1
|
Muravyeva A, Smirnikhina S. Strategies for Modifying Adenoviral Vectors for Gene Therapy. Int J Mol Sci 2024; 25:12461. [PMID: 39596526 PMCID: PMC11595218 DOI: 10.3390/ijms252212461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Adenoviral vectors (AdVs) are effective vectors for gene therapy due to their broad tropism, large capacity, and high transduction efficiency, making them widely used as oncolytic vectors and for creating vector-based vaccines. This review also considers the application of adenoviral vectors in oncolytic virotherapy and gene therapy for inherited diseases, analyzing strategies to enhance their efficacy and specificity. However, despite significant progress in this field, the use of adenoviral vectors is limited by their high immunogenicity, low specificity to certain cell types, and limited duration of transgene expression. Various strategies and technologies aimed at improving the characteristics of adenoviral vectors are being developed to overcome these limitations. Significant attention is being paid to the creation of tissue-specific promoters, which allow for the controlled expression of transgenes, as well as capsid modifications that enhance tropism to target cells, which also play a key role in reducing immunogenicity and increasing the efficiency of gene delivery. This review focuses on modern approaches to adenoviral vector modifications made to enhance their effectiveness in gene therapy, analyzing the current achievements, challenges, and prospects for applying these technologies in clinical practice, as well as identifying future research directions necessary for successful clinical implementation.
Collapse
Affiliation(s)
| | - Svetlana Smirnikhina
- Laboratory of Genome Editing, Research Centre for Medical Genetics, Moskvorechye, 1, 115522 Moscow, Russia
| |
Collapse
|
2
|
Choi A, Javius-Jones K, Hong S, Park H. Cell-Based Drug Delivery Systems with Innate Homing Capability as a Novel Nanocarrier Platform. Int J Nanomedicine 2023; 18:509-525. [PMID: 36742991 PMCID: PMC9893846 DOI: 10.2147/ijn.s394389] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023] Open
Abstract
Nanoparticle-based drug delivery systems have been designed to treat various diseases. However, many problems remain, such as inadequate tumor targeting and poor therapeutic outcomes. To overcome these obstacles, cell-based drug delivery systems have been developed. Candidates for cell-mediated drug delivery include blood cells, immune cells, and stem cells with innate tumor tropism and low immunogenicity; they act as a disguise to deliver the therapeutic payload. In drug delivery systems, therapeutic agents are encapsulated intracellularly or attached to the surface of the plasma membrane and transported to the desired site. Here, we review the pros and cons of cell-based therapies and discuss their homing mechanisms in the tumor microenvironment. In addition, different strategies to load therapeutic agents inside or on the surface of circulating cells and the current applications for a wide range of disease treatments are summarized.
Collapse
Affiliation(s)
- Anseo Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea,Correspondence: Hansoo Park; Seungpyo Hong, School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea, Tel +82-2 820 5804, Fax +82-2 813 8159, Email ;
| |
Collapse
|
3
|
Alyami EM, Tarar A, Peng CA. Less phagocytosis of viral vectors by tethering with CD47 ectodomain. J Mater Chem B 2021; 10:64-77. [PMID: 34846059 DOI: 10.1039/d1tb01815a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many viral vectors, which are effective when administrated in situ, lack efficacy when delivered intravenously. The key reason for this is the rapid clearance of the viruses from the blood circulation via the immune system before they reach target sites. Therefore, avoiding their clearance by the immune system is essential. In this study, lentiviral vectors were tethered with the ectodomain of self-marker protein CD47 to suppress phagocytosis via interacting with SIRPα on the outer membrane of macrophage cells. CD47 ectodomain and core-streptavidin fusion gene (CD47ED-coreSA) was constructed into pET-30a(+) plasmid and transformed into Lemo21 (DE3) competent E. coli cells. The expressed CD47ED-coreSA chimeric protein was purified by cobalt-nitrilotriacetate affinity column and characterized by SDS-PAGE and western blot. The purified chimeric protein was anchored on biotinylated lentivirus via biotin-streptavidin binding. The CD47ED-capped lentiviruses encoding GFP were used to infect J774A.1 macrophage cells to assess the impact on phagocytosis. Our results showed that the overexpressed CD47ED-coreSA chimeric protein was purified and bound on the surface of biotinylated lentivirus which was confirmed via immunoblotting assay. The process to produce biotinylated lentivirus did not affect native viral infectivity. It was shown that the level of GFP expression in J774A.1 macrophages transduced with CD47ED-lentiviruses was threefold lower in comparison to control lentiviruses, indicating an antiphagocytic effect triggered by the interaction of CD47ED and SIRPα. Through the test of blocking antibodies against CD47ED and/or SIRPα, it was confirmed that the phagocytosis inhibition was mediated through the CD47ED-SIRPα axis signaling. In conclusion, surface immobilization of CD47ED on lentiviral vectors inhibits their phagocytosis by macrophages. The chimeric protein of CD47 ectodomain and core-streptavidin is effective in mediating the surface binding and endowing the lentiviral nanoparticles with the antiphagocytic property.
Collapse
Affiliation(s)
- Esmael M Alyami
- Department of Chemical and Biological Engineering, University of Idaho, Engineering Physics Building 410, 875 Perimeter Drive, Moscow, ID 83844-0904, USA.
| | - Ammar Tarar
- Department of Chemical and Biological Engineering, University of Idaho, Engineering Physics Building 410, 875 Perimeter Drive, Moscow, ID 83844-0904, USA.
| | - Ching-An Peng
- Department of Chemical and Biological Engineering, University of Idaho, Engineering Physics Building 410, 875 Perimeter Drive, Moscow, ID 83844-0904, USA.
| |
Collapse
|
4
|
Nano-engineered immune cells as "guided missiles" for cancer therapy. J Control Release 2021; 341:60-79. [PMID: 34785315 DOI: 10.1016/j.jconrel.2021.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Immune cells can actively regulate tumors or inflammatory sites and have good biocompatibility and safety. Currently, they are one of the most promising candidates for drug delivery systems. Moreover, immune cells can significantly extend the circulation time of nanoparticles and have broad-spectrum tumor-targeting properties. This article first introduces the immune cell types most commonly used in recent years, analyzes their advantages and disadvantages, and elucidates their application in anti-tumor therapy. Next, the various ways of loading nanoparticles on immune cells that have been used in recent years are summarized and simply divided into two categories: backpacks and Trojan horses. Finally, the two "mountains" that stand in front of us when using immune cells as cell carriers, off-target problems and effective release strategies, are discussed.
Collapse
|
5
|
Weklak D, Pembaur D, Koukou G, Jönsson F, Hagedorn C, Kreppel F. Genetic and Chemical Capsid Modifications of Adenovirus Vectors to Modulate Vector-Host Interactions. Viruses 2021; 13:1300. [PMID: 34372506 PMCID: PMC8310343 DOI: 10.3390/v13071300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
Adenovirus-based vectors are playing an important role as efficacious genetic vaccines to fight the current COVID-19 pandemic. Furthermore, they have an enormous potential as oncolytic vectors for virotherapy and as vectors for classic gene therapy. However, numerous vector-host interactions on a cellular and noncellular level, including specific components of the immune system, must be modulated in order to generate safe and efficacious vectors for virotherapy or classic gene therapy. Importantly, the current widespread use of Ad vectors as vaccines against COVID-19 will induce antivector immunity in many humans. This requires the development of strategies and techniques to enable Ad-based vectors to evade pre-existing immunity. In this review article, we discuss the current status of genetic and chemical capsid modifications as means to modulate the vector-host interactions of Ad-based vectors.
Collapse
Affiliation(s)
| | | | | | | | | | - Florian Kreppel
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Street 10, 58453 Witten, Germany; (D.W.); (D.P.); (G.K.); (F.J.); (C.H.)
| |
Collapse
|
6
|
Iacobucci I, Monaco V, Cozzolino F, Monti M. From classical to new generation approaches: An excursus of -omics methods for investigation of protein-protein interaction networks. J Proteomics 2020; 230:103990. [PMID: 32961344 DOI: 10.1016/j.jprot.2020.103990] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/11/2020] [Accepted: 08/31/2020] [Indexed: 01/24/2023]
Abstract
Functional Proteomics aims to the identification of in vivo protein-protein interaction (PPI) in order to piece together protein complexes, and therefore, cell pathways involved in biological processes of interest. Over the years, proteomic approaches used for protein-protein interaction investigation have relied on classical biochemical protocols adapted to a global overview of protein-protein interactions, within so-called "interactomics" investigation. In particular, their coupling with advanced mass spectrometry instruments and innovative analytical methods led to make great strides in the PPIs investigation in proteomics. In this review, an overview of protein complexes purification strategies, from affinity purification approaches, including proximity-dependent labeling techniques and cross-linking strategy for the identification of transient interactions, to Blue Native Gel Electrophoresis (BN-PAGE) and Size Exclusion Chromatography (SEC) employed in the "complexome profiling", has been reported, giving a look to their developments, strengths and weakness and providing to readers several recent applications of each strategy. Moreover, a section dedicated to bioinformatic databases and platforms employed for protein networks analyses was also included.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- Department of Chemical Sciences, University Federico II of Naples, Strada Comunale Cinthia, 26, 80126 Naples, Italy; CEINGE Advanced Biotechnologies, Via G. Salvatore 486, 80145 Naples, Italy
| | - Vittoria Monaco
- CEINGE Advanced Biotechnologies, Via G. Salvatore 486, 80145 Naples, Italy
| | - Flora Cozzolino
- Department of Chemical Sciences, University Federico II of Naples, Strada Comunale Cinthia, 26, 80126 Naples, Italy; CEINGE Advanced Biotechnologies, Via G. Salvatore 486, 80145 Naples, Italy.
