gene vectors. The low seroprevalence in humans of adenovirus serotype 43 (Ad43), an otherwise unstudied species D Ad, identified this rare serotype as an attractive new human gene therapy vector platform. Thus, in this study we wished to assess biological properties of Ad43 essential to its vectorization. We found that (1) Ad43 virions do not bind blood coagulation factor X and cause low random transduction upon vascular delivery; (2) they clear host tissues more quickly than do traditionally used Ad5 vectors; (3) Ad43 uses CD46 as primary receptor; (4) Ad43 can use integrins as alternative primary receptors. As the first step toward vectorization of Ad43, we demonstrated that the primary receptor specificity of the Ad43 fiber can be altered to achieve infection via Her2, an established oncotarget. Whereas this modification required use of the Ad5 fiber shaft, the presence of this domain in chimeric virions did not make them susceptible for neutralization by anti-Ad5 antibodies.
Collapse
|
18
|
Coxsackievirus Adenovirus Receptor Loss Impairs Adult Neurogenesis, Synapse Content, and Hippocampus Plasticity. J Neurosci 2017; 36:9558-71. [PMID: 27629708 DOI: 10.1523/jneurosci.0132-16.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/26/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Although we are beginning to understand the late stage of neurodegenerative diseases, the molecular defects associated with the initiation of impaired cognition are poorly characterized. Here, we demonstrate that in the adult brain, the coxsackievirus and adenovirus receptor (CAR) is located on neuron projections, at the presynapse in mature neurons, and on the soma of immature neurons in the hippocampus. In a proinflammatory or diseased environment, CAR is lost from immature neurons in the hippocampus. Strikingly, in hippocampi of patients at early stages of late-onset Alzheimer's disease (AD), CAR levels are significantly reduced. Similarly, in triple-transgenic AD mice, CAR levels in hippocampi are low and further reduced after systemic inflammation. Genetic deletion of CAR from the mouse brain triggers deficits in adult neurogenesis and synapse homeostasis that lead to impaired hippocampal plasticity and cognitive deficits. We propose that post-translational CAR loss of function contributes to cognitive defects in healthy and diseased-primed brains. SIGNIFICANCE STATEMENT This study addressed the role of the coxsackievirus and adenovirus receptor (CAR), a single-pass cell adhesion molecule, in the adult brain. Our results demonstrate that CAR is expressed by mature neurons throughout the brain. In addition, we propose divergent roles for CAR in immature neurons, during neurogenesis, and at the mature synapse. Notably, CAR loss of function also affects hippocampal plasticity.
Collapse
|
19
|
Defining a Novel Role for the Coxsackievirus and Adenovirus Receptor in Human Adenovirus Serotype 5 Transduction In Vitro in the Presence of Mouse Serum. J Virol 2017; 91:JVI.02487-16. [PMID: 28381574 PMCID: PMC5446653 DOI: 10.1128/jvi.02487-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/24/2017] [Indexed: 01/02/2023] Open
Abstract
Human adenoviral serotype 5 (HAdV-5) vectors have predominantly hepatic tropism when delivered intravascularly, resulting in immune activation and toxicity. Coagulation factor X (FX) binding to HAdV-5 mediates liver transduction and provides protection from virion neutralization in mice. FX is dispensable for liver transduction in mice lacking IgM antibodies or complement, suggesting that alternative transduction pathways exist. To identify novel factor(s) mediating HAdV-5 FX-independent entry, we investigated HAdV-5 transduction in vitro in the presence of serum from immunocompetent C57BL/6 or immunocompromised mice lacking IgM antibodies (Rag 2-/- and NOD-scid-gamma [NSG]). Sera from all three mouse strains enhanced HAdV-5 transduction of A549 cells. While inhibition of HAdV-5-FX interaction with FX-binding protein (X-bp) inhibited transduction in the presence of C57BL/6 serum, it had negligible effect on the enhanced transduction observed in the presence of Rag 2-/- or NSG serum. Rag 2-/- serum also enhanced transduction of the FX binding-deficient HAdV-5HVR5*HVR7*E451Q (AdT*). Interestingly, Rag 2-/- serum enhanced HAdV-5 transduction in a FX-independent manner in CHO-CAR and SKOV3-CAR cells (CHO or SKOV3 cells transfected to stably express human coxsackievirus and adenovirus receptor [CAR]). Additionally, blockade of CAR with soluble HAdV-5 fiber knob inhibited mouse serum-enhanced transduction in A549 cells, suggesting a potential role for CAR. Transduction of HAdV-5 KO1 and HAdV-5/F35 (CAR binding deficient) in the presence of Rag 2-/- serum was equivalent to that of HAdV-5, indicating that direct interaction between HAdV-5 and CAR is not required. These data suggest that FX may protect HAdV-5 from neutralization but has minimal contribution to HAdV-5 transduction in the presence of immunocompromised mouse serum. Alternatively, transduction occurs via an unidentified mouse serum protein capable of bridging HAdV-5 to CAR.IMPORTANCE The intravascular administration of HAdV-5 vectors can result in acute liver toxicity, transaminitis, thrombocytopenia, and injury to the vascular endothelium, illustrating challenges yet to overcome for HAdV-5-mediated systemic gene therapy. The finding that CAR and potentially an unidentified factor present in mouse serum might be important mediators of HAdV-5 transduction highlights that a better understanding of the complex biology defining the interplay between adenovirus immune recognition and cellular uptake mechanisms is still required. These findings are important to inform future optimization and development of HAdV-5-based adenoviral vectors for gene therapy.
