1
|
Zhao F, Wang J, Zhang Y, Hu J, Li C, Liu S, Li R, Du R. In vivo Fate of Targeted Drug Delivery Carriers. Int J Nanomedicine 2024; 19:6895-6929. [PMID: 39005963 PMCID: PMC11246094 DOI: 10.2147/ijn.s465959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
This review aimed to systematically investigate the intracellular and subcellular fate of various types of targeting carriers. Upon entering the body via intravenous injection or other routes, a targeting carrier that can deliver therapeutic agents initiates their journey. If administered intravenously, the carrier initially faces challenges presented by the blood circulation before reaching specific tissues and interacting with cells within the tissue. At the subcellular level, the car2rier undergoes processes, such as drug release, degradation, and metabolism, through specific pathways. While studies on the fate of 13 types of carriers have been relatively conclusive, these studies are incomplete and lack a comprehensive analysis. Furthermore, there are still carriers whose fate remains unclear, underscoring the need for continuous research. This study highlights the importance of comprehending the in vivo and intracellular fate of targeting carriers and provides valuable insights into the operational mechanisms of different carriers within the body. By doing so, researchers can effectively select appropriate carriers and enhance the successful clinical translation of new formulations.
Collapse
Affiliation(s)
- Fan Zhao
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jitong Wang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yu Zhang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jinru Hu
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Chenyang Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Shuainan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Key Laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
- Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Ruofei Du
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
2
|
Barbieri E, Mollica GN, Moore BD, Sripada SA, Shastry S, Kilgore RE, Loudermilk CM, Whitacre ZH, Kilgour KM, Wuestenhagen E, Aldinger A, Graalfs H, Rammo O, Schulte MM, Johnson TF, Daniele MA, Menegatti S. Peptide ligands targeting the vesicular stomatitis virus G (VSV-G) protein for the affinity purification of lentivirus particles. Biotechnol Bioeng 2024; 121:618-639. [PMID: 37947118 DOI: 10.1002/bit.28594] [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: 08/23/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
The recent uptick in the approval of ex vivo cell therapies highlights the relevance of lentivirus (LV) as an enabling viral vector of modern medicine. As labile biologics, however, LVs pose critical challenges to industrial biomanufacturing. In particular, LV purification-currently reliant on filtration and anion-exchange or size-exclusion chromatography-suffers from long process times and low yield of transducing particles, which translate into high waiting time and cost to patients. Seeking to improve LV downstream processing, this study introduces peptides targeting the enveloped protein Vesicular stomatitis virus G (VSV-G) to serve as affinity ligands for the chromatographic purification of LV particles. An ensemble of candidate ligands was initially discovered by implementing a dual-fluorescence screening technology and a targeted in silico approach designed to identify sequences with high selectivity and tunable affinity. The selected peptides were conjugated on Poros resin and their LV binding-and-release performance was optimized by adjusting the flow rate, composition, and pH of the chromatographic buffers. Ligands GKEAAFAA and SRAFVGDADRD were selected for their high product yield (50%-60% of viral genomes; 40%-50% of HT1080 cell-transducing particles) upon elution in PIPES buffer with 0.65 M NaCl at pH 7.4. The peptide-based adsorbents also presented remarkable values of binding capacity (up to 3·109 TU per mL of resin, or 5·1011 vp per mL of resin, at the residence time of 1 min) and clearance of host cell proteins (up to a 220-fold reduction of HEK293 HCPs). Additionally, GKEAAFAA demonstrated high resistance to caustic cleaning-in-place (0.5 M NaOH, 30 min) with no observable loss in product yield and quality.
Collapse
Affiliation(s)
- Eduardo Barbieri
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Gina N Mollica
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Brandyn D Moore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Sobhana A Sripada
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Shriarjun Shastry
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, North Carolina, USA
| | - Ryan E Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Casee M Loudermilk
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Zachary H Whitacre
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Katie M Kilgour
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | | | | | | | | | | | - Thomas F Johnson
- Department of Biochemical Engineering, University College London, London, UK
| | - Michael A Daniele
- North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, Raleigh, North Carolina, USA
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina, USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, North Carolina, USA
- North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, Raleigh, North Carolina, USA
- LigaTrap Technologies LLC, Raleigh, North Carolina, USA
| |
Collapse
|
3
|
Liu H, Luo GF, Shang Z. Plant-derived nanovesicles as an emerging platform for cancer therapy. Acta Pharm Sin B 2024; 14:133-154. [PMID: 38239235 PMCID: PMC10792991 DOI: 10.1016/j.apsb.2023.08.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/14/2023] [Accepted: 08/26/2023] [Indexed: 01/22/2024] Open
Abstract
Plant-derived nanovesicles (PDNVs) derived from natural green products have emerged as an attractive nanoplatform in biomedical application. They are usually characterized by unique structural and biological functions, such as the bioactive lipids/proteins/nucleic acids as therapeutics and targeting groups, immune-modulation, and long-term circulation. With the rapid development of nanotechnology, materials, and synthetic chemistry, PDNVs can be engineered with multiple functions for efficient drug delivery and specific killing of diseased cells, which represent an innovative biomaterial with high biocompatibility for fighting against cancer. In this review, we provide an overview of the state-of-the-art studies concerning the development of PDNVs for cancer therapy. The original sources, methods for obtaining PDNVs, composition and structure are introduced systematically. With an emphasis on the featured application, the inherent anticancer properties of PDNVs as well as the strategies in constructing multifunctional PDNVs-based nanomaterials will be discussed in detail. Finally, some scientific issues and technical challenges of PDNVs as promising options in improving anticancer therapy will be discussed, which are expected to promote the further development of PDNVs in clinical translation.
Collapse
Affiliation(s)
- Hanzhe Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guo-Feng Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhengjun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
4
|
Nonclinical pharmacokinetics and biodistribution of VSV-GP using methods to decouple input drug disposition and viral replication. Mol Ther Methods Clin Dev 2022; 28:190-207. [PMID: 36700123 PMCID: PMC9843450 DOI: 10.1016/j.omtm.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Viral replication places oncolytic viruses (OVs) in a unique niche in the field of drug pharmacokinetics (PK) as their self-amplification obscures exposure-response relationships. Moreover, standard bioanalytical techniques are unable to distinguish the input from replicated drug products. Here, we combine two novel approaches to characterize PK and biodistribution (BD) after systemic administration of vesicular stomatitis virus pseudotyped with lymphocytic choriomeningitis virus glycoprotein (VSV-GP) in healthy mice. First: to decouple input drug PK/BD versus replication PK/BD, we developed and fully characterized a replication-incompetent tool virus that retained all other critical attributes of the drug. We used this approach to quantify replication in blood and tissues and to determine its impact on PK and BD. Second: to discriminate the genomic and antigenomic viral RNA strands contributing to replication dynamics in tissues, we developed an in situ hybridization method using strand-specific probes and assessed their spatiotemporal distribution in tissues. This latter approach demonstrated that distribution, transcription, and replication localized to tissue-resident macrophages, indicating their role in PK and BD. Ultimately, our study results in a refined PK/BD profile for a replicating OV, new proposed PK parameters, and deeper understanding of OV PK/BD using unique approaches that could be applied to other replicating vectors.
Collapse
|
5
|
Arsenijevic Y, Berger A, Udry F, Kostic C. Lentiviral Vectors for Ocular Gene Therapy. Pharmaceutics 2022; 14:pharmaceutics14081605. [PMID: 36015231 PMCID: PMC9414879 DOI: 10.3390/pharmaceutics14081605] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 12/10/2022] Open
Abstract
This review offers the basics of lentiviral vector technologies, their advantages and pitfalls, and an overview of their use in the field of ophthalmology. First, the description of the global challenges encountered to develop safe and efficient lentiviral recombinant vectors for clinical application is provided. The risks and the measures taken to minimize secondary effects as well as new strategies using these vectors are also discussed. This review then focuses on lentiviral vectors specifically designed for ocular therapy and goes over preclinical and clinical studies describing their safety and efficacy. A therapeutic approach using lentiviral vector-mediated gene therapy is currently being developed for many ocular diseases, e.g., aged-related macular degeneration, retinopathy of prematurity, inherited retinal dystrophies (Leber congenital amaurosis type 2, Stargardt disease, Usher syndrome), glaucoma, and corneal fibrosis or engraftment rejection. In summary, this review shows how lentiviral vectors offer an interesting alternative for gene therapy in all ocular compartments.
Collapse
Affiliation(s)
- Yvan Arsenijevic
- Unit Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, 1004 Lausanne, Switzerland;
- Correspondence: (Y.A.); (C.K.)
| | - Adeline Berger
- Group Epigenetics of ocular diseases, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, 1004 Lausanne, Switzerland;
| | - Florian Udry
- Unit Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, 1004 Lausanne, Switzerland;
| | - Corinne Kostic
- Group for Retinal Disorder Research, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, 1004 Lausanne, Switzerland
- Correspondence: (Y.A.); (C.K.)
| |
Collapse
|
6
|
Deng L, Liang P, Cui H. Pseudotyped lentiviral vectors: Ready for translation into targeted cancer gene therapy? Genes Dis 2022. [PMID: 37492721 PMCID: PMC10363566 DOI: 10.1016/j.gendis.2022.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Gene therapy holds great promise for curing cancer by editing the deleterious genes of tumor cells, but the lack of vector systems for efficient delivery of genetic material into specific tumor sites in vivo has limited its full therapeutic potential in cancer gene therapy. Over the past two decades, increasing studies have shown that lentiviral vectors (LVs) modified with different glycoproteins from a donating virus, a process referred to as pseudotyping, have altered tropism and display cell-type specificity in transduction, leading to selective tumor cell killing. This feature of LVs together with their ability to enable high efficient gene delivery in dividing and non-dividing mammalian cells in vivo make them to be attractive tools in future cancer gene therapy. This review is intended to summarize the status quo of some typical pseudotypings of LVs and their applications in basic anti-cancer studies across many malignancies. The opportunities of translating pseudotyped LVs into clinic use in cancer therapy have also been discussed.