| | - Maria Monti
- Department of Chemical Sciences, University Federico II of Naples, Strada Comunale Cinthia, 26, 80126 Naples, Italy; CEINGE Advanced Biotechnologies, Via G. Salvatore 486, 80145 Naples, Italy.
| |
Collapse
|
7
|
Abstract
Mutations in approximately 80 genes have been implicated as the cause of various genetic kidney diseases. However, gene delivery to kidney cells from the blood is inefficient because of the natural filtering functions of the glomerulus, and research into and development of gene therapy directed toward kidney disease has lagged behind as compared with hepatic, neuromuscular, and ocular gene therapy. This lack of progress is in spite of numerous genetic mouse models of human disease available to the research community and many vectors in existence that can theoretically deliver genes to kidney cells with high efficiency. In the past decade, several groups have begun to develop novel injection techniques in mice, such as retrograde ureter, renal vein, and direct subcapsular injections to help resolve the issue of gene delivery to the kidney through the blood. In addition, the ability to retarget vectors specifically toward kidney cells has been underutilized but shows promise. This review discusses how recent advances in gene delivery to the kidney and the field of gene therapy can leverage the wealth of knowledge of kidney genetics to work toward developing gene therapy products for patients with kidney disease.
Collapse
Affiliation(s)
- Jeffrey D Rubin
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN, USA
| | - Michael A Barry
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Oncolytic Adenoviruses: Strategies for Improved Targeting and Specificity. Cancers (Basel) 2020; 12:cancers12061504. [PMID: 32526919 PMCID: PMC7352392 DOI: 10.3390/cancers12061504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is a major health problem. Most of the treatments exhibit systemic toxicity, as they are not targeted or specific to cancerous cells and tumors. Adenoviruses are very promising gene delivery vectors and have immense potential to deliver targeted therapy. Here, we review a wide range of strategies that have been tried, tested, and demonstrated to enhance the specificity of oncolytic viruses towards specific cancer cells. A combination of these strategies and other conventional therapies may be more effective than any of those strategies alone.
Collapse
|
9
|
Barry MA, Rubin JD, Lu SC. Retargeting adenoviruses for therapeutic applications and vaccines. FEBS Lett 2020; 594:1918-1946. [PMID: 31944286 PMCID: PMC7311308 DOI: 10.1002/1873-3468.13731] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022]
Abstract
Adenoviruses (Ads) are robust vectors for therapeutic applications and vaccines, but their use can be limited by differences in their in vitro and in vivo pharmacologies. This review emphasizes that there is not just one Ad, but a whole virome of diverse viruses that can be used as therapeutics. It discusses that true vector targeting involves not only retargeting viruses, but importantly also detargeting the viruses from off-target cells.
Collapse
Affiliation(s)
- Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Department of Immunology, Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey D Rubin
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Shao-Chia Lu
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Hagedorn C, Kreppel F. Capsid Engineering of Adenovirus Vectors: Overcoming Early Vector-Host Interactions for Therapy. Hum Gene Ther 2018; 28:820-832. [PMID: 28854810 DOI: 10.1089/hum.2017.139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Adenovirus-based vectors comprise the most frequently used vector type in clinical studies to date. Both intense lab research and insights from the clinical trials reveal the importance of a comprehensive understanding of vector-host interactions. Especially for systemic intravenous adenovirus vector delivery, it is paramount to develop safe and efficacious vectors. Very early vector-host interactions that take place in blood long before the first cell is being transduced are phenomena triggered by the surface, shape, and size of the adenovirus vector particles. Not surprisingly, a multitude of different technologies ranging from genetics to chemistry has been developed to alter the adenovirus vector surface. In this review, we discuss the most important technologies and evaluate them for their suitability to overcome hurdles imposed by early vector-host interactions.
Collapse
Affiliation(s)
- Claudia Hagedorn
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University , Witten, Germany
| | - Florian Kreppel
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University , Witten, Germany
| |
Collapse
|
11
|
Revaud J, Unterfinger Y, Rol N, Suleman M, Shaw J, Galea S, Gavard F, Lacour SA, Coulpier M, Versillé N, Havenga M, Klonjkowski B, Zanella G, Biacchesi S, Cordonnier N, Corthésy B, Ben Arous J, Richardson JP. Firewalls Prevent Systemic Dissemination of Vectors Derived from Human Adenovirus Type 5 and Suppress Production of Transgene-Encoded Antigen in a Murine Model of Oral Vaccination. Front Cell Infect Microbiol 2018; 8:6. [PMID: 29423380 PMCID: PMC5788964 DOI: 10.3389/fcimb.2018.00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/09/2018] [Indexed: 01/09/2023] Open
Abstract
To define the bottlenecks that restrict antigen expression after oral administration of viral-vectored vaccines, we tracked vectors derived from the human adenovirus type 5 at whole body, tissue, and cellular scales throughout the digestive tract in a murine model of oral delivery. After intragastric administration of vectors encoding firefly luciferase or a model antigen, detectable levels of transgene-encoded protein or mRNA were confined to the intestine, and restricted to delimited anatomical zones. Expression of luciferase in the form of multiple small bioluminescent foci in the distal ileum, cecum, and proximal colon suggested multiple crossing points. Many foci were unassociated with visible Peyer's patches, implying that transduced cells lay in proximity to villous rather than follicle-associated epithelium, as supported by detection of transgene-encoded antigen in villous epithelial cells. Transgene-encoded mRNA but not protein was readily detected in Peyer's patches, suggesting that post-transcriptional regulation of viral gene expression might limit expression of transgene-encoded antigen in this tissue. To characterize the pathways by which the vector crossed the intestinal epithelium and encountered sentinel cells, a fluorescent-labeled vector was administered to mice by the intragastric route or inoculated into ligated intestinal loops comprising a Peyer's patch. The vector adhered selectively to microfold cells in the follicle-associated epithelium, and, after translocation to the subepithelial dome region, was captured by phagocytes that expressed CD11c and lysozyme. In conclusion, although a large number of crossing events took place throughout the intestine within and without Peyer's patches, multiple firewalls prevented systemic dissemination of vector and suppressed production of transgene-encoded protein in Peyer's patches.
Collapse
Affiliation(s)
- Julien Revaud
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France.,SEPPIC Paris La Défense, Paris, France
| | - Yves Unterfinger
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Nicolas Rol
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Muhammad Suleman
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Julia Shaw
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Sandra Galea
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Françoise Gavard
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Sandrine A Lacour
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Muriel Coulpier
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | | | | | - Bernard Klonjkowski
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Gina Zanella
- Anses, Epidemiology Unit, Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | | | - Nathalie Cordonnier
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | - Jennifer P Richardson
- UMR Virologie INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| |
Collapse
|
12
|
Moghaddam-Taaheri P, Karlsson AJ. Protein Labeling in Live Cells for Immunological Applications. Bioconjug Chem 2018; 29:680-685. [DOI: 10.1021/acs.bioconjchem.7b00722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
13
|
Nguyen TV, Anguiano-Zarate SS, Matchett WE, Barry ME, Barry MA. Retargeted and detargeted adenovirus for gene delivery to the muscle. Virology 2017; 514:118-123. [PMID: 29172089 DOI: 10.1016/j.virol.2017.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 01/09/2023]
Abstract
We previously selected muscle binding peptides 12.51 and 12.52 from "context-specific" phage display libraries for introduction into adenovirus (Ad) vectors. In this work, these peptides were inserted into the hypervariable region (HVR) 5 loop of the Ad5 hexon protein to display 720 peptides per virions. HVR-12.51 and 12.52 increased transduction of C2C12 cells up to 20-fold when compared to unmodified Ad5. 12.51 increased in vivo muscle transduction 2 to 7-fold over unmodified Ad after intramuscular injection in mice and hamsters. 12.52 did not increase muscle transduction. Notably, insertion of 12.51 into the hexon reduced liver transduction 80-fold when compared to unmodified Ad5 after intravenous injection. Increased muscle transduction in mice translated into increased immune responses after gene-based vaccination. These data suggest there are merits to retargeting and detargeting benefits to modifying the hexons of Ads with peptide ligands.