Collapse
|
20
|
Matthäus C, Langhorst H, Schütz L, Jüttner R, Rathjen FG. Cell-cell communication mediated by the CAR subgroup of immunoglobulin cell adhesion molecules in health and disease. Mol Cell Neurosci 2016; 81:32-40. [PMID: 27871939 DOI: 10.1016/j.mcn.2016.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022] Open
Abstract
The immunoglobulin superfamily represents a diverse set of cell-cell contact proteins and includes well-studied members such as NCAM1, DSCAM, L1 or the contactins which are strongly expressed in the nervous system. In this review we put our focus on the biological function of a less understood subgroup of Ig-like proteins composed of CAR (coxsackievirus and adenovirus receptor), CLMP (CAR-like membrane protein) and BT-IgSF (brain and testis specific immunoglobulin superfamily). The CAR-related proteins are type I transmembrane proteins containing an N-terminal variable (V-type) and a membrane proximal constant (C2-type) Ig domain in their extracellular region which are implicated in homotypic adhesion. They are highly expressed during embryonic development in a variety of tissues including the nervous system whereby in adult stages the protein level of CAR and CLMP decreases, only BT-IgSF expression increases within age. CAR-related proteins are concentrated at specialized cell-cell communication sites such as gap or tight junctions and are present at the plasma membrane in larger protein complexes. Considerable progress has been made on the molecular structure and interactions of CAR while research on CLMP and BT-IgSF is at an early stage. Studies on mouse mutants revealed biological functions of CAR in the heart and for CLMP in the gastrointestinal and urogenital systems. Furthermore, CAR and BT-IgSF appear to regulate synaptic function in the hippocampus.
Collapse
Affiliation(s)
- Claudia Matthäus
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany.
| | - Hanna Langhorst
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany
| | - Laura Schütz
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany
| | - René Jüttner
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany
| | - Fritz G Rathjen
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany.
| |
Collapse
|
21
|
Bhatia S, O'Bryan SM, Rivera AA, Curiel DT, Mathis JM. CXCL12 retargeting of an adenovirus vector to cancer cells using a bispecific adapter. Oncolytic Virother 2016; 5:99-113. [PMID: 27957479 PMCID: PMC5113939 DOI: 10.2147/ov.s112107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ad vectors are promising delivery vehicles for cancer therapeutic interventions. However, their application is limited by promiscuous tissue tropism and hepatotoxicity. This limitation can be avoided by altering the native tropism of Ads so that they can be redirected to the target cells through alternate cellular receptors. The CXCR4 chemokine receptor belongs to a large superfamily of G-protein-coupled receptors and is known to be upregulated in a wide variety of cancers, including breast cancer and melanoma. These receptors have been associated with cancer cell survival, progression, and metastasis. In the current study, an Ad to cancer cells overexpressing CXCR4 by using a bispecific adapter, sCAR-CXCL12, was retargeted. The sCAR-CXCL12 adapter contained the soluble ectodomain form of the native Ad5 receptor (sCAR), which was fused to a mature human chemokine ligand, CXCL12, through a short peptide linker. A dramatic increase in the infectivity of cancer cells using a targeted Ad vector compared with an untargeted vector was observed. Furthermore, sCAR-CXCL12 attenuated Ad infection of liver ex vivo and in vivo and enhanced Ad vector infection of xenograft tumors implanted in immunodeficient SCID-bg mice. Thus, the sCAR-CXCL12 adapter could be used to retarget Ad vectors to chemokine receptor-positive tumors.
Collapse
Affiliation(s)
- Shilpa Bhatia
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Samia M O'Bryan
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Angel A Rivera
- Departments of Pathology and Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - David T Curiel
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - J Michael Mathis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| |
Collapse
|
22
|
Abstract
For decades, effective cancer gene therapy has been a tantalising prospect; for a therapeutic modality potentially able to elicit highly effective and selective responses, definitive efficacy outcomes have often seemed out of reach. However, steady progress in vector development and accumulated experience from previous clinical studies has finally led the field to its first licensed therapy. Following a pivotal phase III trial, Imlygic (talimogene laherparepvec/T-Vec) received US approval as a treatment for cutaneous and subcutaneous melanoma in October 2015, followed several weeks later by its European authorisation. These represent the first approvals for an oncolytic virotherapy. Imlygic is an advanced-generation herpesvirus-based vector optimised for oncolytic and immunomodulatory activities. Many other oncolytic agents currently remain in development, providing hope that current success will be followed by other diverse vectors that may ultimately come to constitute a new class of clinical anti-cancer agents. In this review, we discuss some of the key oncolytic viral agents developed in the adenovirus and herpesvirus classes, and the prospects for further enhancing their efficacy by combining them with novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alan E. Bilsland
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, G61 1QH, UK
| | | | - T. R. Jeffry Evans
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, G61 1QH, UK
| |
Collapse
|
23
|
Lang S, Wang L, Wang Z, Zhu R, Yan J, Wang B, Wu J, Zhang H, Wu H, Zhou Y, Kong W, Yu B, Yu X. Localization of neutralization epitopes on adenovirus fiber knob from species C. J Gen Virol 2016; 97:955-962. [PMID: 26801881 DOI: 10.1099/jgv.0.000410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Although potential neutralization epitopes on the fiber knob of adenovirus (AdV) serotype 2 (Ad2) and Ad5 have been revealed, few studies have been carried out to identify neutralization epitopes on the knob from a broader panel of AdV serotypes. In this study, based on sequence and structural analysis of knobs from Ad1, Ad2, Ad5 and Ad6 (all from species C), several trimeric chimeric knob proteins were expressed in Escherichia coli to identify the locations of neutralization epitopes on the knobs by analysing their reactivity with mouse and rabbit polyclonal sera raised against AdVs and human sera with natural AdV infection. The dominant neutralization epitopes were located mainly in the N-terminal part of knobs from Ad1, Ad2 and Ad5, but they seemed to be located in the C-terminal part of the Ad6 knob, with some individual differences in rabbit and human populations. Our study adds to our understanding of humoral immune responses to AdVs and will facilitate the construction of more desirable capsid-modified recombinant Ad5 vectors.
