1
|
Moreira AS, Bezemer S, Faria TQ, Detmers F, Hermans P, Sierkstra L, Coroadinha AS, Peixoto C. Implementation of Novel Affinity Ligand for Lentiviral Vector Purification. Int J Mol Sci 2023; 24:3354. [PMID: 36834764 PMCID: PMC9966744 DOI: 10.3390/ijms24043354] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
The use of viral vectors as therapeutic products for multiple applications such as vaccines, cancer treatment, or gene therapies, has been growing exponentially. Therefore, improved manufacturing processes are needed to cope with the high number of functional particles required for clinical trials and, eventually, commercialization. Affinity chromatography (AC) can be used to simplify purification processes and generate clinical-grade products with high titer and purity. However, one of the major challenges in the purification of Lentiviral vectors (LVs) using AC is to combine a highly specific ligand with a gentle elution condition assuring the preservation of vector biological activity. In this work, we report for the first time the implementation of an AC resin to specifically purify VSV-G pseudotyped LVs. After ligand screening, different critical process parameters were assessed and optimized. A dynamic capacity of 1 × 1011 total particles per mL of resin was determined and an average recovery yield of 45% was found for the small-scale purification process. The established AC robustness was confirmed by the performance of an intermediate scale providing an infectious particles yield of 54%, which demonstrates the scalability and reproducibility of the AC matrix. Overall, this work contributes to increasing downstream process efficiency by delivering a purification technology that enables high purity, scalability, and process intensification in a single step, contributing to time-to-market reduction.
Collapse
Affiliation(s)
- Ana Sofia Moreira
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
- ITQB Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sandra Bezemer
- Thermo Fisher Scientific, 2333 CH Leiden, The Netherlands
| | - Tiago Q. Faria
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Frank Detmers
- Thermo Fisher Scientific, 2333 CH Leiden, The Netherlands
| | - Pim Hermans
- Thermo Fisher Scientific, 2333 CH Leiden, The Netherlands
| | | | - Ana Sofia Coroadinha
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Cristina Peixoto
- IBET Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| |
Collapse
|
2
|
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
|
3
|
Goulding J, Kondrashov A, Mistry SJ, Melarangi T, Vo NTN, Hoang DM, White CW, Denning C, Briddon SJ, Hill SJ. The use of fluorescence correlation spectroscopy to monitor cell surface β2-adrenoceptors at low expression levels in human embryonic stem cell-derived cardiomyocytes and fibroblasts. FASEB J 2021; 35:e21398. [PMID: 33710675 DOI: 10.1096/fj.202002268r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 12/31/2022]
Abstract
The importance of cell phenotype in determining the molecular mechanisms underlying β2 -adrenoceptor (β2AR) function has been noted previously when comparing responses in primary cells and recombinant model cell lines. Here, we have generated haplotype-specific SNAP-tagged β2AR human embryonic stem (ES) cell lines and applied fluorescence correlation spectroscopy (FCS) to study cell surface receptors in progenitor cells and in differentiated fibroblasts and cardiomyocytes. FCS was able to quantify SNAP-tagged β2AR number and diffusion in both ES-derived cardiomyocytes and CRISPR/Cas9 genome-edited HEK293T cells, where the expression level was too low to detect using standard confocal microscopy. These studies demonstrate the power of FCS in investigating cell surface β2ARs at the very low expression levels often seen in endogenously expressing cells. Furthermore, the use of ES cell technology in combination with FCS allowed us to demonstrate that cell surface β2ARs internalize in response to formoterol-stimulation in ES progenitor cells but not following their differentiation into ES-derived fibroblasts. This indicates that the process of agonist-induced receptor internalization is strongly influenced by cell phenotype and this may have important implications for drug treatment with long-acting β2AR agonists.
Collapse
Affiliation(s)
- Joëlle Goulding
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Alexander Kondrashov
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,Division of Cancer & Stem Cells, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Sarah J Mistry
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Tony Melarangi
- Division of Cancer & Stem Cells, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Nguyen T N Vo
- Division of Cancer & Stem Cells, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Duc M Hoang
- Division of Cancer & Stem Cells, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK.,Department of Cellular Manufacturing, Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| | - Carl W White
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Harry Perkins Institute of Medical Research and Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
| | - Chris Denning
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,Division of Cancer & Stem Cells, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Stephen J Briddon
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Stephen J Hill
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
4
|
Perry C, Rayat ACME. Lentiviral Vector Bioprocessing. Viruses 2021; 13:268. [PMID: 33572347 PMCID: PMC7916122 DOI: 10.3390/v13020268] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Lentiviral vectors (LVs) are potent tools for the delivery of genes of interest into mammalian cells and are now commonly utilised within the growing field of cell and gene therapy for the treatment of monogenic diseases and adoptive therapies such as chimeric antigen T-cell (CAR-T) therapy. This is a comprehensive review of the individual bioprocess operations employed in LV production. We highlight the role of envelope proteins in vector design as well as their impact on the bioprocessing of lentiviral vectors. An overview of the current state of these operations provides opportunities for bioprocess discovery and improvement with emphasis on the considerations for optimal and scalable processing of LV during development and clinical production. Upstream culture for LV generation is described with comparisons on the different transfection methods and various bioreactors for suspension and adherent producer cell cultivation. The purification of LV is examined, evaluating different sequences of downstream process operations for both small- and large-scale production requirements. For scalable operations, a key focus is the development in chromatographic purification in addition to an in-depth examination of the application of tangential flow filtration. A summary of vector quantification and characterisation assays is also presented. Finally, the assessment of the whole bioprocess for LV production is discussed to benefit from the broader understanding of potential interactions of the different process options. This review is aimed to assist in the achievement of high quality, high concentration lentiviral vectors from robust and scalable processes.