Collapse
Affiliation(s)
- Tien V Nguyen
- Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN, United States; Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | | | - William E Matchett
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN, United States
| | - Mary E Barry
- Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN, United States; Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Michael A Barry
- Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN, United States; Department of Immunology, Mayo Clinic, Rochester, MN, United States; Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
14
|
The use of BirA-BAP system to study the effect of US2 and US11 on MHC class I heavy chain in cells. Immunol Lett 2017; 190:233-239. [PMID: 28860039 DOI: 10.1016/j.imlet.2017.08.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/06/2017] [Accepted: 08/25/2017] [Indexed: 11/24/2022]
Abstract
Biotinylation has been extensively used for antibody tagging, affinity-based purification, and in protein/DNA-protein interaction studies. Here we describe the use of biotinylation to study the turn-over of proteins in cells. We use the prokaryotic biotin ligase (BirA) to biotinylate the human leukocyte antigen (HLA)-A2 (A2) heavy chain (HC), which was engineered to contain a biotin acceptor peptide (BAP). Controlled availability of biotin in combination with visualization using streptavidin-conjugated peroxidase made it possible to detect biotinylated BAP-A2. Further, we exploited the effects of human cytomegalovirus (HCMV) unique short (US) proteins US2 and US11 on the turn-over of BAP-A2 HC. The full-length BAP-A2 HC and its mutants lacking either the cytosolic tail (tail-less) or both the transmembrane and cytosolic regions (soluble) were expressed via recombinant adenoviruses (rAd). The effect of US2, US11 and a control HCMV protein US9, also expressed via rAd, on each of the BAP- A2 forms was assessed. Experiments using this system showed that US2 and US11 cause proteasome-mediated degradation of full-length BAP-A2 HC but only US2 could cause degradation of tail-less BAP-A2. The results demonstrate that the technique of biotinylation can be used to study protein turn-over in cells.
Collapse
|
15
|
Calton CM, Bronnimann MP, Manson AR, Li S, Chapman JA, Suarez-Berumen M, Williamson TR, Molugu SK, Bernal RA, Campos SK. Translocation of the papillomavirus L2/vDNA complex across the limiting membrane requires the onset of mitosis. PLoS Pathog 2017; 13:e1006200. [PMID: 28463988 PMCID: PMC5412990 DOI: 10.1371/journal.ppat.1006200] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/25/2017] [Indexed: 11/20/2022] Open
Abstract
The human papillomavirus type 16 (HPV16) L2 protein acts as a chaperone to ensure that the viral genome (vDNA) traffics from endosomes to the trans-Golgi network (TGN) and eventually the nucleus, where HPV replication occurs. En route to the nucleus, the L2/vDNA complex must translocate across limiting intracellular membranes. The details of this critical process remain poorly characterized. We have developed a system based on subcellular compartmentalization of the enzyme BirA and its cognate substrate to detect membrane translocation of L2-BirA from incoming virions. We find that L2 translocation requires transport to the TGN and is strictly dependent on entry into mitosis, coinciding with mitotic entry in synchronized cells. Cell cycle arrest causes retention of L2/vDNA at the TGN; only release and progression past G2/M enables translocation across the limiting membrane and subsequent infection. Microscopy of EdU-labeled vDNA reveals a rapid and dramatic shift in vDNA localization during early mitosis. At late G2/early prophase vDNA egresses from the TGN to a pericentriolar location, accumulating there through prometaphase where it begins to associate with condensed chromosomes. By metaphase and throughout anaphase the vDNA is seen bound to the mitotic chromosomes, ensuring distribution into both daughter nuclei. Mutations in a newly defined chromatin binding region of L2 potently blocked translocation, suggesting that translocation is dependent on chromatin binding during prometaphase. This represents the first time a virus has been shown to functionally couple the penetration of limiting membranes to cellular mitosis, explaining in part the tropism of HPV for mitotic basal keratinocytes.
Collapse
Affiliation(s)
- Christine M. Calton
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Matthew P. Bronnimann
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Ariana R. Manson
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Shuaizhi Li
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson, Arizona, United States of America
| | - Janice A. Chapman
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Marcela Suarez-Berumen
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Tatum R. Williamson
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Sudheer K. Molugu
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Ricardo A. Bernal
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Samuel K. Campos
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
16
|
Zhang T, Suryawanshi YR, Woyczesczyk HM, Essani K. Targeting Melanoma with Cancer-Killing Viruses. Open Virol J 2017; 11:28-47. [PMID: 28567163 PMCID: PMC5420172 DOI: 10.2174/1874357901711010028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022] Open
Abstract
Melanoma is the deadliest skin cancer with ever-increasing incidence. Despite the development in diagnostics and therapies, metastatic melanoma is still associated with significant morbidity and mortality. Oncolytic viruses (OVs) represent a class of novel therapeutic agents for cancer by possessing two closely related properties for tumor reduction: virus-induced lysis of tumor cells and induction of host anti-tumor immune responses. A variety of viruses, either in "natural" or in genetically modified forms, have exhibited a remarkable therapeutic efficacy in regressing melanoma in experimental and/or clinical studies. This review provides a comprehensive summary of the molecular and cellular mechanisms of action of these viruses, which involve manipulating and targeting the abnormalities of melanoma, and can be categorized as enhancing viral tropism, targeting the tumor microenvironment and increasing the innate and adaptive antitumor responses. Additionally, this review describes the "biomarkers" and deregulated pathways of melanoma that are responsible for melanoma initiation, progression and metastasis. Advances in understanding these abnormalities of melanoma have resulted in effective targeted and immuno-therapies, and could potentially be applied for engineering OVs with enhanced oncolytic activity in future.
Collapse
Affiliation(s)
- Tiantian Zhang
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Yogesh R. Suryawanshi
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Helene M. Woyczesczyk
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| |
Collapse
|
17
|
Furin Cleavage of L2 during Papillomavirus Infection: Minimal Dependence on Cyclophilins. J Virol 2016; 90:6224-6234. [PMID: 27122588 DOI: 10.1128/jvi.00038-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/21/2016] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Despite an abundance of evidence supporting an important role for the cleavage of minor capsid protein L2 by cellular furin, direct cleavage of capsid-associated L2 during human papillomavirus 16 (HPV16) infection remains poorly characterized. The conserved cleavage site, close to the L2 N terminus, confounds observation and quantification of the small cleavage product by SDS-PAGE. To overcome this difficulty, we increased the size shift by fusing a compact protein domain, the Propionibacterium shermanii transcarboxylase domain (PSTCD), to the N terminus of L2. The infectious PSTCD-L2 virus displayed an appreciable L2 size shift during infection of HaCaT keratinocytes. Cleavage under standard cell culture conditions rarely exceeded 35% of total L2. Cleavage levels were enhanced by the addition of exogenous furin, and the absolute levels of infection correlated to the level of L2 cleavage. Cleavage occurred on both the HaCaT cell surface and extracellular matrix (ECM). Contrary to current models, experiments on the involvement of cyclophilins revealed little, if any, role for these cellular enzymes in the modulation of furin cleavage. HPV16 L2 contains two consensus cleavage sites, Arg5 (2RHKR5) and Arg12 (9RTKR12). Mutant PSTCD-L2 viruses demonstrated that although furin can cleave either site, cleavage must occur at Arg12, as cleavage at Arg5 alone is insufficient for successful infection. Mutation of the conserved cysteine residues revealed that the Cys22-Cys28 disulfide bridge is not required for cleavage. The PSTCD-L2 virus or similar N-terminal fusions will be valuable tools to study additional cellular and viral determinants of furin cleavage. IMPORTANCE Furin cleavage of minor capsid protein L2 during papillomavirus infection has been difficult to directly visualize and quantify, confounding efforts to study this important step of HPV infection. Fusion of a small protein domain to the N terminus greatly facilitates direct visualization of the cleavage product, revealing important characteristics of this critical process. Contrary to the current model, we found that cleavage is largely independent of cyclophilins, suggesting that cyclophilins act either in parallel to or downstream of furin to trigger exposure of a conserved N-terminal L2 epitope (RG-1) during infection. Based on this finding, we strongly caution against using L2 RG-1 epitope exposure as a convenient but indirect proxy of furin cleavage.
Collapse
|
18
|
Treatment of osteoarthritis using a helper-dependent adenoviral vector retargeted to chondrocytes. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16008. [PMID: 27626040 PMCID: PMC5008224 DOI: 10.1038/mtm.2016.8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 10/29/2015] [Accepted: 10/29/2015] [Indexed: 11/09/2022]
Abstract
Osteoarthritis (OA) is a joint disease characterized by degeneration of the articular cartilage, subchondral bone remodeling, and secondary inflammation. It is among the top three causes of chronic disability, and currently there are no treatment options to prevent disease progression. The localized nature of OA makes it an ideal candidate for gene and cell therapy. However, gene and cell therapy of OA is impeded by inefficient gene transduction of chondrocytes. In this study, we developed a broadly applicable system that retargets cell surface receptors by conjugating antibodies to the capsid of helper-dependent adenoviral vectors (HDVs). Specifically, we applied this system to retarget chondrocytes by conjugating an HDV to an α-10 integrin monoclonal antibody (a10mab). We show that a10mab-conjugated HDV (a10mabHDV)-infected chondrocytes efficiently in vitro and in vivo while detargeting other cell types. The therapeutic index of an intra-articular injection of 10mabHDV-expressing proteoglycan 4 (PRG4) into a murine model of post-traumatic OA was 10-fold higher than with standard HDV. Moreover, we show that PRG4 overexpression from articular, superficial zone chondrocytes is effective for chondroprotection in postinjury OA and that α-10 integrin is an effective protein for chondrocyte targeting.