Collapse
Affiliation(s)
- Shuai Lang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Lizheng Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Zixuan Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Rui Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Jingyi Yan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Baoming Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Yan Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina.,Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, PRChina.,Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PRChina
| |
Collapse
|
24
|
Abstract
Human adenovirus (Ad) has been used extensively to develop gene transfer vectors for vaccine and gene therapy applications. A major factor limiting the efficacy of the current generation of Ad vectors is their inability to accomplish specific gene delivery to the cells of interest. Transductional targeting strategies seek to redirect virus binding to the appropriate cellular receptor to increase infection efficiency in selected cell types to achieve therapeutic intervention. These efforts mainly focused on incorporating targeting ligands by means of chemical conjugation or genetic modification of Ad capsid proteins and using bispecific adapter molecules to mediate virus recognition of target cells. This review summarizes current progress in Ad tropism modification maneuvers that embody genetic capsid modification and adapter-based approaches that have encouraging implications for further development of advanced vectors suitable for clinical translation.
Collapse
|
25
|
|
26
|
Dakin RS, Parker AL, Delles C, Nicklin SA, Baker AH. Efficient transduction of primary vascular cells by the rare adenovirus serotype 49 vector. Hum Gene Ther 2015; 26:312-9. [PMID: 25760682 PMCID: PMC4442572 DOI: 10.1089/hum.2015.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/06/2015] [Indexed: 01/16/2023] Open
Abstract
Neointima formation and vascular remodeling through vascular smooth muscle cell migration and proliferation can limit the long-term success of coronary interventions, for example, in coronary artery bypass grafting (CABG). Ex vivo gene therapy has the potential to reduce unnecessary cell proliferation and limit neointima formation in vascular pathologies. To date, the species C adenovirus serotype 5 has been commonly used for preclinical gene therapy; however, its suitability is potentially limited by relatively poor tropism for vascular cells and high levels of preexisting immunity in the population. To avoid these limitations, novel species of adenovirus are being tested; here we investigate the potential of adenovirus 49 (Ad49) for use in gene therapy. Transduction of primary human vascular cells by a range of adenovirus serotypes was assessed; Ad49 demonstrated highest transduction of both vascular smooth muscle and endothelial cells. Gene transfer with Ad49 in vascular smooth muscle and endothelial cells was possible following short exposure times (<1 hr) and with low MOI, which is clinically relevant. Ex vivo delivery to surplus CABG tissue showed efficient gene transfer with Ad49, consistent with the in vitro findings. Luminal infusion of Ad49GFP into intact CABG samples ex vivo resulted in efficient vessel transduction. In addition, no seroprevalence rates to Ad49 were observed in a Scottish cohort of patients from cardiovascular clinics, thus circumventing issues with preexisting immunity. Our results show that Ad49 has tropism for vascular cells in vitro and ex vivo and demonstrate that Ad49 may be an improved vector for local vascular gene therapy compared with current alternatives.
Collapse
Affiliation(s)
- Rachel S. Dakin
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Alan L. Parker
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Stuart A. Nicklin
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Andrew H. Baker
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| |
Collapse
|
27
|
Lenman A, Liaci AM, Liu Y, Årdahl C, Rajan A, Nilsson E, Bradford W, Kaeshammer L, Jones MS, Frängsmyr L, Feizi T, Stehle T, Arnberg N. Human adenovirus 52 uses sialic acid-containing glycoproteins and the coxsackie and adenovirus receptor for binding to target cells. PLoS Pathog 2015; 11:e1004657. [PMID: 25674795 PMCID: PMC4335501 DOI: 10.1371/journal.ppat.1004657] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/05/2015] [Indexed: 11/25/2022] Open
Abstract
Most adenoviruses attach to host cells by means of the protruding fiber protein that binds to host cells via the coxsackievirus and adenovirus receptor (CAR) protein. Human adenovirus type 52 (HAdV-52) is one of only three gastroenteritis-causing HAdVs that are equipped with two different fiber proteins, one long and one short. Here we show, by means of virion-cell binding and infection experiments, that HAdV-52 can also attach to host cells via CAR, but most of the binding depends on sialylated glycoproteins. Glycan microarray, flow cytometry, surface plasmon resonance and ELISA analyses reveal that the terminal knob domain of the long fiber (52LFK) binds to CAR, and the knob domain of the short fiber (52SFK) binds to sialylated glycoproteins. X-ray crystallographic analysis of 52SFK in complex with 2-O-methylated sialic acid combined with functional studies of knob mutants revealed a new sialic acid binding site compared to other, known adenovirus:glycan interactions. Our findings shed light on adenovirus biology and may help to improve targeting of adenovirus-based vectors for gene therapy.