Collapse
Affiliation(s)
- Christopher Perry
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Gower St, London WC1E 6BT, UK;
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Andrea C. M. E. Rayat
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Gower St, London WC1E 6BT, UK;
| |
Collapse
|
5
|
Stevens LM, Zhang Y, Volnov Y, Chen G, Stein DS. Isolation of secreted proteins from Drosophila ovaries and embryos through in vivo BirA-mediated biotinylation. PLoS One 2019; 14:e0219878. [PMID: 31658274 PMCID: PMC6816556 DOI: 10.1371/journal.pone.0219878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023] Open
Abstract
The extraordinarily strong non-covalent interaction between biotin and avidin (kD = 10-14-10-16) has permitted this interaction to be used in a wide variety of experimental contexts. The Biotin Acceptor Peptide (BAP), a 15 amino acid motif that can be biotinylated by the E. coli BirA protein, has been fused to proteins-of-interest, making them substrates for in vivo biotinylation. Here we report on the construction and characterization of a modified BirA bearing signals for secretion and endoplasmic reticulum (ER) retention, for use in experimental contexts requiring biotinylation of secreted proteins. When expressed in the Drosophila female germline or ovarian follicle cells under Gal4-mediated transcriptional control, the modified BirA protein could be detected and shown to be enzymatically active in ovaries and progeny embryos. Surprisingly, however, it was not efficiently retained in the ER, and instead appeared to be secreted. To determine whether this secreted protein, now designated secBirA, could biotinylate secreted proteins, we generated BAP-tagged versions of two secreted Drosophila proteins, Torsolike (Tsl) and Gastrulation Defective (GD), which are normally expressed maternally and participate in embryonic pattern formation. Both Tsl-BAP and GD-BAP were shown to exhibit normal patterning activity. Co-expression of Tsl-BAP together with secBirA in ovarian follicle cells resulted in its biotinylation, which permitted its isolation from both ovaries and progeny embryos using Avidin-coupled affinity matrix. In contrast, co-expression with secBirA in the female germline did not result in detectable biotinylation of GD-BAP, possibly because the C-terminal location of the BAP tag made it inaccessible to BirA in vivo. Our results indicate that secBirA directs biotinylation of proteins bound for secretion in vivo, providing access to powerful experimental approaches for secreted proteins-of-interest. However, efficient biotinylation of target proteins may vary depending upon the location of the BAP tag or other structural features of the protein.
Collapse
Affiliation(s)
- Leslie M. Stevens
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- Section of Molecular Cell and Developmental Biology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Yuan Zhang
- Section of Molecular Cell and Developmental Biology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Yuri Volnov
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Geng Chen
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- Section of Molecular Cell and Developmental Biology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - David S. Stein
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- Section of Molecular Cell and Developmental Biology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
6
|
Reja SI, Minoshima M, Hori Y, Kikuchi K. Development of an effective protein-labeling system based on smart fluorogenic probes. J Biol Inorg Chem 2019; 24:443-455. [PMID: 31152238 DOI: 10.1007/s00775-019-01669-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 12/23/2022]
Abstract
Proteins are an important component of living systems and play a crucial role in various physiological functions. Fluorescence imaging of proteins is a powerful tool for monitoring protein dynamics. Fluorescent protein (FP)-based labeling methods are frequently used to monitor the movement and interaction of cellular proteins. However, alternative methods have also been developed that allow the use of synthetic fluorescent probes to target a protein of interest (POI). Synthetic fluorescent probes have various advantages over FP-based labeling methods. They are smaller in size than the fluorescent proteins, offer a wide variety of colors and have improved photochemical properties. There are various chemical recognition-based labeling techniques that can be used for labeling a POI with a synthetic probe. In this review, we focus on the development of protein-labeling systems, particularly the SNAP-tag, BL-tag, and PYP-tag systems, and understanding the fluorescence behavior of the fluorescently labeled target protein in these systems. We also discuss the smart fluorogenic probes for these protein-labeling systems and their applications. The fluorogenic protein labeling will be a useful tool to investigate complex biological phenomena in future work on cell biology.
Collapse
Affiliation(s)
- Shahi Imam Reja
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masafumi Minoshima
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Hori
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazuya Kikuchi
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan.
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
7
|
Mekkaoui L, Parekh F, Kotsopoulou E, Darling D, Dickson G, Cheung GW, Chan L, MacLellan-Gibson K, Mattiuzzo G, Farzaneh F, Takeuchi Y, Pule M. Lentiviral Vector Purification Using Genetically Encoded Biotin Mimic in Packaging Cell. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:155-165. [PMID: 30547049 PMCID: PMC6258877 DOI: 10.1016/j.omtm.2018.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 10/17/2018] [Indexed: 11/17/2022]
Abstract
Lentiviral vectors (LVs) have recently witnessed an increasing demand in research and clinical applications. Their current purification processes represent the main bottleneck in their widespread use, as the methods used are cumbersome and yield low recoveries. We aimed to develop a one-step method to specifically purify LVs, with high yields and reduced levels of impurities, using the biotin-streptavidin system. Herein, packaging HEK293T cells were genetically engineered with a cyclical biotin-mimicking peptide displayed on a CD8α stalk, termed cTag8. LVs were modified with cTag8 by its passive incorporation onto viral surfaces during budding, without viral protein engineering or hindrance on infectivity. Expression of cTag8 on LVs allowed complete capture of infectious particles by streptavidin magnetic beads. As cTag8 binds streptavidin in the nanomolar range, the addition of micromolar concentrations of biotin resulted in the release of captured LVs by competitive elution, with overall yields of ≥60%. Analysis of eluted LVs revealed high purity with a >3-log and 2-log reduction in DNA contamination and host cell proteins, respectively. This one-step purification was also tested for scalable vector processing using monolith affinity chromatography, with an encouraging preliminary overall yield of 20%. This method will be of valuable use for both research and clinical applications of LVs.