Collapse
|
19
|
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
|
20
|
Jaykumar AB, Caceres PS, Sablaban I, Tannous BA, Ortiz PA. Real-time monitoring of NKCC2 endocytosis by total internal reflection fluorescence (TIRF) microscopy. Am J Physiol Renal Physiol 2015; 310:F183-91. [PMID: 26538436 DOI: 10.1152/ajprenal.00104.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 10/30/2015] [Indexed: 11/22/2022] Open
Abstract
The apical Na-K-2Cl cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb (TAL). The amount of NKCC2 at the apical membrane of TAL cells is determined by exocytic delivery, recycling, and endocytosis. Surface biotinylation allows measurement of NKCC2 endocytosis, but it has low time resolution and does not allow imaging of the dynamic process of endocytosis. We hypothesized that total internal reflection fluorescence (TIRF) microscopy imaging of labeled NKCC2 would allow monitoring of NKCC2 endocytosis in polarized Madin-Darby canine kidney (MDCK) and TAL cells. Thus we generated a NKCC2 construct containing a biotin acceptor domain (BAD) sequence between the transmembrane domains 5 and 6. Once expressed in polarized MDCK or TAL cells, surface NKCC2 was specifically biotinylated by exogenous biotin ligase (BirA). We also demonstrate that expression of a secretory form of BirA in TAL cells induces metabolic biotinylation of NKCC2. Labeling biotinylated surface NKCC2 with fluorescent streptavidin showed that most apical NKCC2 was located within small discrete domains or clusters referred to as "puncta" on the TIRF field. NKCC2 puncta were observed to disappear from the TIRF field, indicating an endocytic event which led to a decrease in the number of surface puncta at a rate of 1.18 ± 0.16%/min in MDCK cells, and a rate 1.09 ± 0.08%/min in TAL cells (n = 5). Treating cells with a cholesterol-chelating agent (methyl-β-cyclodextrin) completely blocked NKCC2 endocytosis. We conclude that TIRF microscopy of labeled NKCC2 allows the dynamic imaging of individual endocytic events at the apical membrane of TAL cells.
Collapse
Affiliation(s)
- Ankita Bachhawat Jaykumar
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan; and
| | - Paulo S Caceres
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan; and
| | - Ibrahim Sablaban
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pablo A Ortiz
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan; and
| |
Collapse
|
21
|
Weaver EA, Camacho ZT, Hillestad ML, Crosby CM, Turner MA, Guenzel AJ, Fadel HJ, Mercier GT, Barry MA. Mucosal vaccination by adenoviruses displaying reovirus sigma 1. Virology 2015; 482:60-6. [PMID: 25827529 DOI: 10.1016/j.virol.2015.02.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 02/17/2015] [Accepted: 02/25/2015] [Indexed: 10/23/2022]
Abstract
We developed adenovirus serotype 5 (Ad5) vectors displaying the sigma 1 protein from reovirus as mucosal vaccines. Ad5-sigma retargets to JAM-1 and sialic acid, but has 40-fold reduced gene delivery when compared to Ad5. While weaker at transduction, Ad5-sigma generates stronger T cell responses than Ad5 when used for mucosal immunization. In this work, new Ad5-fiber-sigma vectors were generated by varying the number of fiber β-spiral shaft repeats (R) between the fiber tail and sigma. Increasing chimera length led to decreasing insertion of these proteinsAd5 virions. Ad-R3 and R14 vectors effectively targeted JAM-1 in vitro while R20 did not. When wereused to immunize mice by the intranasal route, Ad5-R3-sigma produced higher serum and vaginal antibody responses than Ad5. These data suggest optimized Ad-sigma vectors may be useful vectors for mucosal vaccination.
Collapse
Affiliation(s)
- Eric A Weaver
- Department of Internal Medicine, Division of Infectious Diseases, Translational Immunovirology and Biodefense Program, Mayo Clinic, Rochester, MN 55902, USA
| | - Zenaido T Camacho
- Department of Cell Biology, Department of Natural Sciences, Western New Mexico University, Silver City, NM 88062, USA
| | | | - Catherine M Crosby
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN 55902, USA
| | - Mallory A Turner
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN 55902, USA
| | - Adam J Guenzel
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN 55902, USA
| | - Hind J Fadel
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN 55902, USA
| | - George T Mercier
- Department of Physics, University of Houston, Houston, TX 77004, USA
| | - Michael A Barry
- Department of Internal Medicine, Division of Infectious Diseases, Translational Immunovirology and Biodefense Program, Mayo Clinic, Rochester, MN 55902, USA; Department of Immunology and Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA.
| |
Collapse
|
22
|
Su Y, Xie Z, Kim GB, Dong C, Yang J. Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 2015; 1:201-217. [PMID: 25984572 PMCID: PMC4428174 DOI: 10.1021/ab500179h] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Drug delivery systems, particularly nanomaterial-based drug delivery systems, possess a tremendous amount of potential to improve diagnostic and therapeutic effects of drugs. Controlled drug delivery targeted to a specific disease is designed to significantly improve the pharmaceutical effects of drugs and reduce their side effects. Unfortunately, only a few targeted drug delivery systems can achieve high targeting efficiency after intravenous injection, even with the development of numerous surface markers and targeting modalities. Thus, alternative drug and nanomedicine targeting approaches are desired. Circulating cells, such as erythrocytes, leukocytes, and stem cells, present innate disease sensing and homing properties. Hence, using living cells as drug delivery carriers has gained increasing interest in recent years. This review highlights the recent advances in the design of cell-mediated drug delivery systems and targeting mechanisms. The approaches of drug encapsulation/conjugation to cell-carriers, cell-mediated targeting mechanisms, and the methods of controlled drug release are elaborated here. Cell-based "live" targeting and delivery could be used to facilitate a more specific, robust, and smart payload distribution for the next-generation drug delivery systems.
Collapse
Affiliation(s)
- Yixue Su
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Zhiwei Xie
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Gloria B. Kim
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Cheng Dong
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| |
Collapse
|
23
|
Abstract
Simian immunodeficiency virus (SIV) is a robust pathogen used in non-human primates to model HIV vaccines. SIV encodes a number of potential vaccine targets. By far the largest and most conserved protein target in SIV is its gag-pol protein that bears many epitopes to drive multivalent immune T cell responses. While gag-pol is an attractive antigen, it is only translated after a frame shift between gag and pol with the effect that gag and pol are expressed at an approximate 10/1 ratio. The codon bias of native lentiviral genes are also mismatched with the abundance of tRNAs in mammalian cells resulting in poor expression of unmodified SIV genes. To provide a better SIV gag-pol immunogen for gene-based vaccination, we codon-optimized the full gag-pol sequence from SIVmac239. To increase pol expression, we artificially moved the pol sequence in frame to gag to bypass the need for a translational frame shift for its expression. Finally, we inserted four "self-cleaving" picornavirus sequences into gag p24, protease, reverse transcriptase, and into integrase to fragment the proteins for potentially better immune presentation. We demonstrate that these immunogens are well expressed in vitro and drive similar antibody and T cell responses with or without cleavage sequences.
Collapse
|
24
|
Leoh LS, Morizono K, Kershaw KM, Chen ISY, Penichet ML, Daniels-Wells TR. Gene delivery in malignant B cells using the combination of lentiviruses conjugated to anti-transferrin receptor antibodies and an immunoglobulin promoter. J Gene Med 2014; 16:11-27. [PMID: 24436117 DOI: 10.1002/jgm.2754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/05/2013] [Accepted: 01/09/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND We previously developed an antibody-avidin fusion protein (ch128.1Av) specific for the human transferrin receptor 1 (TfR1; CD71) to be used as a delivery vector for cancer therapy and showed that ch128.1Av delivers the biotinylated plant toxin saporin-6 into malignant B cells. However, as a result of widespread expression of TfR1, delivery of the toxin to normal cells is a concern. Therefore, we explored the potential of a dual targeted lentiviral-mediated gene therapy strategy to restrict gene expression to malignant B cells. Targeting occurs through the use of ch128.1Av or its parental antibody without avidin (ch128.1) and through transcriptional regulation using an immunoglobulin promoter. METHODS Flow cytometry was used to detect the expression of enhanced green fluorescent protein (EGFP) in a panel of cell lines. Cell viability after specific delivery of the therapeutic gene FCU1, a chimeric enzyme consisting of cytosine deaminase genetically fused to uracil phosphoribosyltransferse that converts the 5-fluorocytosine (5-FC) prodrug into toxic metabolites, was monitored using the MTS or WST-1 viability assay. RESULTS We found that EGFP was specifically expressed in a panel of human malignant B-cell lines, but not in human malignant T-cell lines. EGFP expression was observed in all cell lines when a ubiquitous promoter was used. Furthermore, we show the decrease of cell viability in malignant plasma cells in the presence of 5-FC and the FCU1 gene. CONCLUSIONS The present study demonstrates that gene expression can be restricted to malignant B cells and suggests that this dual targeted gene therapy strategy may help to circumvent the potential side effects of certain TfR1-targeted protein delivery approaches.