Collapse
Affiliation(s)
- Annasara Lenman
- Division of Virology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - A. Manuel Liaci
- University of Tübingen, Interfaculty Institute of Biochemistry, Tübingen, Germany
| | - Yan Liu
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Carin Årdahl
- Division of Virology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Anandi Rajan
- Division of Virology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Emma Nilsson
- Division of Virology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Will Bradford
- University of Tübingen, Interfaculty Institute of Biochemistry, Tübingen, Germany
| | - Lisa Kaeshammer
- University of Tübingen, Interfaculty Institute of Biochemistry, Tübingen, Germany
| | - Morris S. Jones
- Division of Infectious Diseases, Naval Medical Center, San Diego, California, United States of America
| | - Lars Frängsmyr
- Division of Virology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Ten Feizi
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Thilo Stehle
- University of Tübingen, Interfaculty Institute of Biochemistry, Tübingen, Germany
- Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Niklas Arnberg
- Division of Virology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| |
Collapse
|
28
|
miR-466 is putative negative regulator of Coxsackie virus and Adenovirus Receptor. FEBS Lett 2014; 589:246-54. [PMID: 25497012 DOI: 10.1016/j.febslet.2014.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 11/18/2014] [Accepted: 12/02/2014] [Indexed: 01/01/2023]
Abstract
This study aimed at elucidating how Coxsackie B virus (CVB) perturbs the host's microRNA (miRNA) regulatory pathways that lead to antiviral events. The results of miRNA profiling in rat pancreatic cells infection models revealed that rat rno-miR-466d was up-regulated in CVB infection. Furthermore, in silico studies showed that Coxsackie virus and Adenovirus Receptor (CAR), a cellular receptor, was one of the rno-miR-466d targets involved in viral entry. Subsequent experiments also proved that both the rno-miR-466d and the human hsa-miR-466, which are orthologs of the miR-467 gene family, could effectively down-regulate the levels of rat and human CAR protein expression, respectively.
Collapse
|
29
|
Abstract
ABSTRACT Viruses are a diverse class of nanoparticles. However, they have evolved a few common mechanisms that enable successful infection of their host cells. The first stage of this process involves entry into the cell. For enveloped viruses this process has been well characterized. For nonenveloped viruses, the focus of this review, the entry mechanisms are less well understood. For these viruses, a typical pathway involves receptor attachment followed by internalization into cellular vesicles and subsequent viral escape to the cytosol and transport to the site of genome replication. Significantly, these viruses have evolved numerous mechanisms to fulfill this seemingly simple infection scheme. We focus on the latest observations for several families of nonenveloped viruses and highlight specific members for eukaryotic families: Adenoviridae, Papillomaviridae, Parvoviridae, Picornaviridae, Polyomaviridae and Reoviridae; and prokaryotic families: Microviridae, Myoviridae, Podoviridae and Siphoviridae.
Collapse
Affiliation(s)
- Bridget Lins
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
30
|
Lopez-Gordo E, Denby L, Nicklin SA, Baker AH. The importance of coagulation factors binding to adenovirus: historical perspectives and implications for gene delivery. Expert Opin Drug Deliv 2014; 11:1795-813. [DOI: 10.1517/17425247.2014.938637] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Estrella Lopez-Gordo
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Laura Denby
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Stuart A Nicklin
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Andrew H Baker
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK ;
| |
Collapse
|
31
|
Peptide-based technologies to alter adenoviral vector tropism: ways and means for systemic treatment of cancer. Viruses 2014; 6:1540-63. [PMID: 24699364 PMCID: PMC4014709 DOI: 10.3390/v6041540] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/15/2014] [Accepted: 03/20/2014] [Indexed: 12/11/2022] Open
Abstract
Due to the fundamental progress in elucidating the molecular mechanisms of human diseases and the arrival of the post-genomic era, increasing numbers of therapeutic genes and cellular targets are available for gene therapy. Meanwhile, the most important challenge is to develop gene delivery vectors with high efficiency through target cell selectivity, in particular under in situ conditions. The most widely used vector system to transduce cells is based on adenovirus (Ad). Recent endeavors in the development of selective Ad vectors that target cells or tissues of interest and spare the alteration of all others have focused on the modification of the virus broad natural tropism. A popular way of Ad targeting is achieved by directing the vector towards distinct cellular receptors. Redirecting can be accomplished by linking custom-made peptides with specific affinity to cellular surface proteins via genetic integration, chemical coupling or bridging with dual-specific adapter molecules. Ideally, targeted vectors are incapable of entering cells via their native receptors. Such altered vectors offer new opportunities to delineate functional genomics in a natural environment and may enable efficient systemic therapeutic approaches. This review provides a summary of current state-of-the-art techniques to specifically target adenovirus-based gene delivery vectors.