Collapse
Affiliation(s)
- Leila Mekkaoui
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Farhaan Parekh
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | | | - David Darling
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Glenda Dickson
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Gordon W. Cheung
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Lucas Chan
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Kirsty MacLellan-Gibson
- National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK
| | - Giada Mattiuzzo
- National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK
| | - Farzin Farzaneh
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Yasuhiro. Takeuchi
- National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK
- Division of Infection and Immunity, University College London, Rayne Building, 5 University Street, London WC1E 6JF, UK
| | - Martin Pule
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
- Corresponding author: Martin Pule, UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK.
| |
Collapse
|
8
|
Schimert KI, Cheng W. A method for tethering single viral particles for virus-cell interaction studies with optical tweezers. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2018; 10723:107233B. [PMID: 30872888 PMCID: PMC6411052 DOI: 10.1117/12.2500050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Direct optical trapping of single viral particles allows characterization of individual particles in suspension with single-molecule sensitivity. Alternative to direct optical trapping of particles, individual particles may be tethered specifically in suspension for manipulation by optical tweezers indirectly, which could be useful for studies of virus-cell interactions. One specific example is the interactions between cell surface receptors and the envelope glycoproteins (Env) on the surface of human immunodeficiency virus type 1 (HIV-1). Env binds to cellular receptors and undergoes a series of conformational changes, culminating in fusion of the viral and cellular membranes that mediates viral entry into cells. In addition to being required for cellular infection, Env is also the sole target for neutralizing antibodies. Thus, significant research has focused on elucidating the structure of Env and the mechanism of HIV-1 entry. However, current methods are unable to resolve the dynamics and stoichiometry of Env binding to cellular receptors during the entry process. Fluorescence and electron microscopy have visualized Env clusters in the viral membrane, but the extent to which these clusters actually bind to cellular receptors, and the mechanism of cluster formation, remain unclear. We describe the development of an optical tweezers technique that can potentially address these questions by delivering a single HIV-1 virion to a live cell with minimal perturbation to the system. Our method can be used to quantitatively probe the physical interactions between Env and cellular receptors in their native environment, which may reveal critical parameters in HIV-1 entry. Furthermore, our method can be used to investigate other protein-protein interactions in the context of live cells, such as the recognition of particulate antigens by B cells, thus offering insight into fundamental features of protein-mediated receptor activation.
Collapse
Affiliation(s)
- Kristin I Schimert
- Biophysics Program, University of Michigan, 930 North University Avenue, Room 4028, Ann Arbor, MI 48109, USA
| | - Wei Cheng
- Biophysics Program, University of Michigan, 930 North University Avenue, Room 4028, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan Medical School; University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA
- Department of Biological Chemistry, University of Michigan Medical School; University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
McNally DJ, Darling D, Farzaneh F, Levison PR, Slater NKH. Optimised concentration and purification of retroviruses using membrane chromatography. J Chromatogr A 2014; 1340:24-32. [PMID: 24685165 PMCID: PMC4003387 DOI: 10.1016/j.chroma.2014.03.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 03/04/2014] [Accepted: 03/08/2014] [Indexed: 01/21/2023]
Abstract
An in investigation in to the use of membrane chromatography for the purification of a γ-retrovirus was undertaken. The first report of a capacity for γ-retrovirus binding to a membrane chromatography device is presented. A process that produces a large increase in concentration and purity of the studied γ-retrovirus was identified. Proteomic techniques were used to identify the protein impurities removed and co-purified with the virus containing eluate.
The ability of an anion exchange membrane to purify a γ-retrovirus was assessed and optimised with respect to different loading and wash buffers. Recoveries of infectious virus greater than 50% were consistently obtained, while specific titre was increased up to one thousand fold when compared to the material loaded. Specific proteins removed and retained by this optimised process were identified by mass spectrometry. It was possible to successfully bind and elute the equivalent of 1.27 × 108 Ifu/ml of ion exchange membrane. This could then be highly concentrated, with infectious virus concentrated to a maximum of 420-fold compared to the load.
Collapse
Affiliation(s)
- D J McNally
- Department of Chemical Engineering and Biotechnology, New Museums Site, Pembroke St, Cambridge CB2 3RA, UK.