Collapse
Affiliation(s)
- Lai Sum Leoh
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
25
|
Carozza M, Rodrigues V, Unterfinger Y, Galea S, Coulpier M, Klonjkowski B, Thiaucourt F, Totté P, Richardson J. An adenoviral vector expressing lipoprotein A, a major antigen of Mycoplasma mycoides subspecies mycoides, elicits robust immune responses in mice. Vaccine 2014; 33:141-8. [PMID: 25444801 DOI: 10.1016/j.vaccine.2014.10.088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 10/24/2022]
Abstract
Contagious bovine pleuropneumonia (CBPP), caused by Mycoplasma mycoides subsp. mycoides small colony type (MmmSC), is a devastating respiratory disease of cattle. In sub-Saharan Africa, where CBPP is enzootic, live attenuated vaccines are deployed but afford only short-lived protection. In cattle, recovery from experimental MmmSC infection has been associated with the presence of CD4(+) T lymphocytes that secrete interferon gamma in response to MmmSC, and in particular to the lipoprotein A (LppA) antigen. In an effort to develop a better vaccine against CBPP, a viral vector (Ad5-LppA) that expressed LppA was generated from human adenovirus type 5. The LppA-specific immune responses elicited by the Ad5-LppA vector were evaluated in mice, and compared to those elicited by recombinant LppA formulated with a potent adjuvant. Notably, a single administration of Ad5-LppA, but not recombinant protein, sufficed to elicit a robust LppA-specific humoral response. After a booster administration, both vector and recombinant protein elicited strong LppA-specific humoral and cell-mediated responses. Ex vivo stimulation of splenocytes induced extensive proliferation of CD4(+) T cells for mice immunized with vector or protein, and secretion of T helper 1-associated and proinflammatory cytokines for mice immunized with Ad5-LppA. Our study - by demonstrating the potential of a viral-vectored prototypic vaccine to elicit prompt and robust immune responses against a major antigen of MmmSC - represents a first step in developing a recombinant vaccine against CBPP.
Collapse
Affiliation(s)
- Marlène Carozza
- Centre International de Recherche en Agronomie pour le Développement, UMR CMAEE, Montpellier, France; INRA, UMR 1309 CMAEE, Montpellier, France; INRA, UMR 1161 Virologie, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; ANSES, UMR Virologie, 23 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UMR Virologie, Maisons-Alfort F-94704, France
| | - Valérie Rodrigues
- Centre International de Recherche en Agronomie pour le Développement, UMR CMAEE, Montpellier, France; INRA, UMR 1309 CMAEE, Montpellier, France
| | - Yves Unterfinger
- INRA, UMR 1161 Virologie, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; ANSES, UMR Virologie, 23 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UMR Virologie, Maisons-Alfort F-94704, France
| | - Sandra Galea
- INRA, UMR 1161 Virologie, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; ANSES, UMR Virologie, 23 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UMR Virologie, Maisons-Alfort F-94704, France
| | - Muriel Coulpier
- INRA, UMR 1161 Virologie, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; ANSES, UMR Virologie, 23 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UMR Virologie, Maisons-Alfort F-94704, France
| | - Bernard Klonjkowski
- INRA, UMR 1161 Virologie, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; ANSES, UMR Virologie, 23 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UMR Virologie, Maisons-Alfort F-94704, France
| | - François Thiaucourt
- Centre International de Recherche en Agronomie pour le Développement, UMR CMAEE, Montpellier, France; INRA, UMR 1309 CMAEE, Montpellier, France
| | - Philippe Totté
- Centre International de Recherche en Agronomie pour le Développement, UMR CMAEE, Montpellier, France; INRA, UMR 1309 CMAEE, Montpellier, France
| | - Jennifer Richardson
- INRA, UMR 1161 Virologie, 7 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; ANSES, UMR Virologie, 23 avenue du Général de Gaulle, 94700 Maisons-Alfort, France; Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UMR Virologie, Maisons-Alfort F-94704, France.
| |
Collapse
|
26
|
Streptavidin–biotin technology: improvements and innovations in chemical and biological applications. Appl Microbiol Biotechnol 2013; 97:9343-53. [DOI: 10.1007/s00253-013-5232-z] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 12/25/2022]
|
27
|
Sapsford KE, Algar WR, Berti L, Gemmill KB, Casey BJ, Oh E, Stewart MH, Medintz IL. Functionalizing nanoparticles with biological molecules: developing chemistries that facilitate nanotechnology. Chem Rev 2013; 113:1904-2074. [PMID: 23432378 DOI: 10.1021/cr300143v] [Citation(s) in RCA: 854] [Impact Index Per Article: 71.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kim E Sapsford
- Division of Biology, Department of Chemistry and Materials Science, Office of Science and Engineering Laboratories, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Cancer gene therapy approaches have benefited greatly from the utilization of molecular-based therapeutics. Of these, adenovirus-based interventions hold much promise as a platform for targeted therapeutic delivery to tumors. However, a barrier to this progression is the lack of native adenovirus receptor expression on a variety of cancer types. As such, any adenovirus-based cancer therapy must take into consideration retargeting the vector to nonnative cellular surface receptors. Predicated upon the knowledge gained in native adenovirus biology, several strategies to transductionally retarget adenovirus have emerged. Herein, we describe the biological hurdles as well as strategies utilized in adenovirus transductional targeting, covering the progress of both adapter-based and genetic manipulation-based targeting. Additionally, we discuss recent translation of these targeting strategies into a clinical setting.
Collapse
Affiliation(s)
- Matthew S Beatty
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | | |
Collapse
|
29
|
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
|
30
|
Murugan S, Saarela U, Airenne K, Shan J, Skovorodkin I, Ylä-Herttuala S, Vainio SJ. Conditional expression of Lodavin, an avidin-tagged LDL receptor, for biotin-mediated applications in vivo. Genesis 2012; 50:693-9. [PMID: 22467513 DOI: 10.1002/dvg.22028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/20/2012] [Accepted: 03/22/2012] [Indexed: 11/12/2022]
Abstract
Lodavin represents an engineered fusion protein that consists of a cytoplasmic and a transmembrane domain of the human low-density lipoprotein receptor coupled to an extracellular avidin monomer. Biotinylated compounds have been successfully targeted to Lodavin-expressing cells that have been transduced by a Lodavin-containing virus, and the targeting is based on the high affinity between biotin and avidin. We engineered a Rosa26 (R26R) knock-in Lodavin mouse to develop biotin-based applications such as targeted drug delivery, cell purification, and tissue imaging in vivo. A cDNA encoding Lodavin was inserted downstream of a floxed βgeo resistance gene in the R26R locus in embryonic stem cells, and a germ line-derived R26RLodavin mouse line was generated. Efficient removal of the floxed βgeo cassette and conditional activation of Lodavin expression was achieved as a result of crossing the R26RLodavin mice with HoxB7-Cre, Wnt4-Cre, or Tie1-Cre mice. In summary, the R26RLodavin mouse line may provide a useful tool for testing and developing applications with the aid of avidin and biotin interaction.
Collapse
Affiliation(s)
- Subramanian Murugan
- Oulu Centre for Cell-Matrix Research, Biocenter Oulu, Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | |
Collapse
|
31
|
Akter F, Mie M, Kobatake E. Immuno-rolling circle amplification using a multibinding fusion protein. Anal Biochem 2011; 416:174-9. [DOI: 10.1016/j.ab.2011.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 05/02/2011] [Accepted: 05/03/2011] [Indexed: 10/18/2022]
|
32
|
Khare R, Chen CY, Weaver EA, Barry MA. Advances and future challenges in adenoviral vector pharmacology and targeting. Curr Gene Ther 2011; 11:241-58. [PMID: 21453281 PMCID: PMC3267160 DOI: 10.2174/156652311796150363] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/14/2011] [Accepted: 03/15/2011] [Indexed: 11/26/2022]
Abstract
Adenovirus is a robust vector for therapeutic applications, but its use is limited by our understanding of its complex in vivo pharmacology. In this review we describe the necessity of identifying its natural, widespread, and multifaceted interactions with the host since this information will be crucial for efficiently redirecting virus into target cells. In the rational design of vectors, the notion of overcoming a sequence of viral "sinks" must be combined with re-targeting to target populations with capsid as well as shielding the vectors from pre-existing or toxic immune responses. It must also be noted that most known adenoviral pharmacology is deduced from the most commonly used serotypes, Ad5 and Ad2. However, these serotypes may not represent all adenoviruses, and may not even represent the most useful vectors for all purposes. Chimeras between Ad serotypes may become useful in engineering vectors that can selectively evade substantial viral traps, such as Kupffer cells, while retaining the robust qualities of Ad5. Similarly, vectorizing other Ad serotypes may become useful in avoiding immunity against Ad5 altogether. Taken together, this research on basic adenovirus biology will be necessary in developing vectors that interact more strategically with the host for the most optimal therapeutic effect.