Collapse
|
32
|
Schreiber J, Langhorst H, Jüttner R, Rathjen FG. The IgCAMs CAR, BT-IgSF, and CLMP: Structure, Function, and Diseases. ADVANCES IN NEUROBIOLOGY 2014; 8:21-45. [DOI: 10.1007/978-1-4614-8090-7_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Development of a generic adenovirus delivery system based on structure-guided design of bispecific trimeric DARPin adapters. Proc Natl Acad Sci U S A 2013; 110:E869-77. [PMID: 23431166 DOI: 10.1073/pnas.1213653110] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adenoviruses (Ads) have shown promise as vectors for gene delivery in clinical trials. Efficient viral targeting to a tissue of choice requires both ablation of the virus' original tropism and engineering of an efficient receptor-mediated uptake by a specific cell population. We have developed a series of adapters binding to the virus with such high affinity that they remain fully bound for >10 d, block its natural receptor binding site and mediate interaction with a surface receptor of choice. The adapter contains two fused modules, both consisting of designed ankyrin repeat proteins (DARPins), one binding to the fiber knob of adenovirus serotype 5 and the other binding to various tumor markers. By solving the crystal structure of the complex of the trimeric knob with three bound DARPins at 1.95-Å resolution, we could use computer modeling to design a link to a trimeric protein of extraordinary kinetic stability, the capsid protein SHP from the lambdoid phage 21. We arrived at a module which binds the knob like a trimeric clamp. When this clamp was fused with DARPins of varying specificities, it enabled adenovirus serotype 5-mediated delivery of a transgene in a human epidermal growth factor receptor 2-, epidermal growth factor receptor-, or epithelial cell adhesion molecule-dependent manner with transduction efficiencies comparable to or even exceeding those of Ad itself. With these adapters, efficiently produced in Escherichia coli, Ad can be converted rapidly to new receptor specificities using any ligand as the receptor-binding moiety. Prefabricated Ads with different payloads thus can be retargeted readily to many cell types of choice.
Collapse
|
34
|
Reetz J, Herchenröder O, Schmidt A, Pützer BM. Vector Technology and Cell Targeting: Peptide-Tagged Adenoviral Vectors as a Powerful Tool for Cell Specific Targeting. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
35
|
Mechanisms of human adenovirus inactivation by sunlight and UVC light as examined by quantitative PCR and quantitative proteomics. Appl Environ Microbiol 2012; 79:1325-32. [PMID: 23241978 DOI: 10.1128/aem.03457-12] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Human adenoviruses (HAdV) are important pathogens in both industrialized and developing nations. HAdV has been shown to be relatively resistant to monochromatic UVC light. Polychromatic UVC light, in contrast, is a more effective means of disinfection, presumably due to the involvement of viral proteins in the inactivation mechanism. Solar disinfection of HAdV, finally, is only poorly understood. In this paper, the kinetics and mechanism of HAdV inactivation by UVC light and direct and indirect solar disinfection are elucidated. PCR and mass spectrometry were employed to quantify the extent of genome and protein degradation and to localize the affected regions in the HAdV proteins. For this purpose, we used for the first time an approach involving stable isotope labeling by amino acids in cell culture (SILAC) of a human virus. Inactivation by UVC light and the full sunlight spectrum were found to efficiently inactivate HAdV, whereas UVA-visible light only caused inactivation in the presence of external sensitizers (indirect solar disinfection). Genome damage was significant for UVC but was less important for solar disinfection. In contrast, indirect solar disinfection exhibited extensive protein degradation. In particular, the fiber protein and the amino acids responsible for host binding within the fiber protein were shown to degrade. In addition, the central domain of the penton protein was damaged, which may inhibit interactions with the fiber protein and lead to a disruption of the initial stages of infection. Damage to the hexon protein, however, appeared to affect only regions not directly involved in the infectious cycle.
Collapse
|
36
|
A new finding concerning adenoviral-mediated gene transfer: A high-level, cell-specific transgene expression in the neural stem cells of adult mice. J Virol Methods 2012; 186:1-6. [DOI: 10.1016/j.jviromet.2012.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/05/2012] [Accepted: 07/09/2012] [Indexed: 01/28/2023]
|
37
|
HER3 targeting of adenovirus by fiber modification increases infection of breast cancer cells in vitro, but not following intratumoral injection in mice. Cancer Gene Ther 2012; 19:888-98. [PMID: 23099884 DOI: 10.1038/cgt.2012.79] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Despite the tremendous potential of adenovirus (Ad) as a delivery vector for cancer gene therapy, its use in clinical settings has been limited, mainly as a result of the limited infectivity in many tumors and the wide tissue tropism associated with Ad. To modify the tropism of the virus, we have inserted the epidermal growth factor-like domain of the human heregulin-α (HRG) into the HI loop of Ad5 fiber. This insertion had no adverse effect on fiber trimerization nor did it affect incorporation of the modified fiber into infectious viral particles. Virions bearing modified fiber displayed growth characteristics and viral yields indistinguishable from those of wild-type (wt) virus. Most importantly, HRG-tagged virions showed enhanced infection of cells expressing the cognate receptors HER3/ErbB3 and HER4/ErbB4. This was significantly reduced in the presence of soluble HRG. Furthermore, HER3-expressing Chinese hamster ovary (CHO) cells were transduced by the HRG-modified virus, but not by wt virus. In contrast, CHO cells expressing the coxsackie-Ad receptor were transduced with both viruses. However, infection of an in vivo breast cancer xenograft model after intratumoral injection was similar with both viruses, suggesting that the tumor microenvironment and/or the route of delivery have important roles in infection of target cells with fiber-modified Ads.