| | - D Darling
- King's College London, 123 Coldharbour Lane, London SE5 9NU, UK
| | - F Farzaneh
- King's College London, 123 Coldharbour Lane, London SE5 9NU, UK
| | - P R Levison
- Pall Europe Limited, 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, Hampshire, UK
| | - N K H Slater
- Department of Chemical Engineering and Biotechnology, New Museums Site, Pembroke St, Cambridge CB2 3RA, UK
| |
Collapse
|
10
|
Postexit surface engineering of retroviral/lentiviral vectors. BIOMED RESEARCH INTERNATIONAL 2013; 2013:253521. [PMID: 23691494 PMCID: PMC3652111 DOI: 10.1155/2013/253521] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/15/2013] [Indexed: 11/25/2022]
Abstract
Gene delivery vectors based on retroviral or lentiviral particles are considered powerful tools for biomedicine and biotechnology applications. Such vectors require modification at the genomic level in the form of rearrangements to allow introduction of desired genes and regulatory elements (genotypic modification) as well as engineering of the physical virus particle (phenotypic modification) in order to mediate efficient and safe delivery of the genetic information to the target cell nucleus. Phenotypic modifications are typically introduced at the genomic level through genetic manipulation of the virus producing cells. However, this paper focuses on methods which allow modification of viral particle surfaces after they have exited the cell, that is, directly on the viral particles in suspension. These methods fall into three categories: (i) direct covalent chemical modification, (ii) membrane-topic reagents, and (iii) adaptor systems. Current applications of such techniques will be introduced and their advantages and disadvantages will be discussed.
Collapse
|
11
|
Segura MM, Mangion M, Gaillet B, Garnier A. New developments in lentiviral vector design, production and purification. Expert Opin Biol Ther 2013; 13:987-1011. [PMID: 23590247 DOI: 10.1517/14712598.2013.779249] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Lentiviruses are a very potent class of viral vectors for which there is presently a rapidly growing interest for a number of gene therapy. However, their construction, production and purification need to be performed according to state-of-the-art techniques in order to obtain sufficient quantities of high purity material of any usefulness and safety. AREAS COVERED The recent advances in the field of recombinant lentivirus vector design, production and purification will be reviewed with an eye toward its utilization for gene therapy. Such a review should be helpful for the potential user of this technology. EXPERT OPINION The principal hurdles toward the use of recombinant lentivirus as a gene therapy vector are the low titer at which it is produced as well as the difficulty to purify it at an acceptable level without degrading it. The recent advances in the bioproduction of this vector suggest these issues are about to be resolved, making the retrovirus gene therapy a mature technology.
Collapse
Affiliation(s)
- Maria Mercedes Segura
- Chemical Engineering Department, Universitat Autònoma de Barcelona, Campus Bellaterra, Cerdanyola del Vallès (08193), Barcelona, Spain
| | | | | | | |
Collapse
|
12
|
Reddy LH, Arias JL, Nicolas J, Couvreur P. Magnetic nanoparticles: design and characterization, toxicity and biocompatibility, pharmaceutical and biomedical applications. Chem Rev 2012; 112:5818-78. [PMID: 23043508 DOI: 10.1021/cr300068p] [Citation(s) in RCA: 1121] [Impact Index Per Article: 93.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- L Harivardhan Reddy
- Laboratoire de Physico-Chimie, Pharmacotechnie et Biopharmacie, Université Paris-Sud XI, UMR CNRS, Faculté de Pharmacie, IFR, Châtenay-Malabry, France
| | | | | | | |
Collapse
|
13
|
Darton N, Darling D, Townsend M, McNally D, Farzaneh F, Slater N. Lentivirus capture directly from cell culture with Q-functionalised microcapillary film chromatography. J Chromatogr A 2012; 1251:236-239. [DOI: 10.1016/j.chroma.2012.06.072] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 01/19/2023]
|
14
|
Ta HT, Peter K, Hagemeyer CE. Enzymatic Antibody Tagging: Toward a Universal Biocompatible Targeting Tool. Trends Cardiovasc Med 2012; 22:105-11. [DOI: 10.1016/j.tcm.2012.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Roux KJ, Kim DI, Raida M, Burke B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. ACTA ACUST UNITED AC 2012; 196:801-10. [PMID: 22412018 PMCID: PMC3308701 DOI: 10.1083/jcb.201112098] [Citation(s) in RCA: 1546] [Impact Index Per Article: 128.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Proximity-dependent biotin identification (BioID) is a new approach making use of biotin ligase fusion proteins for the identification of both interacting and neighboring proteins in their native cellular environment. We have developed a new technique for proximity-dependent labeling of proteins in eukaryotic cells. Named BioID for proximity-dependent biotin identification, this approach is based on fusion of a promiscuous Escherichia coli biotin protein ligase to a targeting protein. BioID features proximity-dependent biotinylation of proteins that are near-neighbors of the fusion protein. Biotinylated proteins may be isolated by affinity capture and identified by mass spectrometry. We apply BioID to lamin-A (LaA), a well-characterized intermediate filament protein that is a constituent of the nuclear lamina, an important structural element of the nuclear envelope (NE). We identify multiple proteins that associate with and/or are proximate to LaA in vivo. The most abundant of these include known interactors of LaA that are localized to the NE, as well as a new NE-associated protein named SLAP75. Our results suggest BioID is a useful and generally applicable method to screen for both interacting and neighboring proteins in their native cellular environment.
Collapse
Affiliation(s)
- Kyle J Roux
- Sanford Children's Health Research Center, Sanford Research/University of South Dakota, Sioux Falls, SD 57104, USA.
| | | | | | | |
Collapse
|
16
|
Ng S, Jafari MR, Derda R. Bacteriophages and viruses as a support for organic synthesis and combinatorial chemistry. ACS Chem Biol 2012; 7:123-38. [PMID: 21988453 DOI: 10.1021/cb200342h] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Display of polypeptide on the coat proteins of bacteriophages and viruses is a powerful tool for selection and amplification of libraries of great diversity. Chemical diversity of these libraries, however, is limited to libraries made of natural amino acid side chains. Bacteriophages and viruses can be modified chemically; peptide libraries presented on phage thus can be functionalized to yield moieties that cannot be encoded genetically. In this review, we summarize the possibilities for using bacteriophage and viral particles as support for the synthesis of diverse chemically modified peptide libraries. This review critically summarizes the key chemical considerations for on-phage syntheses such as selection of reactions compatible with protein of phage, modification of phage "support" that renders it more suitable for reactions, and characterization of reaction efficiency.