Collapse
|
7
|
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
|
8
|
Tagging and Capturing of Lentiviral Vectors Using Short RNAs. Int J Mol Sci 2021; 22:ijms221910263. [PMID: 34638603 PMCID: PMC8508951 DOI: 10.3390/ijms221910263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/19/2021] [Indexed: 11/22/2022] Open
Abstract
Lentiviral (LV) vectors have emerged as powerful tools for transgene delivery ex vivo but in vivo gene therapy applications involving LV vectors have faced a number of challenges, including the low efficiency of transgene delivery, a lack of tissue specificity, immunogenicity to both the product encoded by the transgene and the vector, and the inactivation of the vector by the human complement cascade. To mitigate these issues, several engineering approaches, involving the covalent modification of vector particles or the incorporation of specific protein domains into the vector’s envelope, have been tested. Short synthetic oligonucleotides, including aptamers bound to the surface of LV vectors, may provide a novel means with which to retarget LV vectors to specific cells and to shield these vectors from neutralization by sera. The purpose of this study was to develop strategies to tether nucleic acid sequences, including short RNA sequences, to LV vector particles in a specific and tight fashion. To bind short RNA sequences to LV vector particles, a bacteriophage lambda N protein-derived RNA binding domain (λN), fused to the measles virus hemagglutinin protein, was used. The λN protein bound RNA sequences bearing a boxB RNA hairpin. To test this approach, we used an RNA aptamer specific to the human epidermal growth factor receptor (EGFR), which was bound to LV vector particles via an RNA scaffold containing a boxB RNA motif. The results obtained confirmed that the EGFR-specific RNA aptamer bound to cells expressing EGFR and that the boxB containing the RNA scaffold was bound specifically to the λN RNA binding domain attached to the vector. These results show that LV vectors can be equipped with nucleic acid sequences to develop improved LV vectors for in vivo applications.
Collapse
|
9
|
Bacteriophage Technology and Modern Medicine. Antibiotics (Basel) 2021; 10:antibiotics10080999. [PMID: 34439049 PMCID: PMC8388951 DOI: 10.3390/antibiotics10080999] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/26/2022] Open
Abstract
The bacteriophage (or phage for short) has been used as an antibacterial agent for over a century but was abandoned in most countries after the discovery and broad use of antibiotics. The worldwide emergence and high prevalence of antimicrobial-resistant (AMR) bacteria have led to a revival of interest in the long-forgotten antibacterial therapy with phages (phage therapy) as an alternative approach to combatting AMR bacteria. The rapid progress recently made in molecular biology and genetic engineering has accelerated the generation of phage-related products with superior therapeutic potentials against bacterial infection. Nowadays, phage-based technology has been developed for many purposes, including those beyond the framework of antibacterial treatment, such as to suppress viruses by phages, gene therapy, vaccine development, etc. Here, we highlighted the current progress in phage engineering technology and its application in modern medicine.
Collapse
|
10
|
Lentiviral Vectors Delivered with Biomaterials as Therapeutics for Spinal Cord Injury. Cells 2021; 10:cells10082102. [PMID: 34440872 PMCID: PMC8394044 DOI: 10.3390/cells10082102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating trauma that can cause permanent disability, life-long chronic issues for sufferers and is a big socioeconomic burden. Regenerative medicine aims to overcome injury caused deficits and restore function after SCI through gene therapy and tissue engineering approaches. SCI has a multifaceted pathophysiology. Due to this, producing therapies that target multiple different cellular and molecular mechanisms might prove to be a superior approach in attempts at regeneration. Both biomaterials and nucleic acid delivery via lentiviral vectors (LVs) have proven to promote repair and restoration of function post SCI in animal models. Studies indicate that a combination of biomaterials and LVs is more effective than either approach alone. This review presents studies supporting the use of LVs and LVs delivered with biomaterials in therapies for SCI and summarises methods to combine LVs with biomaterials for SCI treatment. By summarising this knowledge this review aims to demonstrate how LV delivery with biomaterials can augment/compliment both LV and biomaterial therapeutic effects in SCI.
Collapse
|
11
|
Comisel RM, Kara B, Fiesser FH, Farid SS. Lentiviral vector bioprocess economics for cell and gene therapy commercialization. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2020.107868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
12
|
Yousefi H, Mahmud A, Chang D, Das J, Gomis S, Chen JB, Wang H, Been T, Yip L, Coomes E, Li Z, Mubareka S, McGeer A, Christie N, Gray-Owen S, Cochrane A, Rini JM, Sargent EH, Kelley SO. Detection of SARS-CoV-2 Viral Particles Using Direct, Reagent-Free Electrochemical Sensing. J Am Chem Soc 2021; 143:1722-1727. [PMID: 33481575 PMCID: PMC7857138 DOI: 10.1021/jacs.0c10810] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 02/07/2023]
Abstract
The development of new methods for direct viral detection using streamlined and ideally reagent-free assays is a timely and important, but challenging, problem. The challenge of combatting the COVID-19 pandemic has been exacerbated by the lack of rapid and effective methods to identify viral pathogens like SARS-CoV-2 on-demand. Existing gold standard nucleic acid-based approaches require enzymatic amplification to achieve clinically relevant levels of sensitivity and are not typically used outside of a laboratory setting. Here, we report reagent-free viral sensing that directly reads out the presence of viral particles in 5 minutes using only a sensor-modified electrode chip. The approach relies on a class of electrode-tethered sensors bearing an analyte-binding antibody displayed on a negatively charged DNA linker that also features a tethered redox probe. When a positive potential is applied, the sensor is transported to the electrode surface. Using chronoamperometry, the presence of viral particles and proteins can be detected as these species increase the hydrodynamic drag on the sensor. This report is the first virus-detecting assay that uses the kinetic response of a probe/virus complex to analyze the complexation state of the antibody. We demonstrate the performance of this sensing approach as a means to detect, within 5 min, the presence of the SARS-CoV-2 virus and its associated spike protein in test samples and in unprocessed patient saliva.
Collapse
Affiliation(s)
- Hanie Yousefi
- Leslie
Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Alam Mahmud
- The
Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S
3G4, Canada
| | - Dingran Chang
- Leslie
Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Jagotamoy Das
- Leslie
Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Surath Gomis
- The
Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S
3G4, Canada
| | - Jenise B. Chen
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Hansen Wang
- Leslie
Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Terek Been
- Department
of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lily Yip
- Sunnybrook
Research Institute, Toronto, ON M4N 3N5, Canada
| | - Eric Coomes
- Division
of Infectious Disease, Department of Medicine, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Zhijie Li
- Department
of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Allison McGeer
- Department
of Microbiology, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Natasha Christie
- Combined
Containment Level 3 Unit, University of
Toronto, Toronto, ON M5S 1A8, Canada
| | - Scott Gray-Owen
- Department
of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Combined
Containment Level 3 Unit, University of
Toronto, Toronto, ON M5S 1A8, Canada
| | - Alan Cochrane
- Department
of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - James M. Rini
- Department
of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department
of Biochemistry, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Edward H. Sargent
- The
Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S
3G4, Canada
| | - Shana O. Kelley
- Leslie
Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
- Department
of Biochemistry, University of Toronto, Toronto, ON M5G 1M1, Canada
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
13
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
14
|
Gu H, Shi S, Xiao F, Huang Z, Xu J, Chen G, Zhou K, Lu L, Yin X. MiR-1-3p regulates the differentiation of mesenchymal stem cells to prevent osteoporosis by targeting secreted frizzled-related protein 1. Bone 2020; 137:115444. [PMID: 32447074 DOI: 10.1016/j.bone.2020.115444] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/11/2023]
Abstract
Osteoporosis (OP) is a systemic skeletal disorder with the characteristics of bone mass reduction and microarchitecture deterioration, resulting in bone fragility and increased fracture risk. A reduction in the osteoblast-differentiation of bone marrow mesenchymal stem cells (BMSCs) is considered as a basic pathogenesis of osteoporosis. miRNAs play a substantial role in the development and differentiation of BMSCs. In the present study, we found that miR-1-3p was significantly downregulated in the bones of Chinese osteoporotic patients (n = 29). Secreted frizzled-related protein 1 (SFRP1) was predicted as a target gene of miR-1-3p via the TargetScan and PicTar softwares and validated by dual-luciferase reporter assays. The findings revealed that the expression of SFRP1 was inversely correlated with miR-1-3p in osteoporotic patients. We induced mouse MSCs (mMSCs) to osteogenesis or adipogenesis and found that miR-1-3p was upregulated during osteogenesis but downregulated during adipogenesis. The overexpression of miR-1-3p stimulated osteogenesis and inhibited adipogenesis of mMSCs. In addition, ovariectomized (OVX) mice were tested and the function of miR-1-3p in vivo was explored. Immunohistochemistry and histomorphometric assays showed that in vivo inhibition of miR-1-3p increased the expression level of SFRP1 and reduced bone formation and bone mass. Furthermore, tartrate-resistant acid phosphatase (TRAP) staining indicated that the in vivo suppression of miR-1-3p promoted osteoclast activity, suggesting that miR-1-3p may influence bone mass by regulating bone resorption. It can be concluded that miR-1-3p plays a pivotal role in the pathogenesis of osteoporosis via targeting SFRP1 and may be a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Si Shi
- Department of Biochemistry and Molecular Biology, School of medicine, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Fangzhu Xiao
- Department of Orthopedics, The Fifth Hospital of Xiamen, 101 Min 'an Road, Maxiang Town, Xiang 'an District, Xiamen, Fujian Province, 361101, PR China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Lixia Lu
- Department of Biochemistry and Molecular Biology, School of medicine, Tongji University, 1239 Siping Road, Shanghai 200092, PR China.