Collapse
Affiliation(s)
- Reeti Khare
- Virology and Gene Therapy Program, Mayo Graduate School
| | - Christopher Y Chen
- Department of Medicine, Division of Infectious Diseases, Translational Immunovirology and Biodefense Program
| | - Eric A Weaver
- Department of Medicine, Division of Infectious Diseases, Translational Immunovirology and Biodefense Program
| | - Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Translational Immunovirology and Biodefense Program
- Department of Molecular Medicine, Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
33
|
Viral delivery for gene therapy against cell movement in cancer. Adv Drug Deliv Rev 2011; 63:671-7. [PMID: 21616108 DOI: 10.1016/j.addr.2011.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/15/2011] [Accepted: 05/07/2011] [Indexed: 12/17/2022]
Abstract
Viral delivery for cancer gene therapy is a promising approach, where traditional radiotherapy or chemotherapy to limit proliferation and movement of cancer cells has met resistance. Based on the new understanding of the biology of the viral vectors, therapeutic viral vectors for cancer gene therapy have been improved for greater safety and efficacy as well as transitioned from being non-replicating to replication-competent. Traditional oncolytic vectors have focused on eliminating tumor growth, while novel vectors simultaneously target epithelial-to-mesenchymal transition (EMT) in cancer cells, which could further prevent and reverse the aggressive tumor progression. In this review, we highlight the illustrative examples of cancer gene therapy in clinical trials as well as preclinical data and include proposals on methods to further enhance the safety and efficacy of oncolytic viral vectors in cancer gene therapy.
Collapse
|
34
|
Selective transduction of dendritic cells in human lymph nodes and superior induction of high-avidity melanoma-reactive cytotoxic T cells by a CD40-targeted adenovirus. J Immunother 2011; 33:706-15. [PMID: 20664356 DOI: 10.1097/cji.0b013e3181eccbd4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Targeted delivery of tumor antigen genes to dendritic cells (DCs) using adenoviral (Ad) vectors holds great potential for cancer immunotherapy. We previously showed that CD40 targeting of Ad vectors enhanced specific transduction of DC in human skin, while simultaneously ensuring their stable maturation and superior allogeneic T-cell stimulatory capacity. In this study, we evaluated whether CD40-targeted Ad encoding the full-length melanoma antigen recognized by T cells-1 (CD40-Ad-MART-1) could be used to efficiently and selectively transduce conventional and plasmacytoid DC to prime melanoma-specific CD8(+) T-effector cells in human melanoma-draining sentinel lymph nodes (SLNs). CD40 targeting of Ad was achieved using a bispecific fusion protein, binding and neutralizing the Ad fiber knob through soluble coxsackie and adenovirus receptor while retargeting the virus to hCD40 through the tumor necrosis factor-like domain of mCD40L. Selective transduction of conventional and plasmacytoid DC subsets by CD40-Ad was observed in suspensions of human melanoma-draining SLN. Moreover, CD40-Ad-MART-1 enhanced the expansion of functional MART-1-specific CD8(+) T cells from SLN with concomitant decreases in CD4:CD8 T-cell ratios and CD4(+)CD25(hi)FoxP3(+) regulatory T-cell rates. Additional studies revealed that transduction and activation of monocyte-derived DCs with CD40-Ad-MART-1 significantly enhanced their priming efficiency of functional CD8(+) effector T cells with high avidity. These findings provide preclinical evidence of possible efficacy of this approach for cancer immunotherapy.
Collapse
|
35
|
Unnatural amino acid incorporation onto adenoviral (Ad) coat proteins facilitates chemoselective modification and retargeting of Ad type 5 vectors. J Virol 2011; 85:7546-54. [PMID: 21613404 DOI: 10.1128/jvi.00118-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Surface modification of adenovirus vectors can improve tissue-selective targeting, attenuate immunogenicity, and enable imaging of particle biodistribution, thus significantly improving therapeutic potential. Currently, surface engineering is constrained by a combination of factors, including impact on viral fitness, limited access to functionality, or incomplete control over the site of modification. Here, we report a two-step labeling process involving an initial metabolic placement of a uniquely reactive unnatural amino acid, azidohomoalanine (Aha), followed by highly specific chemical modification. As genetic modification of adenovirus is unnecessary, vector production is exceedingly straightforward. Aha incorporation demonstrated no discernible impact on either virus production or infectivity of the resultant particles. "Click" chemical modification of surface-exposed azides was highly selective, allowing for the attachment of a wide range of functionality. Decoration of human adenovirus type 5 (hAd5) with folate, a known cancer-targeting moiety, provided an ∼20-fold increase in infection of murine breast cancer cells (4T1) in a folate receptor-dependent manner. This study demonstrates that incorporation of unnatural amino acids can provide a flexible, straightforward route for the selective chemical modification of adenoviral vectors.
Collapse
|
36
|
Niers JM, Chen JW, Weissleder R, Tannous BA. Enhanced in vivo imaging of metabolically biotinylated cell surface reporters. Anal Chem 2011; 83:994-9. [PMID: 21214190 DOI: 10.1021/ac102758m] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metabolic biotinylation of intracellular and secreted proteins as well as surface receptors in mammalian cells provides a versatile way to monitor gene expression; to purify and target viral vectors; to monitor cell and tumor distribution in real time in vivo; to label cells for isolation; and to tag proteins for purification, localization, and trafficking. Here, we show that metabolic biotinylation of proteins fused to the bacterial biotin acceptor peptides (BAP) varies among different mammalian cell types and can be enhanced by over 10-fold upon overexpression of the bacterial biotin ligase directed to the same cellular compartment as the fusion protein. We also show that in vivo imaging of metabolically biotinylated cell surface receptors using streptavidin conjugates is significantly enhanced upon coexpression of bacterial biotin ligase in the secretory pathway. These findings have practical applications in designing more efficient targeting and imaging strategies.
Collapse
Affiliation(s)
- Johanna M Niers
- Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
37
|
Cannon JP, O'Driscoll M, Litman GW. Construction, expression, and purification of chimeric protein reagents based on immunoglobulin fc regions. Methods Mol Biol 2011; 748:51-67. [PMID: 21701966 DOI: 10.1007/978-1-61779-139-0_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recombinant fusion proteins incorporating experimental protein domains fused to immunoglobulin Fc regions have become widely utilized in studies of protein-ligand interactions. The advantages of these systems include an inherent increase in avidity provided by the multimerization of Fc regions, combined with robust detection methods based on numerous commercially available secondary reagents directed against the Fc tag. We describe a set of methods for subcloning, expression, and purification of chimeric protein reagents containing a protein domain (or domains) of interest fused to a C-terminal moiety derived from the Fc region of either IgG or IgM.
Collapse
Affiliation(s)
- John P Cannon
- Department of Pediatrics, University of South Florida College of Medicine, University of South Florida and All Children's Hospital Children's Research Institute, St. Petersburg, FL, USA.
| | | | | |
Collapse
|
38
|
Segura MM, Kamen AA, Garnier A. Overview of current scalable methods for purification of viral vectors. Methods Mol Biol 2011; 737:89-116. [PMID: 21590394 DOI: 10.1007/978-1-61779-095-9_4] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As a result of the growing interest in the use of viruses for gene therapy and vaccines, many virus-based products are being developed. The manufacturing of viruses poses new challenges for process developers and regulating authorities that need to be addressed to ensure quality, efficacy, and safety of the final product. The design of suitable purification strategies will depend on a multitude of variables including the vector production system and the nature of the virus. In this chapter, we provide an overview of the most commonly used purification methods for viral gene therapy vectors. Current chromatography options available for large-scale purification of γ-retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes simplex virus, baculovirus, and poxvirus vectors are presented.
Collapse
Affiliation(s)
- María Mercedes Segura
- Department of Biochemistry and Molecular Biology, Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | | | | |
Collapse
|
39
|
Shin S, Salvay DM, Shea LD. Lentivirus delivery by adsorption to tissue engineering scaffolds. J Biomed Mater Res A 2010; 93:1252-9. [PMID: 19827108 DOI: 10.1002/jbm.a.32619] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Biomaterial scaffolds capable of localized gene delivery are being investigated for numerous regenerative medicine applications and as model systems for fundamental studies of tissue formation. In this manuscript, we investigate the delivery of lentivirus from a tissue engineering scaffold using a surface immobilization strategy. Poly(lactide-co-glycolide) (PLG) was employed as the biomaterial for delivery, which has been widely used for a number of tissue engineering applications. The virus was immobilized by freezing and subsequent lyophilization of the virus with the scaffold. The presence of sucrose during freezing and lyophilization maintained the activity of the lentivirus, and was similar to an adenovirus control. Collagen and fibronectin were investigated for their ability to enhance surface immobilization. Fibronectin modestly increased binding and transduction of the adenovirus, yet did not significantly impact the lentivirus delivery. Most of the immobilized lentivirus was released from the scaffold within 24 h. In vivo implantation of the scaffolds yielded transgene expression that persisted for at least 4 weeks. These findings indicate the potential for delivering lentivirus from tissue engineering scaffolds using a surface immobilization strategy. To our knowledge, this report is the first to investigate lentivirus delivery from porous tissue engineering scaffolds. Delivery of lentiviral vectors from PLG scaffolds could provide an efficient and versatile gene delivery system for use with in vitro and in vivo models of tissue formation, and ultimately for therapeutic applications.