Collapse
Affiliation(s)
- Simon Ng
- Department
of Chemistry and Alberta Innovates Centre
for Carbohydrate Science, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Mohammad R. Jafari
- Department
of Chemistry and Alberta Innovates Centre
for Carbohydrate Science, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Ratmir Derda
- Department
of Chemistry and Alberta Innovates Centre
for Carbohydrate Science, University of Alberta, Edmonton, AB T6G 2G2, Canada
| |
Collapse
|
17
|
Han H, Liu Q, He W, Ong K, Liu X, Gao B. An efficient vector system to modify cells genetically. PLoS One 2011; 6:e26380. [PMID: 22096482 PMCID: PMC3214020 DOI: 10.1371/journal.pone.0026380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 09/26/2011] [Indexed: 11/18/2022] Open
Abstract
The transfer of foreign genes into mammalian cells has been essential for understanding the functions of genes and mechanisms of genetic diseases, for the production of coding proteins and for gene therapy applications. Currently, the identification and selection of cells that have received transferred genetic material can be accomplished by methods, including drug selection, reporter enzyme detection and GFP imaging. These methods may confer antibiotic resistance, or be disruptive, or require special equipment. In this study, we labeled genetically modified cells with a cell surface biotinylation tag by co-transfecting cells with BirA, a biotin ligase. The modified cells can be quickly isolated for downstream applications using a simple streptavidin bead method. This system can also be used to screen cells expressing two sets of genes from separate vectors.
Collapse
Affiliation(s)
- Huamin Han
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Graduate University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Qingjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Wen He
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Graduate University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Kristy Ong
- UCL Institute of Child Health, London, United Kingdom
| | - Xiaoli Liu
- Epigen Biotec Ltd, Beijing, People's Republic of China
| | - Bin Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
- China-Japan Joint Laboratory of Molecular Immunology and Microbiology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
18
|
Plank C, Zelphati O, Mykhaylyk O. Magnetically enhanced nucleic acid delivery. Ten years of magnetofection-progress and prospects. Adv Drug Deliv Rev 2011; 63:1300-31. [PMID: 21893135 PMCID: PMC7103316 DOI: 10.1016/j.addr.2011.08.002] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/28/2022]
Abstract
Nucleic acids carry the building plans of living systems. As such, they can be exploited to make cells produce a desired protein, or to shut down the expression of endogenous genes or even to repair defective genes. Hence, nucleic acids are unique substances for research and therapy. To exploit their potential, they need to be delivered into cells which can be a challenging task in many respects. During the last decade, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as magnetofection. In this review we summarize the progress and achievements in this field of research. We discuss magnetic formulations of vectors for nucleic acid delivery and their characterization, mechanisms of magnetofection, and the application of magnetofection in viral and nonviral nucleic acid delivery in cell culture and in animal models. We summarize results that have been obtained with using magnetofection in basic research and in preclinical animal models. Finally, we describe some of our recent work and end with some conclusions and perspectives.
Collapse
|
19
|
Nesbeth DN, Perez-Pardo MA, Ali S, Ward J, Keshavarz-Moore E. Growth and productivity impacts of periplasmic nuclease expression in an Escherichia coli Fab' fragment production strain. Biotechnol Bioeng 2011; 109:517-27. [PMID: 21898368 DOI: 10.1002/bit.23316] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 08/03/2011] [Accepted: 08/17/2011] [Indexed: 11/09/2022]
Abstract
Host cell engineering is becoming a realistic option in whole bioprocess strategies to maximize product manufacturability. High molecular weight (MW) genomic DNA currently hinders bioprocessing of Escherichia coli by causing viscosity in homogenate feedstocks. We previously showed that co-expressing Staphylococcal nuclease and human Fab' fragment in the periplasm of E. coli enables auto-hydrolysis of genomic DNA upon cell disruption, with a consequent reduction in feedstock viscosity and improvement in clarification performance. Here we report the impact of periplasmic nuclease expression on stability of DNA and Fab' fragment in homogenates, host-strain growth kinetics, cell integrity at harvest and Fab' fragment productivity. Nuclease and Fab' plasmids were shown to exert comparable levels of growth burden on the host W3110 E. coli strain. Nuclease co-expression did not compromise either the growth performance or volumetric yield of the production strain. 0.5 g/L Fab' fragment (75 L scale) and 0.7 g/L (20 L scale) was achieved for both unmodified and cell-engineered production strains. Unexpectedly, nuclease-modified cells achieved maximum Fab' levels 8-10 h earlier than the original, unmodified production strain. Scale-down studies of homogenates showed that nuclease-mediated hydrolysis of high MW DNA progressed to completion within minutes of homogenization, even when homogenates were chilled on ice, with no loss of Fab' product and no need for additional co-factors or buffering.