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China.
| |
Collapse
|
15
|
Carbonaro-Sarracino DA, Tarantal AF, Lee CCI, Kaufman ML, Wandro S, Jin X, Martinez M, Clark DN, Chun K, Koziol C, Hardee CL, Wang X, Kohn DB. Dosing and Re-Administration of Lentiviral Vector for In Vivo Gene Therapy in Rhesus Monkeys and ADA-Deficient Mice. Mol Ther Methods Clin Dev 2020; 16:78-93. [PMID: 31871959 PMCID: PMC6909201 DOI: 10.1016/j.omtm.2019.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022]
Abstract
Adenosine deaminase (ADA)-deficient mice and healthy rhesus monkeys were studied to determine the impact of age at treatment, vector dosage, dosing schedule, repeat administration, biodistribution, and immunogenicity after systemic delivery of lentiviral vectors (LVs). In Ada -/- mice, neonatal treatment resulted in broad vector marking across all tissues analyzed, whereas adult treatment resulted in marking restricted to the liver, spleen, and bone marrow. Intravenous administration to infant rhesus monkeys also resulted in dose-dependent marking in the liver, spleen, and bone marrow. Using an ELISA to monitor anti-vector antibody development, Ada -/- neonatal mice did not produce an antibody response, whereas Ada -/- adult mice produced a strong antibody response to vector administration. In mice and monkeys with repeat administration of LV, a strong anti-vector antibody response was shown in response to the second LV administration, which resulted in LV inactivation. Three separate doses administered to immune competent mice resulted in acute toxicity. Pegylation of the vesicular stomatitis virus G protein (VSV-G)-enveloped LVs showed a less robust anti-vector response but did not prevent the inactivation of the second LV administration. These studies identify important factors to consider related to age and timing of administration when implementing systemic delivery of LVs as a potential therapeutic agent.
Collapse
Affiliation(s)
- Denise A. Carbonaro-Sarracino
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alice F. Tarantal
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, Davis, CA 95616, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - C. Chang I. Lee
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, Davis, CA 95616, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Michael L. Kaufman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen Wandro
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiangyang Jin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michele Martinez
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, Davis, CA 95616, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Danielle N. Clark
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Krista Chun
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Colin Koziol
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cinnamon L. Hardee
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Donald B. Kohn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- The Eli & Edythe Broad Center for Stem Cells and Regenerative Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Abstract
The optimal clinical exploitation of viruses as gene therapy or oncolytic vectors will require them to be administered intravenously. Strategies must therefore be deployed to enable viruses to survive the harsh neutralizing environment of the bloodstream and achieve deposition within and throughout target tissues or tumor deposits. This chapter describes the genetic and chemical engineering approaches that are being developed to overcome these challenges.
Collapse
Affiliation(s)
- Claudia A P Hill
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Luca Bau
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
| |
Collapse
|
17
|
Pipperger L, Koske I, Wild N, Müllauer B, Krenn D, Stoiber H, Wollmann G, Kimpel J, von Laer D, Bánki Z. Xenoantigen-Dependent Complement-Mediated Neutralization of Lymphocytic Choriomeningitis Virus Glycoprotein-Pseudotyped Vesicular Stomatitis Virus in Human Serum. J Virol 2019; 93:e00567-19. [PMID: 31243134 PMCID: PMC6714799 DOI: 10.1128/jvi.00567-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023] Open
Abstract
Neutralization by antibodies and complement limits the effective dose and thus the therapeutic efficacy of oncolytic viruses after systemic application. We and others previously showed that pseudotyping of oncolytic rhabdoviruses such as maraba virus and vesicular stomatitis virus (VSV) with the lymphocytic choriomeningitis virus glycoprotein (LCMV-GP) results in only a weak induction of neutralizing antibodies. Moreover, LCMV-GP-pseudotyped VSV (VSV-GP) was significantly more stable in normal human serum (NHS) than VSV. Here, we demonstrate that depending on the cell line used for virus production, VSV-GP showed different complement sensitivities in nonimmune NHS. The NHS-mediated titer reduction of VSV-GP was dependent on activation of the classical complement pathway, mainly by natural IgM antibodies against xenoantigens such as galactose-α-(1,3)-galactose (α-Gal) or N-glycolylneuraminic acid (Neu5Gc) expressed on nonhuman production cell lines. VSV-GP produced on human cell lines was stable in NHS. However, VSV-GP generated in transduced human cells expressing α-Gal became sensitive to NHS. Furthermore, GP-specific antibodies induced complement-mediated neutralization of VSV-GP independently of the producer cell line, suggesting that complement regulatory proteins potentially acquired by the virus during the budding process are not sufficient to rescue the virus from antibody-dependent complement-mediated lysis. Thus, our study points to the importance of a careful selection of cell lines for viral vector production for clinical use.IMPORTANCE Systemic application aims to deliver oncolytic viruses to tumors as well as to metastatic lesions. However, we found that xenoantigens incorporated onto the viral surface from nonhuman production cell lines are recognized by natural antibodies in human serum and that the virus is thereby inactivated by complement lysis. Hence, to maximize the effective dose, careful selection of cell lines for virus production is crucial.
Collapse
Affiliation(s)
- Lisa Pipperger
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Iris Koske
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nicole Wild
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Brigitte Müllauer
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Krenn
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heribert Stoiber
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Guido Wollmann
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothee von Laer
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Bánki
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
18
|
Balakrishnan B, David E. Biopolymers augment viral vectors based gene delivery. J Biosci 2019; 44:84. [PMID: 31502562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The success of viral vectors mediated gene therapy is still hampered by immunogenicity and insufficient transgene expression. Alternatively, non-viral vectors mediated gene delivery has the advantage of low immunogenicity despite showing low transgene expression. By carefully considering the advantages of each approach, hybrid vectors are currently being developed by modifying the viral vectors using non-viral biopolymers. This review provides an overview of the hybrid vectors currently being developed.
Collapse
Affiliation(s)
- Balaji Balakrishnan
- Department of Haematology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | | |
Collapse
|
19
|
|
20
|
Munis AM, Mattiuzzo G, Bentley EM, Collins MK, Eyles JE, Takeuchi Y. Use of Heterologous Vesiculovirus G Proteins Circumvents the Humoral Anti-envelope Immunity in Lentivector-Based In Vivo Gene Delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:126-137. [PMID: 31254925 PMCID: PMC6599914 DOI: 10.1016/j.omtn.2019.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 01/12/2023]
Abstract
Vesicular stomatitis virus Indiana strain glycoprotein (VSVind.G) mediates broad tissue tropism and efficient cellular uptake. Lentiviral vectors (LVs) are particularly promising, as they can efficiently transduce non-dividing cells and facilitate stable genomic transgene integration; therefore, LVs have an enormous untapped potential for gene therapy applications, but the development of humoral and cell-mediated anti-vector responses may restrict their efficacy. We hypothesized that G proteins from different members of the vesiculovirus genus might allow the generation of a panel of serotypically distinct LV pseudotypes with potential for repeated in vivo administration. We found that mice hyperimmunized with VSVind.G were not transduced to any significant degree following intravenous injection of LVs with VSVind.G envelopes, consistent with the thesis that multiple LV administrations would likely be blunted by an adaptive immune response. Excitingly, bioluminescence imaging studies demonstrated that the VSVind-neutralizing response could be evaded by LV pseudotyped with Piry and, to a lesser extent, Cocal virus glycoproteins. Heterologous dosing regimens using viral vectors and oncolytic viruses with Piry and Cocal envelopes could represent a novel strategy to achieve repeated vector-based interventions, unfettered by pre-existing anti-envelope antibodies.
Collapse
Affiliation(s)
- Altar M Munis
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK; Division of Infection and Immunity, University College London, London WC1E 6BT, UK.
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Emma M Bentley
- Division of Virology, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Mary K Collins
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK; Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan
| | - James E Eyles
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Yasuhiro Takeuchi
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK; Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| |
Collapse
|
21
|
Madrigal JL, Shams S, Stilhano RS, Silva EA. Characterizing the encapsulation and release of lentivectors and adeno-associated vectors from degradable alginate hydrogels. Biomater Sci 2019; 7:645-656. [PMID: 30534722 DOI: 10.1039/c8bm01218k] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gene therapy using viral vectors has been licensed for clinical use both in the European Union and the United States. Lentivectors (LV) and adeno-associated vectors (AAV) are two promising and FDA approved gene-therapy viral vectors. Many future applications of these vectors will benefit from targeting specific regions of interest within the body. Therefore, building on the early success of these vectors may depend on finding effective delivery systems to localize therapeutic administration. Degradable alginate hydrogels have been tested as appealing delivery vehicles for the controlled delivery of vector payloads. In this study, we compare the ability of two different degradable alginate hydrogel formulations to efficiently deliver LV and AAV. We propose that release rates of viral vectors are dependent on the physical properties of both the hydrogels and vectors. Here, we demonstrate that the initial strength and degradation rate of alginate hydrogels provides levers of control for tuning LV release but do not provide control in the release of AAV. While both alginate formulations used showed sustained release of both LV and AAV, LV release was shown to be dependent on alginate hydrogel degradation, while AAV release was largely governed by diffusive mechanisms. Altogether, this study demonstrates alginate's use as a possible delivery platform for LV and, for the first time, AAV - highlighting the potential of injectable degradable alginate hydrogels to be used as a versatile delivery tool in gene therapy applications.