Collapse
|
38
|
Reduced infectivity of adenovirus type 5 particles and degradation of entering viral genomes associated with incomplete processing of the preterminal protein. J Virol 2012; 86:13554-65. [PMID: 23035217 DOI: 10.1128/jvi.02337-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
To investigate further the contribution of the adenovirus type 5 (Ad5) E1B 55-kDa protein to genome replication, viral DNA accumulation was examined in primary human fibroblasts and epithelial cells infected with Ad5 or the E1B 55-kDa-null mutant Hr6. Unexpectedly, all cell types were observed to contain a significantly higher concentration of entering Hr6 than of Ad5 DNA, as did an infectious unit of Hr6. However, the great majority of the Hr6 genomes were degraded soon after entry. As this unusual phenotype cannot be ascribed to the Hr6 E1B frameshift mutation (J. S. Chahal and S. J. Flint, J. Virol. 86:3064-3072, 2012), the sequences of the Ad5 and Hr6 genomes were compared by using high-throughput sequencing. Seven previously unrecognized mutations were identified in the Hr6 genome, two of which result in substitutions in virion proteins, G315V in the preterminal protein (preTP) and A406V in fiber protein IV. Previous observations and the visualization by immunofluorescence of greater numbers of viral genomes entering the cytosol of Hr6-infected cells than of Ad5-infected cells indicated that the fiber mutation could not be responsible for the low-infectivity phenotype of Hr6. However, comparison of the forms of terminal protein present in purified virus particles indicated that the production of mature terminal protein from a processing intermediate is impaired in Hr6 particles. We therefore propose that complete processing of preTP within virus particles is necessary for the ability of viral genomes to become localized at appropriate sites and persist in infected cells.
Collapse
|
39
|
Infectivity-selective oncolytic adenovirus developed by high-throughput screening of adenovirus-formatted library. Mol Ther 2012; 21:139-48. [PMID: 23032977 DOI: 10.1038/mt.2012.205] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Adenovirus (Ad) is a potent gene-delivery vehicle and has frequently been used for designing oncolytic viruses. However, lack of selectivity on infection has hampered the achievement of sufficient in vivo efficiency. Here, we developed a novel oncolytic virus system, infectivity-selective oncolytic adenovirus (ISOAd), via direct high-throughput screening of a high-diversity targeting-ligand library in adenoviral format. Through our newly designed rescue virus system, the high-diversity Ad library carrying the random seven amino acid sequences ligand-library in the AB-loop of its fiber-knob region (5 × 10(9) diversity) was successfully generated. During the screening of this library with the cells expressing the target molecule (mesothelin, MSLN), the AB-loop sequence of the virus clones converged to one dominant sequence and a novel MSLN-targeting sequence was isolated. The virus with the isolated motif showed selective infectivity to MSLN-positive cells in vitro. In vivo, it exhibited a selective and potent antitumor effect resulted from the viral replication in MSLN-positive xenografts. The ISOAd is a novel class of oncolytic Ad, which has selectivity at the step of transduction. The selectivity at the stage of infection can open new perspectives in oncolytic Ad therapy for various diseases.
Collapse
|
40
|
Vaccination with adenovirus serotypes 35, 26, and 48 elicits higher levels of innate cytokine responses than adenovirus serotype 5 in rhesus monkeys. J Virol 2012; 86:9590-8. [PMID: 22787208 DOI: 10.1128/jvi.00740-12] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adenovirus (Ad) vaccine vectors have proven highly immunogenic in multiple experimental models, but the innate immune responses induced by these vectors remain poorly characterized. Here we report innate cytokine responses to 5 different Ad vectors in 26 rhesus monkeys. Vaccination with adenovirus serotype 35 (Ad35), Ad26, and Ad48 induced substantially higher levels of antiviral (gamma interferon [IFN-γ], 10-kDa gamma interferon-induced protein [IP-10]) and proinflammatory (interleukin 1 receptor antagonist [IL-1RA], IL-6) cytokines than vaccination with Ad5 on day 1 following immunization. In vitro studies with capsid chimeric vectors and receptor-blocking monoclonal antibodies suggested that fiber-receptor interactions, as well as other capsid components, were critical for triggering these innate responses. Moreover, multiple cell populations, including dendritic cells, monocytes/macrophages, and T lymphocytes, contributed to these innate cytokine profiles. These data demonstrate that Ad35, Ad26, and Ad48, which utilize CD46 as their primary cellular receptor, induce significantly greater innate cytokine responses than Ad5, which uses the coxsackievirus and adenovirus receptor (CAR). These differences in innate triggering result in markedly different immunologic milieus for the subsequent generation of adaptive immune responses by these vaccine vectors.
Collapse
|
41
|
Zeyaullah M, Patro M, Ahmad I, Ibraheem K, Sultan P, Nehal M, Ali A. Oncolytic viruses in the treatment of cancer: a review of current strategies. Pathol Oncol Res 2012; 18:771-81. [PMID: 22714538 DOI: 10.1007/s12253-012-9548-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 05/30/2012] [Indexed: 12/18/2022]
Abstract
Oncolytic viruses are live, replication-competent viruses that replicate selectively in tumor cells leading to the destruction of the tumor cells. Tumor-selective replicating viruses offer appealing advantages over conventional cancer therapy and are promising a new approach for the treatment of human cancer. The development of virotherapeutics is based on several strategies. Virotherapy is not a new concept, but recent technical advances in the genetic modification of oncolytic viruses have improved their tumor specificity, leading to the development of new weapons for the war against cancer. Clinical trials with oncolytic viruses demonstrate the safety and feasibility of an effective virotherapeutic approach. Strategies to overcome potential obstacles and challenges to virotherapy are currently being explored. Systemic administrations of oncolytic viruses will successfully extend novel treatment against a range of tumors. Combination therapy has shown some encouraging antitumor responses by eliciting strong immunity against established cancer.