Collapse
Affiliation(s)
- Darren N Nesbeth
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 7JE, UK.
| | | | | | | | | |
Collapse
|
20
|
Faster generation of hiPSCs by coupling high-titer lentivirus and column-based positive selection. Nat Protoc 2011; 6:701-14. [PMID: 21637193 DOI: 10.1038/nprot.2011.320] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The protocols described here address methods used in two crucial stages in the retroviral reprogramming of somatic cells to produce human induced pluripotent stem cell (hiPSC) lines. The first is an optimized method for producing lentivirus at an efficiency 600-fold greater than previously published, and it includes conjugation of the lentivirus to streptavidin superparamagnetic particles; this process takes 8 d. The second method enables the isolation of true hiPSCs immediately after somatic cell reprogramming and involves column-based positive selection of cells expressing the pluripotency marker TRA-1-81. This process takes 2 h and, as it is directly compatible with feeder-free culture, the time burden of manually identifying and mechanically propagating hiPSC colonies is reduced drastically. Taken together, these methods accelerate the production of hiPSCs and enable lines to be isolated, expanded to approxiamtely 10⁷ cells and cryopreserved within 6-8 weeks.
Collapse
|
21
|
Niers JM, Chen JW, Weissleder R, Tannous BA. Enhanced in vivo imaging of metabolically biotinylated cell surface reporters. Anal Chem 2011; 83:994-9. [PMID: 21214190 DOI: 10.1021/ac102758m] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metabolic biotinylation of intracellular and secreted proteins as well as surface receptors in mammalian cells provides a versatile way to monitor gene expression; to purify and target viral vectors; to monitor cell and tumor distribution in real time in vivo; to label cells for isolation; and to tag proteins for purification, localization, and trafficking. Here, we show that metabolic biotinylation of proteins fused to the bacterial biotin acceptor peptides (BAP) varies among different mammalian cell types and can be enhanced by over 10-fold upon overexpression of the bacterial biotin ligase directed to the same cellular compartment as the fusion protein. We also show that in vivo imaging of metabolically biotinylated cell surface receptors using streptavidin conjugates is significantly enhanced upon coexpression of bacterial biotin ligase in the secretory pathway. These findings have practical applications in designing more efficient targeting and imaging strategies.
Collapse
Affiliation(s)
- Johanna M Niers
- Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
22
|
Dick E, Matsa E, Bispham J, Reza M, Guglieri M, Staniforth A, Watson S, Kumari R, Lochmüller H, Young L, Darling D, Denning C. Two new protocols to enhance the production and isolation of human induced pluripotent stem cell lines. Stem Cell Res 2010; 6:158-67. [PMID: 21095172 DOI: 10.1016/j.scr.2010.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/12/2010] [Accepted: 10/14/2010] [Indexed: 12/31/2022] Open
Abstract
There are two critical stages in the retroviral reprogramming of somatic cells to produce human induced pluripotent stem cell (hiPSC) lines. One is the production of high titer virus required to reprogram somatic cells; the other is identification of true hiPSC colonies from heterogeneous cell populations, and their isolation and expansion to generate a sustainable, pluripotent stem cell line. Here we describe simple, time-saving methods to address the current difficulties at these two critical junctures. First, we have developed a method to increase the number of infectious viral units 600-fold. Second, we have developed a TRA-1-81-based positive selection column method for isolating "true" hiPSCs from the heterogeneous cell populations, which overcomes the labor-intensive and highly subjective method of manual selection of hiPSC colonies. We have used these techniques to produce 8 hiPSC lines from human fibroblasts and we believe that they are of considerable utility to researchers in the hiPSC field.
Collapse
Affiliation(s)
- Emily Dick
- Wolfson Centre for Stem Cells, Tissue Engineering & Modelling, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lesch HP, Kaikkonen MU, Pikkarainen JT, Ylä-Herttuala S. Avidin-biotin technology in targeted therapy. Expert Opin Drug Deliv 2010; 7:551-64. [PMID: 20233034 DOI: 10.1517/17425241003677749] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD The goal of drug targeting is to increase the concentration of the drug in the vicinity of the cells responsible for disease without affecting healthy cells. Many approaches in cancer treatment are limited because of their broad range of unwanted side effects on healthy cells. Targeting can reduce side effects and increase efficacy of drugs in the patient. AREAS COVERED IN THIS REVIEW Avidin, originally isolated from chicken eggs, and its bacterial analogue, streptavidin, from Streptomyces avidinii, have extremely high affinity for biotin. This unique feature is the basis of avidin-biotin technology. This article reviews the current status of avidin-biotin systems and their use for pretargeted drug delivery and vector targeting. WHAT THE READER WILL GAIN The reader will gain an understanding of the following approaches using the avidin-biotin system: i) targeting antibodies and therapeutic molecules are administered separately leading to a reduction of drug dose in normal tissues compared with conventional (radio)immunotherapies; ii) introducing avidin gene into specific tissues by local gene transfer, which subsequently can sequester and concentrate considerable amounts of therapeutic ligands; and iii) enabling transductional targeting of gene therapy vectors. TAKE HOME MESSAGE Avidin and biotin technology has proved to be an extremely versatile tool with broad applications, such as pretargeting, delivering avidin gene into cells enabling targeting of biotinylated compounds and targeting of viral vectors.