Collapse
Affiliation(s)
- Justin L Madrigal
- Department of Biomedical Engineering, University of California, Davis, CA, USA.
| | | | | | | |
Collapse
|
22
|
Abstract
Polymeric matrices inherently protect viral vectors from pre-existing immune conditions, limit dissemination to off-target sites, and can sustain vector release. Advancing methodologies in development of particulate based vehicles have led to improved encapsulation of viral vectors. Polymeric delivery systems have contributed to increasing cellular transduction, responsive release mechanisms, cellular infiltration, and cellular signaling. Synthetic polymers are easily customizable, and are capable of balancing matrix retention with cellular infiltration. Natural polymers contain inherent biorecognizable motifs adding therapeutic efficacy to the incorporated viral vector. Recombinant polymers use highly conserved motifs to carefully engineer matrices, allowing for precise design including elements of vector retention and responsive release mechanisms. Composite polymer systems provide opportunities to create matrices with unique properties. Carefully designed matrices can control spatiotemporal release patterns that synergize with approaches in regenerative medicine and antitumor therapies.
Collapse
Affiliation(s)
- Douglas Steinhauff
- Utah Center for Nanomedicine , Nano Institute of Utah , 36 South Wasatch Drive , Salt Lake City , Utah 84112 , United States
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine , Nano Institute of Utah , 36 South Wasatch Drive , Salt Lake City , Utah 84112 , United States
| |
Collapse
|
23
|
Park J, Inwood S, Kruthiventi S, Jenkins J, Shiloach J, Betenbaugh M. Progressing from transient to stable packaging cell lines for continuous production of lentiviral and gammaretroviral vectors. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Characterization of Antibody Interactions with the G Protein of Vesicular Stomatitis Virus Indiana Strain and Other Vesiculovirus G Proteins. J Virol 2018; 92:JVI.00900-18. [PMID: 30232190 PMCID: PMC6232470 DOI: 10.1128/jvi.00900-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/06/2018] [Indexed: 01/20/2023] Open
Abstract
VSVind.G is currently regarded as the gold-standard envelope glycoprotein to pseudotype lentiviral vectors. However, recently other G proteins derived from vesiculoviruses have been proposed as alternative envelopes. Here, we investigated two commercially available anti-VSVind.G monoclonal antibodies for their ability to cross-react with other vesiculovirus G proteins, identified the epitopes they recognize, and explored their neutralization activity. We have identified 8G5F11, for the first time, as a cross-neutralizing antibody against several vesiculovirus G proteins. Furthermore, we elucidated the two different neutralization mechanisms employed by these two monoclonal antibodies. Understanding how cross-neutralizing antibodies interact with other G proteins may be of interest in the context of host-pathogen interaction and coevolution, as well as providing the opportunity to modify the G proteins and improve G protein-containing medicinal products and vaccine vectors. Vesicular stomatitis virus Indiana strain G protein (VSVind.G) is the most commonly used envelope glycoprotein to pseudotype lentiviral vectors (LV) for experimental and clinical applications. Recently, G proteins derived from other vesiculoviruses (VesG), for example, Cocal virus, have been proposed as alternative LV envelopes with possible advantages over VSVind.G. Well-characterized antibodies that recognize VesG will be useful for vesiculovirus research, development of G protein-containing advanced therapy medicinal products (ATMPs), and deployment of VSVind-based vaccine vectors. Here, we show that one commercially available monoclonal antibody, 8G5F11, binds to and neutralizes G proteins from three strains of VSV, as well as Cocal and Maraba viruses, whereas the other commercially available monoclonal anti-VSVind.G antibody, IE9F9, binds to and neutralizes only VSVind.G. Using a combination of G protein chimeras and site-directed mutations, we mapped the binding epitopes of IE9F9 and 8G5F11 on VSVind.G. IE9F9 binds close to the receptor binding site and competes with soluble low-density lipoprotein receptor (LDLR) for binding to VSVind.G, explaining its mechanism of neutralization. In contrast, 8G5F11 binds close to a region known to undergo conformational changes when the G protein moves to its postfusion structure, and we propose that 8G5F11 cross-neutralizes VesGs by inhibiting this. IMPORTANCE VSVind.G is currently regarded as the gold-standard envelope glycoprotein to pseudotype lentiviral vectors. However, recently other G proteins derived from vesiculoviruses have been proposed as alternative envelopes. Here, we investigated two commercially available anti-VSVind.G monoclonal antibodies for their ability to cross-react with other vesiculovirus G proteins, identified the epitopes they recognize, and explored their neutralization activity. We have identified 8G5F11, for the first time, as a cross-neutralizing antibody against several vesiculovirus G proteins. Furthermore, we elucidated the two different neutralization mechanisms employed by these two monoclonal antibodies. Understanding how cross-neutralizing antibodies interact with other G proteins may be of interest in the context of host-pathogen interaction and coevolution, as well as providing the opportunity to modify the G proteins and improve G protein-containing medicinal products and vaccine vectors.
Collapse
|
25
|
Tijani M, Munis AM, Perry C, Sanber K, Ferraresso M, Mukhopadhyay T, Themis M, Nisoli I, Mattiuzzo G, Collins MK, Takeuchi Y. Lentivector Producer Cell Lines with Stably Expressed Vesiculovirus Envelopes. Mol Ther Methods Clin Dev 2018; 10:303-312. [PMID: 30182034 PMCID: PMC6118154 DOI: 10.1016/j.omtm.2018.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/30/2018] [Indexed: 01/19/2023]
Abstract
Retroviral and lentiviral vectors often use the envelope G protein from the vesicular stomatitis virus Indiana strain (VSVind.G). However, lentivector producer cell lines that stably express VSVind.G have not been reported, presumably because of its cytotoxicity, preventing simple scale-up of vector production. Interestingly, we showed that VSVind.G and other vesiculovirus G from the VSV New Jersey strain (VSVnj), Cocal virus (COCV), and Piry virus (PIRYV) could be constitutively expressed and supported lentivector production for up to 10 weeks. All G-enveloped particles were robust, allowing concentration and freeze-thawing. COCV.G and PIRYV.G were resistant to complement inactivation, and, using chimeras between VSVind.G and COCV.G, the determinant for complement inactivation of VSVind.G was mapped to amino acid residues 136-370. Clonal packaging cell lines using COCV.G could be generated; however, during attempts to establish LV producer cells, vector superinfection was observed following the introduction of a lentivector genome. This could be prevented by culturing the cells with the antiviral drug nevirapine. As an alternative countermeasure, we demonstrated that functional lentivectors could be reconstituted by admixing supernatant from stable cells producing unenveloped virus with supernatant containing envelopes harvested from cells stably expressing VSVind.G, COCV.G, or PIRYV.G.
Collapse
Affiliation(s)
- Maha Tijani
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Altar M. Munis
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Christopher Perry
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
- Department of Biochemical Engineering, University College London, London WC1H 0AH, UK
| | - Khaled Sanber
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Marta Ferraresso
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Tarit Mukhopadhyay
- Department of Biochemical Engineering, University College London, London WC1H 0AH, UK
| | - Michael Themis
- Department of Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Ilaria Nisoli
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Mary K. Collins
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
- Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan
| | - Yasuhiro Takeuchi
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| |
Collapse
|
26
|
Sheikh IS, Keefe KM, Sterling NA, Junker IP, Eneanya CI, Liu Y, Tang XQ, Smith GM. Retrogradely Transportable Lentivirus Tracers for Mapping Spinal Cord Locomotor Circuits. Front Neural Circuits 2018; 12:60. [PMID: 30090059 PMCID: PMC6068242 DOI: 10.3389/fncir.2018.00060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022] Open
Abstract
Retrograde tracing is a key facet of neuroanatomical studies involving long distance projection neurons. Previous groups have utilized a variety of tools ranging from classical chemical tracers to newer methods employing viruses for gene delivery. Here, we highlight the usage of a lentivirus that permits highly efficient retrograde transport (HiRet) from synaptic terminals within the cervical and lumbar enlargements of the spinal cord. By injecting HiRet, we can clearly identify supraspinal and propriospinal circuits innervating motor neuron pools relating to forelimb and hindlimb function. We observed robust labeling of propriospinal neurons, including high fidelity details of dendritic arbors and axon terminals seldom seen with chemical tracers. In addition, we examine changes in interneuronal circuits occurring after a thoracic contusion, highlighting populations that potentially contribute to spontaneous behavioral recovery in this lesion model. Our study demonstrates that the HiRet lentivirus is a unique tool for examining neuronal circuitry within the brain and spinal cord.