Collapse
Affiliation(s)
- Md Zeyaullah
- Department of Microbiology, Faculty of Medicine, Omar Al-Mukhtar University, Al-Baida, Libya.
| | | | | | | | | | | | | |
Collapse
|
42
|
Ballard EN, Trinh VT, Hogg RT, Gerard RD. Peptide targeting of adenoviral vectors to augment tumor gene transfer. Cancer Gene Ther 2012; 19:476-88. [PMID: 22595794 PMCID: PMC3380173 DOI: 10.1038/cgt.2012.23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adenovirus serotype 5 remains one of the most promising vectors for delivering genetic material to cancer cells for imaging or therapy, but optimization of these agents to selectively promote tumor cell infection is needed to further their clinical development. Peptide sequences that bind to specific cell surface receptors have been inserted into adenoviral capsid proteins to improve tumor targeting, often in the background of mutations designed to ablate normal ligand:receptor interactions and thereby reduce off target effects and toxicities in non-target tissues. Different tumor types also express highly variable complements of cell surface receptors, so a customized targeting strategy using a particular peptide in the context of specific adenoviral mutations may be needed to achieve optimal efficacy. To further investigate peptide targeting strategies in adenoviral vectors, we used a set of peptide motifs originally isolated using phage display technology that evince tumor specificity in vivo. To demonstrate their abilities as targeting motifs, we genetically incorporated these peptides into a surface loop of the fiber capsid protein to construct targeted adenovirus vectors. We then systematically evaluated the ability of these peptide targeted vectors to infect several tumor cell types, both in vitro and in vivo, in a variety of mutational backgrounds designed to reduce CAR and/or HSG-mediated binding. Results from this study support previous observations that peptide insertions in the HI loop of the fiber knob domain are generally ineffective when used in combination with HSG detargeting mutations. The evidence also suggests that this strategy can attenuate other fiber knob interactions, such as CAR-mediated binding, and reduce overall viral infectivity. The insertion of peptides into fiber proved more effective for targeting tumor cell types expressing low levels of CAR receptor, as this strategy can partially compensate for the very low infectivity of wild-type adenovirus in those cells. Nevertheless, the incorporation of relatively low affinity peptide ligands into the fiber knob, while effective in vitro, has only minimal targeting efficacy in vivo and highlights the importance of high affinity ligand:receptor interactions to achieve tumor targeting.
Collapse
Affiliation(s)
- E N Ballard
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | | | | | | |
Collapse
|
43
|
Alberti MO, Roth JC, Ismail M, Tsuruta Y, Abraham E, Pereboeva L, Gerson SL, Curiel DT. Derivation of a myeloid cell-binding adenovirus for gene therapy of inflammation. PLoS One 2012; 7:e37812. [PMID: 22624065 PMCID: PMC3356302 DOI: 10.1371/journal.pone.0037812] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 04/24/2012] [Indexed: 11/21/2022] Open
Abstract
The gene therapy field is currently limited by the lack of vehicles that permit efficient gene delivery to specific cell or tissue subsets. Native viral vector tropisms offer a powerful platform for transgene delivery but remain nonspecific, requiring elevated viral doses to achieve efficacy. In order to improve upon these strategies, our group has focused on genetically engineering targeting domains into viral capsid proteins, particularly those based on adenovirus serotype 5 (Ad5). Our primary strategy is based on deletion of the fiber knob domain, to eliminate broad tissue specificity through the human coxsackie-and-adenovirus receptor (hCAR), with seamless incorporation of ligands to re-direct Ad tropism to cell types that express the cognate receptors. Previously, our group and others have demonstrated successful implementation of this strategy in order to specifically target Ad to a number of surface molecules expressed on immortalized cell lines. Here, we utilized phage biopanning to identify a myeloid cell-binding peptide (MBP), with the sequence WTLDRGY, and demonstrated that MBP can be successfully incorporated into a knob-deleted Ad5. The resulting virus, Ad.MBP, results in specific binding to primary myeloid cell types, as well as significantly higher transduction of these target populations ex vivo, compared to unmodified Ad5. These data are the first step in demonstrating Ad targeting to cell types associated with inflammatory disease.
Collapse
Affiliation(s)
- Michael O. Alberti
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Justin C. Roth
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (JCR); (DTC)
| | - Mourad Ismail
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- National Center for Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yuko Tsuruta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Edward Abraham
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Larisa Pereboeva
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Stanton L. Gerson
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- National Center for Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (JCR); (DTC)
| |
Collapse
|
44
|
Sstr2A: a relevant target for the delivery of genes into human glioblastoma cells using fiber-modified adenoviral vectors. Gene Ther 2012; 20:283-97. [PMID: 22592599 DOI: 10.1038/gt.2012.39] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Glioblastomas are the most aggressive of the brain tumors occurring in adults and children. Currently available chemotherapy prolongs the median survival time of patients by only 4 months. The low efficiency of current treatments is partly owing to the blood-brain barrier, which restricts the penetration of most drugs into the central nervous system. Locoregional treatment strategies thus become mandatory. In this context, viral tools are of great interest for the selective delivery of genes into tumoral cells. Gliomas express high levels of type 2 somatostatin receptors (sstr2A), pinpointing them as suitable targets for the improvement of transduction efficiency in these tumors. We designed a new adenoviral vector based on the introduction of the full-length somatostatin (SRIF (somatotropin release-inhibiting factor)) sequence into the HI loop of the HAdV fiber protein. We demonstrate that (i) HAdV-5-SRIF uptake into cells is mediated by sstr2A, (ii) our vector drives high levels of gene expression in cells expressing endogenous sstr2A, with up to 65-fold enhancement and (iii) low doses of HAdV-5-SRIF are sufficient to infect high-grade human primary glioblastoma cells. Adenoviral vectors targeting SRIF receptors might thus represent a promising therapeutic approach to brain tumors.