Collapse
Affiliation(s)
- Hanna P Lesch
- University of Eastern Finland, A.I. Virtanen institute, Department of Biotechnology and Molecular Medicine, Kuopio, Finland
| | | | | | | |
Collapse
|
24
|
Tresilwised N, Pithayanukul P, Mykhaylyk O, Holm PS, Holzmüller R, Anton M, Thalhammer S, Adigüzel D, Döblinger M, Plank C. Boosting Oncolytic Adenovirus Potency with Magnetic Nanoparticles and Magnetic Force. Mol Pharm 2010; 7:1069-89. [DOI: 10.1021/mp100123t] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nittaya Tresilwised
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Pimolpan Pithayanukul
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Olga Mykhaylyk
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Per Sonne Holm
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Regina Holzmüller
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Martina Anton
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Stefan Thalhammer
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Denis Adigüzel
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Markus Döblinger
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Christian Plank
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| |
Collapse
|
25
|
Chen R, Folarin N, Ho VH, McNally D, Darling D, Farzaneh F, Slater NK. Affinity recovery of lentivirus by diaminopelargonic acid mediated desthiobiotin labelling. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878:1939-45. [DOI: 10.1016/j.jchromb.2010.05.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 05/12/2010] [Accepted: 05/17/2010] [Indexed: 11/26/2022]
|
26
|
Host-encoded reporters for the detection and purification of multiple enveloped viruses. J Virol Methods 2010; 167:178-85. [PMID: 20399809 PMCID: PMC2916077 DOI: 10.1016/j.jviromet.2010.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 03/31/2010] [Accepted: 04/08/2010] [Indexed: 02/07/2023]
Abstract
The identification of host cell factors for virus replication holds great promise for the development of new antiviral therapies. Recently, high-throughput screening methods have emerged as powerful tools to identify candidate host factors for therapeutic intervention. The development of assay systems suitable for large-scale automated screening is of particular importance for novel viruses with high pathogenic potential for which limited biological information can be developed in a short period of time. This report presents a general enzymatic reporter system for the detection and characterization of multiple enveloped viruses that does not rely on engineering of the virus. Instead, reporter enzymes are incorporated into virus particles by targeting to lipid microdomains in producer cells. The approach allows a variety of human pathogenic enveloped viruses to be detected by sensitive, inexpensive and automatable enzymatic assays. Tagged viruses can be purified quickly and efficiently by a magnetic bead-based capture method. The method allows general detection of enveloped viruses without prior reference to their sequence.
Collapse
|
27
|
Weber W, Lienhart C, Daoud-El Baba M, Grass RN, Kohler T, Müller R, Stark WJ, Fussenegger M. Magnet-guided transduction of mammalian cells and mice using engineered magnetic lentiviral particles. J Biotechnol 2009; 141:118-22. [PMID: 19433214 DOI: 10.1016/j.jbiotec.2009.02.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 02/16/2009] [Accepted: 02/26/2009] [Indexed: 02/08/2023]
Abstract
Targeted delivery of therapeutic transgenes into specific cells remains a highly relevant challenge for tissue engineering and future gene-based therapies. We have designed streptavidin-pseudotyped lentiviral particles which upon coupling with biotinylated magnetic carbon-coated cobalt nanoparticles could be guided by magnetic fields to site-specifically transduce desired target cells in culture as well as in mice. Magnetic patterns projected onto monolayer cultures were replicated by fluorescent cells following targeted transduction by magnetic lentiviral particles engineered for constitutive expression of the green fluorescent protein (GFP). Even after intravenous injection into mice magnetic GFP-transgenic lentiviral particles could be guided to a preferred transduction site in the animal using a magnetic field. Magnet-guided transgene delivery producing desired patterns of transduced cell populations may enable the design of defined tissue topologies and provide site-specific transduction of therapeutic transgenes for cell-specific interventions in future gene and cancer therapies.
Collapse
Affiliation(s)
- Wilfried Weber
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Cheeks M, Kamal N, Sorrell A, Darling D, Farzaneh F, Slater N. Immobilized metal affinity chromatography of histidine-tagged lentiviral vectors using monolithic adsorbents. J Chromatogr A 2009; 1216:2705-11. [DOI: 10.1016/j.chroma.2008.08.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 08/07/2008] [Accepted: 08/08/2008] [Indexed: 11/30/2022]
|
29
|
Howarth M, Ting AY. Imaging proteins in live mammalian cells with biotin ligase and monovalent streptavidin. Nat Protoc 2008; 3:534-45. [PMID: 18323822 DOI: 10.1038/nprot.2008.20] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This protocol describes a simple and efficient way to label specific cell surface proteins with biophysical probes on mammalian cells. Cell surface proteins tagged with a 15-amino acid peptide are biotinylated by Escherichia coli biotin ligase (BirA), whereas endogenous proteins are not modified. The biotin group then allows sensitive and stable binding by streptavidin conjugates. This protocol describes the optimal use of BirA and streptavidin for site-specific labeling and also how to produce BirA and monovalent streptavidin. Streptavidin is tetravalent and the cross-linking of biotinylated targets disrupts many of streptavidin's applications. Monovalent streptavidin has only a single functional biotin-binding site, but retains the femtomolar affinity, low off-rate and high thermostability of wild-type streptavidin. Site-specific biotinylation and streptavidin staining take only a few minutes, while expression of BirA takes 4 d and expression of monovalent streptavidin takes 8 d.