Collapse
Affiliation(s)
- Imran S Sheikh
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Kathleen M Keefe
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Noelle A Sterling
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ian P Junker
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Chidubem I Eneanya
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yingpeng Liu
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiao-Qing Tang
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - George M Smith
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Center for Neural Rehabilitation and Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
27
|
Garaulet G, Lazcano JJ, Alarcón H, de Frutos S, Martínez-Torrecuadrada JL, Rodríguez A. Display of the Albumin-Binding Domain in the Envelope Improves Lentiviral Vector Bioavailability. Hum Gene Ther Methods 2018; 28:340-351. [PMID: 29160106 DOI: 10.1089/hgtb.2017.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vesicular stomatitis virus G glycoprotein (VSVg) is extensively used for retroviral and lentiviral vector (LV) pseudotyping. However, VSVg pseudotyped vectors are serum inactivated, blocking the in vivo gene delivery. Several strategies have been employed to prevent complement inactivation, including chemical and genetic envelope modifications. This study employed the streptococcal albumin-binding domain (ABD) to generate a construct to express ABD as a glycosylphosphatidylinositol-anchored protein. LV particles bearing ABD are able to bind bovine and human serum albumin in vitro. Neither the lentiviral vector production titer nor the in vitro transduction was affected by the ABD display. The study demonstrated that ABD-bearing LVs are protected from human complement inactivation. More importantly, intravenous administration demonstrated that the presence of ABD significantly reduces lentivector sequestration in liver and bone-marrow cells. Therefore, the use of ABD represents an improvement for in vivo gene therapy applications. The results strongly point to ABD display as a universal strategy to increase the in vivo efficacy of different viral vectors.
Collapse
Affiliation(s)
- Guillermo Garaulet
- 1 Department of Molecular Biology, Universidad Autónoma de Madrid , Madrid, E-28049 Spain
| | - Juan José Lazcano
- 2 Signaling and Inflammation Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC) , Madrid, E-28029 Spain
| | - Hernán Alarcón
- 1 Department of Molecular Biology, Universidad Autónoma de Madrid , Madrid, E-28049 Spain
| | - Sergio de Frutos
- 1 Department of Molecular Biology, Universidad Autónoma de Madrid , Madrid, E-28049 Spain
| | | | - Antonio Rodríguez
- 1 Department of Molecular Biology, Universidad Autónoma de Madrid , Madrid, E-28049 Spain
| |
Collapse
|
28
|
Milani M, Annoni A, Bartolaccini S, Biffi M, Russo F, Di Tomaso T, Raimondi A, Lengler J, Holmes MC, Scheiflinger F, Lombardo A, Cantore A, Naldini L. Genome editing for scalable production of alloantigen-free lentiviral vectors for in vivo gene therapy. EMBO Mol Med 2018; 9:1558-1573. [PMID: 28835507 PMCID: PMC5666310 DOI: 10.15252/emmm.201708148] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Lentiviral vectors (LV) are powerful and versatile vehicles for gene therapy. However, their complex biological composition challenges large-scale manufacturing and raises concerns for in vivo applications, because particle components and contaminants may trigger immune responses. Here, we show that producer cell-derived polymorphic class-I major histocompatibility complexes (MHC-I) are incorporated into the LV surface and trigger allogeneic T-cell responses. By disrupting the beta-2 microglobulin gene in producer cells, we obtained MHC-free LV with substantially reduced immunogenicity. We introduce this targeted editing into a novel stable LV packaging cell line, carrying single-copy inducible vector components, which can be reproducibly converted into high-yield LV producers upon site-specific integration of the LV genome of interest. These LV efficiently transfer genes into relevant targets and are more resistant to complement-mediated inactivation, because of reduced content of the vesicular stomatitis virus envelope glycoprotein G compared to vectors produced by transient transfection. Altogether, these advances support scalable manufacturing of alloantigen-free LV with higher purity and increased complement resistance that are better suited for in vivo gene therapy.
Collapse
Affiliation(s)
- Michela Milani
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita Salute San Raffaele University, Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Bartolaccini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mauro Biffi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Russo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tiziano Di Tomaso
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita Salute San Raffaele University, Milan, Italy
| | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy .,Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
29
|
Modulation of immune responses in lentiviral vector-mediated gene transfer. Cell Immunol 2018; 342:103802. [PMID: 29735164 PMCID: PMC6695505 DOI: 10.1016/j.cellimm.2018.04.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
Abstract
Lentiviral vectors (LV) are widely used vehicles for gene transfer and therapy in pre-clinical animal models and clinical trials with promising safety and efficacy results. However, host immune responses against vector- and/or transgene-derived antigens remain a major obstacle to the success and broad applicability of gene therapy. Here we review the innate and adaptive immunological barriers to successful gene therapy, both in the context of ex vivo and in vivo LV gene therapy, mostly concerning systemic LV delivery and discuss possible means to overcome them, including vector design and production and immune modulatory strategies.
Collapse
|
30
|
Taschuk R, Scruten E, Woodbury M, Cashman N, Potter A, Griebel P, Tikoo SK, Napper S. Induction of PrP Sc-specific systemic and mucosal immune responses in white-tailed deer with an oral vaccine for chronic wasting disease. Prion 2018; 11:368-380. [PMID: 28968152 PMCID: PMC5639826 DOI: 10.1080/19336896.2017.1367083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The ongoing epidemic of chronic wasting disease (CWD) within cervid populations indicates the need for novel approaches for disease management. A vaccine that either reduces susceptibility to infection or reduces shedding of prions by infected animals, or a combination of both, could be of benefit for disease control. The development of such a vaccine is challenged by the unique nature of prion diseases and the requirement for formulation and delivery in an oral format for application in wildlife settings. To address the unique nature of prions, our group targets epitopes, termed disease specific epitopes (DSEs), whose exposure for antibody binding depends on disease-associated misfolding of PrPC into PrPSc. Here, a DSE corresponding to the rigid loop (RL) region, which was immunogenic following parenteral vaccination, was translated into an oral vaccine. This vaccine consists of a replication-incompetent human adenovirus expressing a truncated rabies glycoprotein G recombinant fusion with the RL epitope (hAd5:tgG-RL). Oral immunization of white-tailed deer with hAd5:tgG-RL induced PrPSc-specific systemic and mucosal antibody responses with an encouraging safety profile in terms of no adverse health effects nor prolonged vector shedding. By building upon proven strategies of formulation for wildlife vaccines, these efforts generate a particular PrPSc-specific oral vaccine for CWD as well as providing a versatile platform, in terms of carrier protein and biological vector, for generation of other oral, peptide-based CWD vaccines.
Collapse
Affiliation(s)
- Ryan Taschuk
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,b School of Public Health, University of Saskatchewan , Saskatoon, Saskatchewan , Canada
| | - Erin Scruten
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| | - Murray Woodbury
- c Western College of Veterinary Medicine, University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| | - Neil Cashman
- d Department of Neurology , University of British Columbia , Vancouver , BC , Canada
| | - Andrew Potter
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| | - Philip Griebel
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,b School of Public Health, University of Saskatchewan , Saskatoon, Saskatchewan , Canada
| | - Suresh K Tikoo
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,b School of Public Health, University of Saskatchewan , Saskatoon, Saskatchewan , Canada
| | - Scott Napper
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,e Department of Biochemistry , University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| |
Collapse
|
31
|
Sharon D, Kamen A. Advancements in the design and scalable production of viral gene transfer vectors. Biotechnol Bioeng 2017; 115:25-40. [PMID: 28941274 DOI: 10.1002/bit.26461] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 01/22/2023]
Abstract
The last 10 years have seen a rapid expansion in the use of viral gene transfer vectors, with approved therapies and late stage clinical trials underway for the treatment of genetic disorders, and multiple forms of cancer, as well as prevention of infectious diseases through vaccination. With this increased interest and widespread adoption of viral vectors by clinicians and biopharmaceutical industries, there is an imperative to engineer safer and more efficacious vectors, and develop robust, scalable and cost-effective production platforms for industrialization. This review will focus on major innovations in viral vector design and production systems for three of the most widely used viral vectors: Adenovirus, Adeno-Associated Virus, and Lentivirus.
Collapse
Affiliation(s)
- David Sharon
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
32
|
Santiago DN, Heidbuechel JPW, Kandell WM, Walker R, Djeu J, Engeland CE, Abate-Daga D, Enderling H. Fighting Cancer with Mathematics and Viruses. Viruses 2017; 9:E239. [PMID: 28832539 PMCID: PMC5618005 DOI: 10.3390/v9090239] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
After decades of research, oncolytic virotherapy has recently advanced to clinical application, and currently a multitude of novel agents and combination treatments are being evaluated for cancer therapy. Oncolytic agents preferentially replicate in tumor cells, inducing tumor cell lysis and complex antitumor effects, such as innate and adaptive immune responses and the destruction of tumor vasculature. With the availability of different vector platforms and the potential of both genetic engineering and combination regimens to enhance particular aspects of safety and efficacy, the identification of optimal treatments for patient subpopulations or even individual patients becomes a top priority. Mathematical modeling can provide support in this arena by making use of experimental and clinical data to generate hypotheses about the mechanisms underlying complex biology and, ultimately, predict optimal treatment protocols. Increasingly complex models can be applied to account for therapeutically relevant parameters such as components of the immune system. In this review, we describe current developments in oncolytic virotherapy and mathematical modeling to discuss the benefit of integrating different modeling approaches into biological and clinical experimentation. Conclusively, we propose a mutual combination of these research fields to increase the value of the preclinical development and the therapeutic efficacy of the resulting treatments.