Collapse
|
45
|
Rojas JJ, Thorne SH. Theranostic potential of oncolytic vaccinia virus. Theranostics 2012; 2:363-73. [PMID: 22509200 PMCID: PMC3326721 DOI: 10.7150/thno.3724] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/18/2012] [Indexed: 11/17/2022] Open
Abstract
Biological cancer therapies, such as oncolytic, or replication-selective viruses have advantages over traditional therapeutics as they can employ multiple different mechanisms to target and destroy cancers (including direct cell lysis, immune activation and vascular collapse). This has led to their rapid recent clinical development. However this also makes their pre-clinical and clinical study complex, as many parameters may affect their therapeutic potential and so defining reason for treatment failure or approaches that might enhance their therapeutic activity can be complicated. The ability to non-invasively image viral gene expression in vivo both in pre-clinical models and during clinical testing will considerably enhance the speed of oncolytic virus development as well as increasing the level and type of useful data produced from these studies. Further, subsequent to future clinical approval, imaging of reporter gene expression might be used to evaluate the likelihood of response to oncolytic viral therapy prior to changes in tumor burden. Here different reporter genes used in conjunction with oncolytic viral therapy are described, along with the imaging modalities used to measure their expression, while their applications both in pre-clinical and clinical testing are discussed. Possible future applications for reporter gene expression from oncolytic viruses in the phenotyping of tumors and the personalizing of treatment regimens are also discussed.
Collapse
|
46
|
Duffy MR, Parker AL, Bradshaw AC, Baker AH. Manipulation of adenovirus interactions with host factors for gene therapy applications. Nanomedicine (Lond) 2012; 7:271-88. [DOI: 10.2217/nnm.11.186] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanomedicine based on the use of adenovirus vectors for therapeutic gene delivery shows broad potential. Specific targeting for many gene therapy applications, such as metastatic cancers or cardiovascular diseases requires intravascular delivery of the vector. However, a major barrier to successful adenovirus vector targeting follows systemic delivery, as upon contact with the bloodstream the virus interacts with a variety of host proteins, in particular coagulation factor X, which mediates profound liver gene transfer. This inherent hepatic tropism combined with macrophage scavenging minimizes the efficacy of the virus at the desired sites and induces toxic side effects. Understanding the complex, multifaceted interactions of adenovirus with host factors is of vital importance to the design of safer vectors with improved efficacy and pharmacokinetic profiles. Increased knowledge of adenovirus biology provides the opportunity to develop innovative strategies to detarget the virus from the liver following intravascular delivery and redirect the vector to disease areas.
Collapse
Affiliation(s)
- Margaret R Duffy
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Alan L Parker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Angela C Bradshaw
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Andrew H Baker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
47
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
48
|
Burckhardt CJ, Suomalainen M, Schoenenberger P, Boucke K, Hemmi S, Greber UF. Drifting motions of the adenovirus receptor CAR and immobile integrins initiate virus uncoating and membrane lytic protein exposure. Cell Host Microbe 2011; 10:105-17. [PMID: 21843868 DOI: 10.1016/j.chom.2011.07.006] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 04/01/2011] [Accepted: 07/06/2011] [Indexed: 12/19/2022]
Abstract
Viral particle binding to plasma membrane receptors elicits virus motions, recruits signaling proteins, and triggers membrane bending and fission, finally resulting in endocytic virus uptake. Here we analyze how human adenovirus engages its receptor coxsackievirus adenovirus receptor (CAR) and coreceptor αv integrin to move on the plasma membrane. Virus binding to CAR through fiber knobs gave rise to diffusive motions and actomyosin-2-dependent drifts, while integrin-targeted viruses were spatially more confined. Diffusions, drifts, and confined motions were specifically observed with viral particles that were subsequently internalized. CAR-mediated drifts together with integrin binding supported fiber shedding from adenovirus particles, leading to exposure of the membrane-lytic internal virion protein VI and enhanced viral escape from endosomes. Our results show that adenovirus uncoating is initiated at the plasma membrane by CAR drifting motion and binding to immobile integrins.
Collapse
Affiliation(s)
- Christoph J Burckhardt
- Institute of Molecular Life Science, University of Zürich, Winterthurerstrasse, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Vajda A, Marignol L, Foley R, Lynch TH, Lawler M, Hollywood D. Clinical potential of gene-directed enzyme prodrug therapy to improve radiation therapy in prostate cancer patients. Cancer Treat Rev 2011; 37:643-54. [DOI: 10.1016/j.ctrv.2011.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 03/08/2011] [Accepted: 03/16/2011] [Indexed: 11/30/2022]
|
50
|
Poulin KL, Tong G, Vorobyova O, Pool M, Kothary R, Parks RJ. Use of Cre/loxP recombination to swap cell binding motifs on the adenoviral capsid protein IX. Virology 2011; 420:146-55. [DOI: 10.1016/j.virol.2011.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/25/2011] [Accepted: 09/02/2011] [Indexed: 12/01/2022]
|