Collapse
Affiliation(s)
- Seungjin Shin
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road E156, Evanston, Illinois 60208-3120, USA
| | | | | |
Collapse
|
40
|
Liu Y, Valadon P, Schnitzer JE. Construction of metabolically biotinylated adenovirus with deleted fiber knob as targeting vector. Virol J 2010; 7:316. [PMID: 21073735 PMCID: PMC2993673 DOI: 10.1186/1743-422x-7-316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 11/12/2010] [Indexed: 11/10/2022] Open
Abstract
Gene delivery vectors based on adenovirus, particularly human adenovirus serotype 5 (hAd5) have great potential for the treatment of variety of diseases. However, the tropism of hAd5 needs to be modified to achieve tissue- or cell- specific therapies for the successful application of this vector system to clinic. Here, we modified hAd5 tropism by replacing the fiber knob which contains the coxsackievirus B and adenovirus receptor (CAR)-binding sites with a biotin acceptor peptide, a truncated form of Propionibacterium shermanii 1.3 S transcarboxylase domain (PSTCD), to enable metabolically biotinylation of the virus. We demonstrate here that the new adenovirus no longer shows CAR-dependent cell uptake and transduction. When metabolically biotinylated and avidin-coated, it forms a nano-complex that can be retargeted to distinct cells using biotinylated antibodies. This vector may prove useful in the path towards achieving targeted gene delivery.
Collapse
Affiliation(s)
- Yanzheng Liu
- Proteogenomics Research Institute for Systems Medicine, 11107 Roselle St., San Diego, CA 92121, USA
| | | | | |
Collapse
|
41
|
Tropism-modification strategies for targeted gene delivery using adenoviral vectors. Viruses 2010; 2:2290-2355. [PMID: 21994621 PMCID: PMC3185574 DOI: 10.3390/v2102290] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 10/07/2010] [Indexed: 02/08/2023] Open
Abstract
Achieving high efficiency, targeted gene delivery with adenoviral vectors is a long-standing goal in the field of clinical gene therapy. To achieve this, platform vectors must combine efficient retargeting strategies with detargeting modifications to ablate native receptor binding (i.e. CAR/integrins/heparan sulfate proteoglycans) and “bridging” interactions. “Bridging” interactions refer to coagulation factor binding, namely coagulation factor X (FX), which bridges hepatocyte transduction in vivo through engagement with surface expressed heparan sulfate proteoglycans (HSPGs). These interactions can contribute to the off-target sequestration of Ad5 in the liver and its characteristic dose-limiting hepatotoxicity, thereby significantly limiting the in vivo targeting efficiency and clinical potential of Ad5-based therapeutics. To date, various approaches to retargeting adenoviruses (Ad) have been described. These include genetic modification strategies to incorporate peptide ligands (within fiber knob domain, fiber shaft, penton base, pIX or hexon), pseudotyping of capsid proteins to include whole fiber substitutions or fiber knob chimeras, pseudotyping with non-human Ad species or with capsid proteins derived from other viral families, hexon hypervariable region (HVR) substitutions and adapter-based conjugation/crosslinking of scFv, growth factors or monoclonal antibodies directed against surface-expressed target antigens. In order to maximize retargeting, strategies which permit detargeting from undesirable interactions between the Ad capsid and components of the circulatory system (e.g. coagulation factors, erythrocytes, pre-existing neutralizing antibodies), can be employed simultaneously. Detargeting can be achieved by genetic ablation of native receptor-binding determinants, ablation of “bridging interactions” such as those which occur between the hexon of Ad5 and coagulation factor X (FX), or alternatively, through the use of polymer-coated “stealth” vectors which avoid these interactions. Simultaneous retargeting and detargeting can be achieved by combining multiple genetic and/or chemical modifications.
Collapse
|
42
|
Hofherr SE, Senac JS, Chen CY, Palmer DJ, Ng P, Barry MA. Short-term rescue of neonatal lethality in a mouse model of propionic acidemia by gene therapy. Hum Gene Ther 2010; 20:169-80. [PMID: 19025475 DOI: 10.1089/hum.2008.158] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Propionic acidemia (PA) is a metabolic disorder that causes mental retardation and that can be fatal if untreated. PA is inherited in an autosomal recessive fashion involving mutations in PCCA or PCCB encoding the alpha and beta subunits of propionyl-CoA carboxylase (PCC). Current treatment is based on dietary restriction of substrate amino acids, which attenuates symptoms. However, patients still experience episodes of hyperammonemia that can cause progressive neurologic damage. In this paper, we have tested gene therapy approaches to PA in a stringent mouse model of PCCA deficiency, in which homozygous knockout mice are born but die within 36 hr. In this work, we have delivered first-generation and helper-dependent adenovirus serotype 5 (Ad5) vectors expressing the human PCCA cDNA by intraperitoneal injection into newborn mice. Unmodified Ad5 vectors mediated extensive transduction of the peritoneum with weak liver transduction as determined by luciferase imaging and dsRed expression. In contrast, modification of Ad5 with polyethylene glycol detargeted the virus from the peritoneum and retargeted it for transduction in the liver. When vectors expressing PCCA were injected, significant increases in life span were observed for both the unmodified and polyethylene glycol (PEG)-modified Ad5 vectors. However, this rescue was transient. Similarly, adeno-associated virus serotype 8-mediated transduction also produced only transient rescue. These data show first proof of principle for gene therapy of PA and demonstrate the potential utility of PEG to modify viral tropism in an actual gene therapy application.
Collapse
Affiliation(s)
- Sean E Hofherr
- Division of Infectious Diseases, Department of Internal Medicine, and Translational Immunovirology Program, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
43
|
Lesch HP, Kaikkonen MU, Pikkarainen JT, Ylä-Herttuala S. Avidin-biotin technology in targeted therapy. Expert Opin Drug Deliv 2010; 7:551-64. [PMID: 20233034 DOI: 10.1517/17425241003677749] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD The goal of drug targeting is to increase the concentration of the drug in the vicinity of the cells responsible for disease without affecting healthy cells. Many approaches in cancer treatment are limited because of their broad range of unwanted side effects on healthy cells. Targeting can reduce side effects and increase efficacy of drugs in the patient. AREAS COVERED IN THIS REVIEW Avidin, originally isolated from chicken eggs, and its bacterial analogue, streptavidin, from Streptomyces avidinii, have extremely high affinity for biotin. This unique feature is the basis of avidin-biotin technology. This article reviews the current status of avidin-biotin systems and their use for pretargeted drug delivery and vector targeting. WHAT THE READER WILL GAIN The reader will gain an understanding of the following approaches using the avidin-biotin system: i) targeting antibodies and therapeutic molecules are administered separately leading to a reduction of drug dose in normal tissues compared with conventional (radio)immunotherapies; ii) introducing avidin gene into specific tissues by local gene transfer, which subsequently can sequester and concentrate considerable amounts of therapeutic ligands; and iii) enabling transductional targeting of gene therapy vectors. TAKE HOME MESSAGE Avidin and biotin technology has proved to be an extremely versatile tool with broad applications, such as pretargeting, delivering avidin gene into cells enabling targeting of biotinylated compounds and targeting of viral vectors.
Collapse
Affiliation(s)
- Hanna P Lesch
- University of Eastern Finland, A.I. Virtanen institute, Department of Biotechnology and Molecular Medicine, Kuopio, Finland
| | | | | | | |
Collapse
|
44
|
Chen CY, May SM, Barry MA. Targeting adenoviruses with factor x-single-chain antibody fusion proteins. Hum Gene Ther 2010; 21:739-49. [PMID: 20331369 DOI: 10.1089/hum.2009.190] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract It has been shown that blood clotting factors, including factor X (FX), bind to the adenovirus serotype 5 (Ad5) hexon protein and target the virus to liver hepatocytes after intravenous injection. These factors bind to hexon via their conserved vitamin K-dependent gamma-carboxyglutamic acid (GLA) domains with subnanomolar affinity. In this work, we have used this strong interaction to retarget Ad to new receptors, using the GLA domain of FX fused to single-chain antibody variable fragment (ScFv). We demonstrate that fusion of the GLA domain of human FX to receptor-specific ScFvs will target Ad5 vectors to cells expressing these receptors. Fusion of an alphaHer2 ScFv to GLA increased in vitro transduction of Her2-positive versus Her2-negative cells when compared with untargeted virus. Similar results were obtained with ScFvs against the epidermal growth factor receptor (EGFR) and against the stem cell marker ATP-binding cassette protein G2 (ABCG2). Direct expression of GLA fusion protein from replication-defective or replication-competent Ad increased infection and killing of cancer cells in vitro and in vivo. These data demonstrate the potential of using GLA domains to bridge secreted ligands with intracellularly produced Ad5 vectors for vector targeting.