Collapse
Affiliation(s)
- Daniel N Santiago
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | | | - Wendy M Kandell
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Cancer Biology PhD Program, University of South Florida, Tampa, FL 33612, USA.
| | - Rachel Walker
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Julie Djeu
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Christine E Engeland
- German Cancer Research Center, Heidelberg University, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases Heidelberg, Department of Translational Oncology, Department of Medical Oncology, 69120 Heidelberg, Germany.
| | - Daniel Abate-Daga
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
33
|
Madrigal JL, Stilhano R, Silva EA. Biomaterial-Guided Gene Delivery for Musculoskeletal Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2017; 23:347-361. [PMID: 28166711 PMCID: PMC5749599 DOI: 10.1089/ten.teb.2016.0462] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/11/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy is a promising strategy for musculoskeletal tissue repair and regeneration where local and sustained expression of proteins and/or therapeutic nucleic acids can be achieved. However, the musculoskeletal tissues present unique engineering and biological challenges as recipients of genetic vectors. Targeting specific cell populations, regulating expression in vivo, and overcoming the harsh environment of damaged tissue accompany the general concerns of safety and efficacy common to all applications of gene therapy. In this review, we will first summarize these challenges and then discuss how biomaterial carriers for genetic vectors can address these issues. Second, we will review how limitations specific to given vectors further motivate the utility of biomaterial carriers. Finally, we will discuss how these concepts have been combined with tissue engineering strategies and approaches to improve the delivery of these vectors for musculoskeletal tissue regeneration.
Collapse
Affiliation(s)
- Justin L Madrigal
- Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - Roberta Stilhano
- Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - Eduardo A Silva
- Department of Biomedical Engineering, University of California , Davis, Davis, California
| |
Collapse
|
34
|
Liu X, Li YP, Zhong ZM, Tan HQ, Lin HP, Chen SJ, Fu YC, Xu WC, Wei CJ. Incorporation of Viral Glycoprotein VSV-G Improves the Delivery of DNA by Erythrocyte Ghost into Cells Refractory to Conventional Transfection. Appl Biochem Biotechnol 2017; 181:748-761. [PMID: 27665615 DOI: 10.1007/s12010-016-2246-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/12/2016] [Indexed: 02/05/2023]
Abstract
The objective of this study was to formulate a novel gene delivery system based on the erythrocyte ghost (EG) integrated with fusogenic viral glycoprotein vesicular stomatitis virus glycoprotein G (VSV-G). VSV-G proteins were harvested as condition medium of Ad293 cells carrying a VSV-G transgene and then incorporated into EG. Plasmid DNA was condensed by various transfection reagents. A luciferase expression construct (pGL3-control) and a DsRed expression cassette (pCMV-DsRed) were used to evaluate the delivery efficiency of DNA/EG/VSV-G complexes. VSV-G proteins could be incorporated into EG in static incubation under acidic conditions as evidenced by the Western blot analysis. Condensed plasmid DNA was bound mostly to the outer surface of EG, which could be detected by electromicroscopy and measured by electrophoresis. EG/VSV-G complexes stimulated the delivery of pGL3-control into Ad293 cells significantly with the luciferase activity increased about 4-fold as compared to that of the control. The delivery of pCMV-DsRed was also enhanced with the percentage of DsRed-positive Ad293 cells increased from 55 % to about 80 %. Moreover, the transfection efficiency in 3T3, HeLa, INS-1, and bone marrow stem cell (BMSC) cells increased about 2-3-fold. Finally, confocal microscopy analysis showed that incorporation of VSV-G significantly enhanced the endocytosis of EG into target cells. In the present study, a novel type of non-viral DNA delivery vehicle consisting of EG and fusogenic VSV-G proteins was formulated, which showed superior transfection efficiency even in cells resistant to classical transfection.
Collapse
Affiliation(s)
- Xin Liu
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Yun-Pan Li
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Zhen-Min Zhong
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Hui-Qi Tan
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Hao-Peng Lin
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Shao-Jun Chen
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Yu-Cai Fu
- Laboratory of Cell Senescence, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Wen-Can Xu
- Department of Endocrinology, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Chi-Ju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China.
| |
Collapse
|
35
|
Sosale NG, Ivanovska II, Tsai RK, Swift J, Hsu JW, Alvey CM, Zoltick PW, Discher DE. "Marker of Self" CD47 on lentiviral vectors decreases macrophage-mediated clearance and increases delivery to SIRPA-expressing lung carcinoma tumors. Mol Ther Methods Clin Dev 2016; 3:16080. [PMID: 28053997 PMCID: PMC5148596 DOI: 10.1038/mtm.2016.80] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 02/08/2023]
Abstract
Lentiviruses infect many cell types and are now widely used for gene delivery in vitro, but in vivo uptake of these foreign vectors by macrophages is a limitation. Lentivectors are produced here from packaging cells that overexpress "Marker of Self" CD47, which inhibits macrophage uptake of cells when prophagocytic factors are also displayed. Single particle analyses show "hCD47-Lenti" display properly oriented human-CD47 for interactions with the macrophage's inhibitory receptor SIRPA. Macrophages derived from human and NOD/SCID/Il2rg-/- (NSG) mice show a SIRPA-dependent decrease in transduction, i.e., transgene expression, by hCD47-Lenti compared to control Lenti. Consistent with known "Self" signaling pathways, macrophage transduction by control Lenti is decreased by drug inhibition of Myosin-II to the same levels as hCD47-Lenti. In contrast, human lung carcinoma cells express SIRPA and use it to enhance transduction by hCD47-Lenti- as illustrated by more efficient gene deletion using CRISPR/Cas9. Intravenous injection of hCD47-Lenti into NSG mice shows hCD47 prolongs circulation, unless a blocking anti-SIRPA is preinjected. In vivo transduction of spleen and liver macrophages also decreases for hCD47-Lenti while transduction of lung carcinoma xenografts increases. hCD47 could be useful when macrophage uptake is limiting on other viral vectors that are emerging in cancer treatments (e.g., Measles glycoprotein-pseudotyped lentivectors) and also in targeting various SIRPA-expressing tumors such as glioblastomas.
Collapse
Affiliation(s)
- Nisha G Sosale
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Irena I Ivanovska
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Richard K Tsai
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joe Swift
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jake W Hsu
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cory M Alvey
- Pharmacological Sciences Graduate Group, University of Pennsylvania, Pennsylvania, USA
| | - Philip W Zoltick
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dennis E Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Pharmacological Sciences Graduate Group, University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|
36
|
Stilhano RS, Madrigal JL, Wong K, Williams PA, Martin PK, Yamaguchi FS, Samoto VY, Han SW, Silva EA. Injectable alginate hydrogel for enhanced spatiotemporal control of lentivector delivery in murine skeletal muscle. J Control Release 2016; 237:42-9. [DOI: 10.1016/j.jconrel.2016.06.047] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 12/17/2022]
|
37
|
Yang B, He F, Dai C, Tan R, Ma D, Wang Z, Zhang B, Feng J, Wei L, Zhu H, Chen Z. BATF inhibition prevent acute allograft rejection after cardiac transplantation. Am J Transl Res 2016; 8:3603-3613. [PMID: 27648151 PMCID: PMC5009413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 04/28/2016] [Indexed: 06/06/2023]
Abstract
Acute allograft rejection is a serious and life-threatening complication of organ transplantation. Th17 cells induced inflammation has been described to play an important role in allograft rejection. Since there is a plenty of evidence indicating that transcriptional factor BATF regulates the differentiation of Th17 and follicular T helper cells both in vitro and in vivo, we investigated whether is BATF involved in acute rejection and allograft survival by injecting lentivirus containing BATF shRNA through tail vein before the cardiac transplantation operation. We found that the allograft survival time of the mice treated with BATF shRNA was significantly prolonged compared with that of negative shRNA treated group and the control group. Further pathological analysis revealed that the BATF shRNA treatment group had significantly lower rejection degree than the negative shRNA group, while there was no significant difference between the negative shRNA group and the control group. Furthermore, flow cytometry analysis and quantitative polymerase chain reaction and enzyme-linked immuno sorbent assay were used to determine the proportion of T helper cells, the expression of specific transcription factor and the inflammatory cytokines respectively. Data showed that BATF regulated Th17 and Treg responses during allograft rejection. And BATF inhibition led to reduction of the expression level of Rorγ-t and enhancement of the Foxp-3. In addition, cytokines IL-17A and IL-4 were found decreased. This may indicate BATF as a novel therapy target for treatment of acute allograft rejection.
Collapse
Affiliation(s)
- Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Chen Dai
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Rumeng Tan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Dongxia Ma
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Zhimin Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Bo Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Jincheng Feng
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, Ohio, United States of America
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Ministry of Health and Key Laboratory of Ministry of EducationWuhan, China
| |
Collapse
|
38
|
Kasala D, Yoon AR, Hong J, Kim SW, Yun CO. Evolving lessons on nanomaterial-coated viral vectors for local and systemic gene therapy. Nanomedicine (Lond) 2016; 11:1689-713. [PMID: 27348247 DOI: 10.2217/nnm-2016-0060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Viral vectors are promising gene carriers for cancer therapy. However, virus-mediated gene therapies have demonstrated insufficient therapeutic efficacy in clinical trials due to rapid dissemination to nontarget tissues and to the immunogenicity of viral vectors, resulting in poor retention at the disease locus and induction of adverse inflammatory responses in patients. Further, the limited tropism of viral vectors prevents efficient gene delivery to target tissues. In this regard, modification of the viral surface with nanomaterials is a promising strategy to augment vector accumulation at the target tissue, circumvent the host immune response, and avoid nonspecific interactions with the reticuloendothelial system or serum complement. In the present review, we discuss various chemical modification strategies to enhance the therapeutic efficacy of viral vectors delivered either locally or systemically. We conclude by highlighting the salient features of various nanomaterial-coated viral vectors and their prospects and directions for future research.
Collapse
Affiliation(s)
- Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Jinwoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Sung Wan Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea.,Department of Pharmaceutics & Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| |
Collapse
|
39
|
Lin HP, Zheng DJ, Li YP, Wang N, Chen SJ, Fu YC, Xu WC, Wei CJ. Incorporation of VSV-G produces fusogenic plasma membrane vesicles capable of efficient transfer of bioactive macromolecules and mitochondria. Biomed Microdevices 2016; 18:41. [PMID: 27165101 DOI: 10.1007/s10544-016-0066-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The objective of this study was to determine if plasma membrane vesicles (PMVs) could be exploited for efficient transfer of macro-biomolecules and mitochondria. PMVs were derived from mechanical extrusion, and made fusogenic (fPMVs) by incorporating the glycoprotein G of vesicular stomatitis virus (VSV-G). Confocal microscopy examination revealed that cytoplasmic proteins and mitochondria were enclosed in PMVs as evidenced by tracing with cytoplasmically localized and mitochondria-targeted EGFP, respectively. However, no fluorescence signal was detected in PMVs from cells whose nucleus was labeled with an EGFP-tagged histone H2B. Consistently, qRT-PCR measurement showed that mRNA, miRNA and mitochondrial DNA decreased slightly; while nuclear DNA was not measureable. Further, Western blot analysis revealed that cytoplasmic and membrane-bound proteins fell inconspicuously while nuclear proteins were barely detecsle. In addition, fPMVs carrying cytoplamic DsRed proteins transduced about ~40 % of recipient cells. The transfer of protein was further confirmed by using the inducible Cre/loxP system. Mitochondria transfer was found in about 20 % recipient cells after incubation with fPMVs for 5 h. To verify the functionalities of transferred mitochondria, mitochodria-deficient HeLa cells (Rho0) were generated and cultivated with fPMVs. Cell enumeration demonstrated that adding fPMVs into culture media stimulated Rho0 cell growth by 100 % as compared to the control. Lastly, MitoTracker and JC-1 staining showed that transferred mitochondria maintained normal shape and membrane potential in Rho0 cells. This study established a time-saving and efficient approach to delivering proteins and mitochondria by using fPMVs, which would be helpful for finding a cure to mitochondria-associated diseases. Graphical abstract Schematic of the delivery of macro-biomolecules and organelles by fPMVs. VSV-G-expressing cells were extruded through a 3 μm polycarbonate membrane filter to generate fusogenic plasma membrane vesicles (fPMVs), which contain bioactive molecules and organelles but not the nucleus. fPMVs can be endocytosed by target cells, while the cargo is released due to low-pH induced membrane fusion. These nucleus-free fPMVs are efficient at delivery of cytoplasmic proteins and mitochondria, leading to recovery of mitochondrial biogenesis and proliferative ability in mitochondria-deficient cells.
Collapse
Affiliation(s)
- Hao-Peng Lin
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - De-Jin Zheng
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Yun-Pan Li
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Na Wang
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Shao-Jun Chen
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Yu-Cai Fu
- Laboratory of Cell Senescence, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Wen-Can Xu
- Department of Endocrinology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Chi-Ju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China.
| |
Collapse
|
40
|
Kaneti L, Bronshtein T, Malkah Dayan N, Kovregina I, Letko Khait N, Lupu-Haber Y, Fliman M, Schoen BW, Kaneti G, Machluf M. Nanoghosts as a Novel Natural Nonviral Gene Delivery Platform Safely Targeting Multiple Cancers. NANO LETTERS 2016; 16:1574-82. [PMID: 26901695 DOI: 10.1021/acs.nanolett.5b04237] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Nanoghosts derived from mesenchymal stem cells and retaining their unique surface-associated tumor-targeting capabilities were redesigned as a selective and safe universal nonviral gene-therapy platform. pDNA-loaded nanoghosts efficiently targeted and transfected diverse cancer cells, in vitro and in vivo, in subcutaneous and metastatic orthotopic tumor models, leading to no adverse effects. Nanoghosts loaded with pDNA encoding for a cancer-toxic gene inhibited the growth of metastatic orthotopic lung cancer and subcutaneous prostate cancer models and dramatically prolonged the animals' survival.
Collapse
Affiliation(s)
- Limor Kaneti
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Tomer Bronshtein
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Natali Malkah Dayan
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Inna Kovregina
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Nitzan Letko Khait
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Yael Lupu-Haber
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Miguel Fliman
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Beth W Schoen
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Galoz Kaneti
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Marcelle Machluf
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| |
Collapse
|
41
|
Madrigal JL, Stilhano RS, Siltanen C, Tanaka K, Rezvani SN, Morgan RP, Revzin A, Han SW, Silva EA. Microfluidic generation of alginate microgels for the controlled delivery of lentivectors. J Mater Chem B 2016; 4:6989-6999. [DOI: 10.1039/c6tb02150f] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microgels fabricated through distinct microfluidic procedures encapsulate and release functioning lentivectors in a controlled manner.
Collapse
Affiliation(s)
| | | | | | - Kimberly Tanaka
- Department of Biomedical Engineering
- University of California
- Davis
- USA
| | - Sabah N. Rezvani
- Department of Biomedical Engineering
- University of California
- Davis
- USA
| | - Ryan P. Morgan
- Department of Biomedical Engineering
- University of California
- Davis
- USA
| | - Alexander Revzin
- Department of Biomedical Engineering
- University of California
- Davis
- USA
| | - Sang W. Han
- Department of Biophysics
- Federal University of São Paulo
- São Paulo
- Brazil
| | - Eduardo A. Silva
- Department of Biomedical Engineering
- University of California
- Davis
- USA
| |
Collapse
|
42
|
Pros and Cons of Antigen-Presenting Cell Targeted Tumor Vaccines. J Immunol Res 2015; 2015:785634. [PMID: 26583156 PMCID: PMC4637118 DOI: 10.1155/2015/785634] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 08/26/2015] [Accepted: 09/03/2015] [Indexed: 01/08/2023] Open
Abstract
In therapeutic antitumor vaccination, dendritic cells play the leading role since they decide if, how, when, and where a potent antitumor immune response will take place. Since the disentanglement of the complexity and merit of different antigen-presenting cell subtypes, antitumor immunotherapeutic research started to investigate the potential benefit of targeting these subtypes in situ. This review will discuss which antigen-presenting cell subtypes are at play and how they have been targeted and finally question the true meaning of targeting antitumor-based vaccines.
Collapse
|
43
|
Chu Y, Oum YH, Carrico IS. Surface modification via strain-promoted click reaction facilitates targeted lentiviral transduction. Virology 2015; 487:95-103. [PMID: 26499046 DOI: 10.1016/j.virol.2015.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 11/29/2022]
Abstract
As a result of their ability to integrate into the genome of both dividing and non-dividing cells, lentiviruses have emerged as a promising vector for gene delivery. Targeted gene transduction of specific cells and tissues by lentiviral vectors has been a major goal, which has proven difficult to achieve. We report a novel targeting protocol that relies on the chemoselective attachment of cancer specific ligands to unnatural glycans on lentiviral surfaces. This strategy exhibits minimal perturbation on virus physiology and demonstrates remarkable flexibility. It allows for targeting but can be more broadly useful with applications such as vector purification and immunomodulation.
Collapse
Affiliation(s)
- Yanjie Chu
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| | - Yoon Hyeun Oum
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| | - Isaac S Carrico
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA; Institute of Chemical Biology and Drug Discovery, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA.
| |
Collapse
|
44
|
Xu X, Ling Q, Wang J, Xie H, Wei X, Lu D, Hu Q, Zhang X, Wu L, Zhou L, Zheng S. Donor miR-196a-2 polymorphism is associated with hepatocellular carcinoma recurrence after liver transplantation in a Han Chinese population. Int J Cancer 2015; 138:620-9. [PMID: 26365437 DOI: 10.1002/ijc.29821] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 07/28/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Hangzhou China
| | - Qi Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Hangzhou China
| | - Jianguo Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Haiyang Xie
- Key Laboratory of Combined Multi-Organ Transplantation; Ministry of Public Health; Hangzhou China
| | - Xuyong Wei
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Di Lu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Qichao Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Xuanyu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
| | - Liming Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Hangzhou China
| | - Lin Zhou
- Key Laboratory of Combined Multi-Organ Transplantation; Ministry of Public Health; Hangzhou China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery; First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou Zhejiang China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Hangzhou China
- Key Laboratory of Combined Multi-Organ Transplantation; Ministry of Public Health; Hangzhou China
| |
Collapse
|
45
|
Walthers CM, Seidlits SK. Gene delivery strategies to promote spinal cord repair. Biomark Insights 2015; 10:11-29. [PMID: 25922572 PMCID: PMC4395076 DOI: 10.4137/bmi.s20063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 12/21/2022] Open
Abstract
Gene therapies hold great promise for the treatment of many neurodegenerative disorders and traumatic injuries in the central nervous system. However, development of effective methods to deliver such therapies in a controlled manner to the spinal cord is a necessity for their translation to the clinic. Although essential progress has been made to improve efficiency of transgene delivery and reduce the immunogenicity of genetic vectors, there is still much work to be done to achieve clinical strategies capable of reversing neurodegeneration and mediating tissue regeneration. In particular, strategies to achieve localized, robust expression of therapeutic transgenes by target cell types, at controlled levels over defined time periods, will be necessary to fully regenerate functional spinal cord tissues. This review summarizes the progress over the last decade toward the development of effective gene therapies in the spinal cord, including identification of appropriate target genes, improvements to design of genetic vectors, advances in delivery methods, and strategies for delivery of multiple transgenes with synergistic actions. The potential of biomaterials to mediate gene delivery while simultaneously providing inductive scaffolding to facilitate tissue regeneration is also discussed.