Collapse
Affiliation(s)
- Christopher Y Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
45
|
Howarth JL, Lee YB, Uney JB. Using viral vectors as gene transfer tools (Cell Biology and Toxicology Special Issue: ETCS-UK 1 day meeting on genetic manipulation of cells). Cell Biol Toxicol 2009; 26:1-20. [PMID: 19830583 PMCID: PMC2817806 DOI: 10.1007/s10565-009-9139-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 09/24/2009] [Indexed: 02/06/2023]
Abstract
In recent years, the development of powerful viral gene transfer techniques has greatly facilitated the study of gene function. This review summarises some of the viral delivery systems routinely used to mediate gene transfer into cell lines, primary cell cultures and in whole animal models. The systems described were originally discussed at a 1-day European Tissue Culture Society (ETCS-UK) workshop that was held at University College London on 1st April 2009. Recombinant-deficient viral vectors (viruses that are no longer able to replicate) are used to transduce dividing and post-mitotic cells, and they have been optimised to mediate regulatable, powerful, long-term and cell-specific expression. Hence, viral systems have become very widely used, especially in the field of neurobiology. This review introduces the main categories of viral vectors, focusing on their initial development and highlighting modifications and improvements made since their introduction. In particular, the use of specific promoters to restrict expression, translational enhancers and regulatory elements to boost expression from a single virion and the development of regulatable systems is described.
Collapse
|
46
|
Weaver EA, Barry MA. Effects of shielding adenoviral vectors with polyethylene glycol on vector-specific and vaccine-mediated immune responses. Hum Gene Ther 2009; 19:1369-82. [PMID: 18778197 DOI: 10.1089/hum.2008.091] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Many individuals have been previously exposed to human adenovirus serotype 5 (Ad5). This prior immunity has long been known to hinder its use for gene therapy and as a gene-based vaccine. Given these immunogenicity problems, we have tested whether polyethylene glycol (PEG) can blunt immune effects against Ad5 during systemic and mucosal vaccination. Ad5 vectors were covalently modified with 5-, 20-, and 35-kDa linear PEG polymers and evaluated for their ability to produce immune responses against transgene antigen products and the vector itself. We show that shielding Ad5 with different-sized PEGs generally reduces transduction and primary antibody responses by the intramuscular or intranasal route. In contrast, PEGylated vectors generally appear better at boosting antibody responses in Ad-immune animals. Displaying either glucose or galactose on PEG mediated increased transduction and antibody responses by the intranasal, but not the intramuscular, route. In naive animals, PEGylated vectors generated T cell responses that were equal to or better than those by unmodified Ad. Priming by PEGylated vectors generally enabled better subsequent T cell responses after boost. Priming and boosting by PEGylated vectors produced T cell responses after boost that were equal to or higher than those produced by unmodified vectors. These data indicate that PEGylation can enable more effective application of Ad5 and perhaps other Ad serotype vaccines during prime-boost vaccination.
Collapse
Affiliation(s)
- Eric A Weaver
- Division of Infectious Diseases, Department of Internal Medicine, and Translational Immunovirology and Biodefense Program, Mayo Clinic, Rochester, MN 55902, USA
| | | |
Collapse
|
47
|
Morizono K, Xie Y, Helguera G, Daniels TR, Lane TF, Penichet ML, Chen ISY. A versatile targeting system with lentiviral vectors bearing the biotin-adaptor peptide. J Gene Med 2009; 11:655-63. [PMID: 19455593 PMCID: PMC2834590 DOI: 10.1002/jgm.1345] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Targeted gene transduction in vivo is the ultimate preferred method for gene delivery. We previously developed targeting lentiviral vectors that specifically recognize cell surface molecules with conjugated antibodies and mediate targeted gene transduction both in vitro and in vivo. Although effective in some experimental settings, the conjugation of virus with antibodies is mediated by the interaction between protein A and the Fc region of antibodies, which is not as stable as covalent conjugation. We have now developed a more stable conjugation strategy utilizing the interaction between avidin and biotin. METHODS We inserted the biotin-adaptor-peptide, which was biotinylated by secretory biotin ligase at specific sites, into our targeting envelope proteins, enabling conjugation of the pseudotyped virus with avidin, streptavidin or neutravidin. RESULTS When conjugated with avidin-antibody fusion proteins or the complex of avidin and biotinylated targeting molecules, the vectors could mediate specific transduction to targeted cells recognized by the targeting molecules. When conjugated with streptavidin-coated magnetic beads, transduction by the vectors was targeted to the locations of magnets. CONCLUSIONS This targeting vector system can be used for broad applications of targeted gene transduction using biotinylated targeting molecules or targeting molecules fused with avidin.
Collapse
Affiliation(s)
- Kouki Morizono
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yiming Xie
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Gustavo Helguera
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tracy R. Daniels
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Timothy F. Lane
- Departments of Biological Chemistry and Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Manuel L. Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Departments of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Irvin S. Y. Chen
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Departments of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
48
|
(Strept)avidin-displaying lentiviruses as versatile tools for targeting and dual imaging of gene delivery. Gene Ther 2009; 16:894-904. [PMID: 19440224 DOI: 10.1038/gt.2009.47] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Lentiviruses have shown great promise for human gene therapy. However, no optimal strategies are yet available for noninvasive imaging of virus biodistribution and subsequent transduction in vivo. We have developed a dual-imaging strategy based on avidin-biotin system allowing easy exchange of the surface ligand on HIV-derived lentivirus envelope. This was achieved by displaying avidin or streptavidin fused to the transmembrane anchor of vesicular stomatitis virus G protein on gp64-pseudotyped envelopes. Avidin and streptavidin were efficiently incorporated on virus particles, which consequently showed binding to biotin in ELISA. These vectors, conjugated to biotinylated radionuclides and engineered to express a ferritin transgene, enabled for the first-time dual imaging of virus biodistribution and transduction pattern by single-photon emission computed tomography and magnetic resonance imaging after stereotactic injection into rat brain. In addition, vector retargeting to cancer cells overexpressing CD46, epidermal growth factor and transferrin receptors using biotinylated ligands and antibodies was demonstrated in vitro. In conclusion, we have generated novel lentivirus vectors for noninvasive imaging and targeting of lentivirus-mediated gene delivery. This study suggests that these novel vectors could be applicable for the treatment of central nervous system disorders and cancer.
Collapse
|
49
|
Sharma A, Tandon M, Bangari DS, Mittal SK. Adenoviral vector-based strategies for cancer therapy. CURRENT DRUG THERAPY 2009; 4:117-138. [PMID: 20160875 PMCID: PMC2771947 DOI: 10.2174/157488509788185123] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Definitive treatment of cancer has eluded scientists for decades. Current therapeutic modalities like surgery, chemotherapy, radiotherapy and receptor-targeted antibodies have varied degree of success and generally have moderate to severe side effects. Gene therapy is one of the novel and promising approaches for therapeutic intervention of cancer. Viral vectors in general and adenoviral (Ad) vectors in particular are efficient natural gene delivery systems and are one of the obvious choices for cancer gene therapy. Clinical and preclinical findings with a wide variety of approaches like tumor suppressor and suicide gene therapy, oncolysis, immunotherapy, anti-angiogenesis and RNA interference using Ad vectors have been quite promising, but there are still many hurdles to overcome. Shortcomings like increased immunogenicity, prevalence of preexisting anti-Ad immunity in human population and lack of specific targeting limit the clinical usefulness of Ad vectors. In recent years, extensive research efforts have been made to overcome these limitations through a variety of approaches including the use of conditionally-replicating Ad and specific targeting of tumor cells. In this review, we discuss the potential strengths and limitations of Ad vectors for cancer therapy.
Collapse
Affiliation(s)
| | | | - Dinesh S. Bangari
- Department of Comparative Pathobiology, and Bindley Bioscience Center, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, and Bindley Bioscience Center, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
50
|
Sharma A, Li X, Bangari DS, Mittal SK. Adenovirus receptors and their implications in gene delivery. Virus Res 2009; 143:184-94. [PMID: 19647886 PMCID: PMC2903974 DOI: 10.1016/j.virusres.2009.02.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 02/15/2009] [Indexed: 12/14/2022]
Abstract
Adenoviruses (Ads) have gained popularity as gene delivery vectors for therapeutic and prophylactic applications. Ad entry into host cells involves specific interactions between cell surface receptors and viral capsid proteins. Several cell surface molecules have been identified as receptors for Ad attachment and entry. Tissue tropism of Ad vectors is greatly influenced by their receptor usage. A variety of strategies have been investigated to modify Ad vector tropism by manipulating the receptor-interacting moieties. Many such strategies are aimed at targeting and/or detargeting of Ad vectors. In this review, we discuss the various cell surface molecules that are implicated as receptors for virus attachment and internalization. Special emphasis is given to Ad types that are utilized as gene delivery vectors. Various strategies to modify Ad tropism using the knowledge of Ad receptors are also discussed.
Collapse
Affiliation(s)
- Anurag Sharma
- Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | |
Collapse
|