Collapse
|
46
|
Negro-Demontel ML, Saccardo P, Giacomini C, Yáñez-Muñoz RJ, Ferrer-Miralles N, Vazquez E, Villaverde A, Peluffo H. Comparative analysis of lentiviral vectors and modular protein nanovectors for traumatic brain injury gene therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14047. [PMID: 26015985 PMCID: PMC4362363 DOI: 10.1038/mtm.2014.47] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/25/2014] [Accepted: 08/28/2014] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) remains as one of the leading causes of mortality and morbidity worldwide and there are no effective treatments currently available. Gene therapy applications have emerged as important alternatives for the treatment of diverse nervous system injuries. New strategies are evolving with the notion that each particular pathological condition may require a specific vector. Moreover, the lack of detailed comparative studies between different vectors under similar conditions hampers the selection of an ideal vector for a given pathological condition. The potential use of lentiviral vectors versus several modular protein-based nanovectors was compared using a controlled cortical impact model of TBI under the same gene therapy conditions. We show that variables such as protein/DNA ratio, incubation volume, and presence of serum or chloroquine in the transfection medium impact on both nanovector formation and transfection efficiency in vitro. While lentiviral vectors showed GFP protein 1 day after TBI and increased expression at 14 days, nanovectors showed stable and lower GFP transgene expression from 1 to 14 days. No toxicity after TBI by any of the vectors was observed as determined by resulting levels of IL-1β or using neurological sticky tape test. In fact, both vector types induced functional improvement per se.
Collapse
Affiliation(s)
- María Luciana Negro-Demontel
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo , Montevideo, Uruguay ; Departmento de Histología y Embriología, Facultad de Medicina, UDELAR , Montevideo, Uruguay
| | - Paolo Saccardo
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona, Spain ; Department de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Barcelona, Spain ; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona, Spain
| | - Cecilia Giacomini
- Cátedra de Bioquímica, Departamento de Biociencias, Facultad de Química, UDELAR , Montevideo, Uruguay
| | | | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona, Spain ; Department de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Barcelona, Spain ; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona, Spain
| | - Esther Vazquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona, Spain ; Department de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Barcelona, Spain ; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona , Barcelona, Spain ; Department de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Barcelona, Spain ; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona, Spain
| | - Hugo Peluffo
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo , Montevideo, Uruguay ; Departmento de Histología y Embriología, Facultad de Medicina, UDELAR , Montevideo, Uruguay
| |
Collapse
|
47
|
Carbonaro Sarracino D, Tarantal AF, Lee CCI, Martinez M, Jin X, Wang X, Hardee CL, Geiger S, Kahl CA, Kohn DB. Effects of vector backbone and pseudotype on lentiviral vector-mediated gene transfer: studies in infant ADA-deficient mice and rhesus monkeys. Mol Ther 2014; 22:1803-16. [PMID: 24925206 DOI: 10.1038/mt.2014.88] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 05/11/2014] [Indexed: 01/05/2023] Open
Abstract
Systemic delivery of a lentiviral vector carrying a therapeutic gene represents a new treatment for monogenic disease. Previously, we have shown that transfer of the adenosine deaminase (ADA) cDNA in vivo rescues the lethal phenotype and reconstitutes immune function in ADA-deficient mice. In order to translate this approach to ADA-deficient severe combined immune deficiency patients, neonatal ADA-deficient mice and newborn rhesus monkeys were treated with species-matched and mismatched vectors and pseudotypes. We compared gene delivery by the HIV-1-based vector to murine γ-retroviral vectors pseudotyped with vesicular stomatitis virus-glycoprotein or murine retroviral envelopes in ADA-deficient mice. The vesicular stomatitis virus-glycoprotein pseudotyped lentiviral vectors had the highest titer and resulted in the highest vector copy number in multiple tissues, particularly liver and lung. In monkeys, HIV-1 or simian immunodeficiency virus vectors resulted in similar biodistribution in most tissues including bone marrow, spleen, liver, and lung. Simian immunodeficiency virus pseudotyped with the gibbon ape leukemia virus envelope produced 10- to 30-fold lower titers than the vesicular stomatitis virus-glycoprotein pseudotype, but had a similar tissue biodistribution and similar copy number in blood cells. The relative copy numbers achieved in mice and monkeys were similar when adjusted to the administered dose per kg. These results suggest that this approach can be scaled-up to clinical levels for treatment of ADA-deficient severe combined immune deficiency subjects with suboptimal hematopoietic stem cell transplantation options.
Collapse
Affiliation(s)
- Denise Carbonaro Sarracino
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Alice F Tarantal
- 1] Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, California USA [2] Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - C Chang I Lee
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, California USA
| | - Michele Martinez
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, California USA
| | - Xiangyang Jin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles California, USA
| | - Cinnamon L Hardee
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Sabine Geiger
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Christoph A Kahl
- 1] Division of Research Immunology/BMT, Children's Hospital Los Angeles, Los Angeles, California, USA [2] Current address: Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Donald B Kohn
- 1] Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA [2] Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
48
|
Okamoto KW, Amarasekare P, Petty ITD. Modeling oncolytic virotherapy: is complete tumor-tropism too much of a good thing? J Theor Biol 2014; 358:166-78. [PMID: 24810840 DOI: 10.1016/j.jtbi.2014.04.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 04/22/2014] [Accepted: 04/25/2014] [Indexed: 12/21/2022]
Abstract
The specific targeting of tumor cells by replication-competent oncolytic viruses is considered indispensable for realizing the potential of oncolytic virotherapy. Yet off-target infections by oncolytic viruses may increase virus production, further reducing tumor load. This ability may be critical when tumor-cell scarcity or the onset of an adaptive immune response constrain viral anti-tumoral efficacy. Here we develop a mathematical framework for assessing whether oncolytic viruses with reduced tumor-specificity can more effectively eliminate tumors while keeping losses to normal cell populations low. We find viruses that infect some normal cells can potentially balance the competing goals of tumor elimination and minimizing the effects on normal cell populations. Particularly when infected tissues can be regenerated, moderating rather than completely eliminating the ability of oncolytic viruses to infect and lyse normal cells could improve cancer treatment, with potentially fewer side-effects than conventional treatments such as chemotherapy.
Collapse
Affiliation(s)
- Kenichi W Okamoto
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA; Department of Entomology, North Carolina State University, Raleigh, NC 27695-7613, USA.
| | - Priyanga Amarasekare
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA.
| | - Ian T D Petty
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA.
| |
Collapse
|
49
|
Peptide-based technologies to alter adenoviral vector tropism: ways and means for systemic treatment of cancer. Viruses 2014; 6:1540-63. [PMID: 24699364 PMCID: PMC4014709 DOI: 10.3390/v6041540] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/15/2014] [Accepted: 03/20/2014] [Indexed: 12/11/2022] Open
Abstract
Due to the fundamental progress in elucidating the molecular mechanisms of human diseases and the arrival of the post-genomic era, increasing numbers of therapeutic genes and cellular targets are available for gene therapy. Meanwhile, the most important challenge is to develop gene delivery vectors with high efficiency through target cell selectivity, in particular under in situ conditions. The most widely used vector system to transduce cells is based on adenovirus (Ad). Recent endeavors in the development of selective Ad vectors that target cells or tissues of interest and spare the alteration of all others have focused on the modification of the virus broad natural tropism. A popular way of Ad targeting is achieved by directing the vector towards distinct cellular receptors. Redirecting can be accomplished by linking custom-made peptides with specific affinity to cellular surface proteins via genetic integration, chemical coupling or bridging with dual-specific adapter molecules. Ideally, targeted vectors are incapable of entering cells via their native receptors. Such altered vectors offer new opportunities to delineate functional genomics in a natural environment and may enable efficient systemic therapeutic approaches. This review provides a summary of current state-of-the-art techniques to specifically target adenovirus-based gene delivery vectors.
Collapse
|
50
|
Abstract
UNLABELLED Because of its very low human seroprevalence, vesicular stomatitis virus (VSV) has promise as a systemic oncolytic agent for human cancer therapy. However, as demonstrated in this report, the VSV infectious titer drops by 4 log units during the first hour of exposure to nonimmune human serum. This neutralization occurs relatively slowly and is mediated by the concerted actions of natural IgM and complement. Maraba virus, whose G protein is about 80% homologous to that of VSV, is relatively resistant to the neutralizing activity of nonimmune human serum. We therefore constructed and rescued a recombinant VSV whose G gene was replaced by the corresponding gene from Maraba virus. Comparison of the parental VSV and VSV with Maraba G substituted revealed nearly identical host range properties and replication kinetics on a panel of tumor cell lines. Moreover, in contrast to the parental VSV, the VSV with Maraba G substituted was resistant to nonimmune human serum. Overall, our data suggest that VSV with Maraba G substituted should be further investigated as a candidate for human systemic oncolytic virotherapy applications. IMPORTANCE Oncolytic virotherapy is a promising approach for the treatment of disseminated cancers, but antibody neutralization of circulating oncolytic virus particles remains a formidable barrier. In this work, we developed a pseudotyped vesicular stomatitis virus (VSV) with a glycoprotein of Maraba virus, a closely related but serologically distinct member of the family Rhabdoviridae, which demonstrated greatly diminished susceptibility to both nonimmune and VSV-immune serum neutralization. VSV with Maraba G substituted or lentiviral vectors should therefore be further investigated as candidates for human systemic oncolytic virotherapy and gene therapy applications.
Collapse
|