Collapse
Affiliation(s)
- Mark Howarth
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA.
| | | |
Collapse
|
30
|
Guinn B, Casey G, Collins S, O'Brien T, Alexander MY, Tangney M. Tripartite Meeting in Gene and Cell Therapy, 2008: Irish Society for Gene and Cell Therapy, British Society for Gene Therapy, and International Society for Cell and Gene Therapy of Cancer. Hum Gene Ther 2008; 19:967-78. [DOI: 10.1089/hum.2008.085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Barbara Guinn
- Department of Haematological Medicine, King's College London School of Medicine, London SE5 9NU, United Kingdom
| | - Garrett Casey
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| | - Sara Collins
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| | - Tim O'Brien
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - M. Yvonne Alexander
- Cardiovascular Group, School of Medicine, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Mark Tangney
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| |
Collapse
|
31
|
Kaikkonen MU, Viholainen JI, Närvänen A, Ylä-Herttuala S, Airenne KJ. Targeting and purification of metabolically biotinylated baculovirus. Hum Gene Ther 2008; 19:589-600. [PMID: 18479188 DOI: 10.1089/hum.2007.177] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Targeting viral entry is one of the major goals in the development of vectors for gene therapy. Ideally, the coupling of each new targeting motif would not require changes in vector structure. To achieve this, we developed novel metabolically biotinylated baculoviral vectors by displaying a small biotin acceptor peptide (BAP) fused either to different sites in the baculovirus glycoprotein gp64 or to the transmembrane anchor of vesicular stomatitis virus G protein. Baculoviral particles were biotinylated during vector production by coexpression of Escherichia coli biotin ligase (BirA). The insertion of BAP at amino acid position 283 of gp64 resulted in the most efficient biotin display. Unlike vectors with lower biotin display, these vectors also showed improved transduction when retargeted to transferrin, epidermal growth factor, and CD46 receptors overexpressed on rat glioma and human ovarian carcinoma cells. Biotinylated baculoviral vectors could also be concentrated by one-step magnetic particle-based capture to reach titers up to 10(10) plaque-forming units/ml. These results demonstrate the utility of metabolically biotinylated baculovirus for vector targeting and viral purification applications.
Collapse
Affiliation(s)
- Minna U Kaikkonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, FIN-70211 Kuopio, Finland
| | | | | | | | | |
Collapse
|
32
|
Saini V, Martyshkin DV, Mirov SB, Perez A, Perkins G, Ellisman MH, Towner VD, Wu H, Pereboeva L, Borovjagin A, Curiel DT, Everts M. An adenoviral platform for selective self-assembly and targeted delivery of nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2008; 4:262-269. [PMID: 18200644 DOI: 10.1002/smll.200700403] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Metallic nanoparticles (NPs) can be used for the diagnosis, imaging, and therapy of tumors and cardiovascular disease. However, targeted delivery of NPs to specific cells remains a major limitation for clinical realization of these potential treatment options. Herein, a novel strategy for the specific coupling of NPs to a targeted adenoviral (Ad) platform to deliver NPs to specific cells is defined. Genetic manipulation of the gene-therapy vector is combined with a specific chemical coupling strategy. In particular, a high-affinity interaction between a sequence of six-histidine amino acid residues genetically incorporated into Ad capsid proteins and nickel(II) nitrilotriacetic acid on the surface of gold NPs is employed. The selective self-assembly of gold NPs and Ad vectors into multifunctional platforms does not negatively affect the targeting of Ad to specific cells. This opens the possibility of using Ad vectors for targeted NP delivery, thereby providing a new type of combinatorial approach for the treatment of diseases that involves both nanotechnology and gene therapy.
Collapse
Affiliation(s)
- Vaibhav Saini
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mykhaylyk O, Antequera YS, Vlaskou D, Plank C. Generation of magnetic nonviral gene transfer agents and magnetofection in vitro. Nat Protoc 2008; 2:2391-411. [PMID: 17947981 DOI: 10.1038/nprot.2007.352] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
This protocol details how to design and conduct experiments to deliver nucleic acids to adherent and suspension cell cultures in vitro by magnetic force-assisted transfection using self-assembled complexes of nucleic acids and cationic lipids or polymers (nonviral gene vectors), which are associated with magnetic (nano) particles. These magnetic complexes are sedimented onto the surface of the cells to be transfected within minutes by the application of a magnetic gradient field. As the diffusion barrier to nucleic acid delivery is overcome, the full vector dose is targeted to the cell surface and transfection is synchronized. In this manner, the transfection process is accelerated and transfection efficiencies can be improved up to several 1,000-fold compared with transfections carried out with nonmagnetic gene vectors. This protocol describes how to accomplish the following stages: synthesis of magnetic nanoparticles for magnetofection; testing the association of DNA with the magnetic components of the transfection complex; preparation of magnetic lipoplexes and polyplexes; magnetofection; and data processing. The synthesis and characterization of magnetic nanoparticles can be accomplished within 3-5 d. Cell culture and transfection is then estimated to take 3 d. Transfected gene expression analysis, cell viability assays and calibration will probably take a few hours. This protocol can be used for cells that are difficult to transfect, such as primary cells, and may also be applied to viral nucleic acid delivery. With only minor alterations, this protocol can also be useful for magnetic cell labeling for cell tracking studies and, as it is, will be useful for screening vector compositions and novel magnetic nanoparticle preparations for optimized transfection efficiency in any cell type.
Collapse
Affiliation(s)
- Olga Mykhaylyk
- Institute of Experimental Oncology, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany.
| | | | | | | |
Collapse
|
34
|
al Yacoub N, Romanowska M, Haritonova N, Foerster J. Optimized production and concentration of lentiviral vectors containing large inserts. J Gene Med 2007; 9:579-84. [PMID: 17533614 DOI: 10.1002/jgm.1052] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Generation of high titer lentiviral stocks and efficient virus concentration are central to maximize the utility of lentiviral technology. Here we evaluate published protocols for lentivirus production on a range of transfer vectors differing in size (7.5-13.2 kb). We present a modified virus production protocol robustly yielding useful titers (up to 10(7)/ml) for a range of different transfer vectors containing packaging inserts up to 7.5 kb. Moreover, we find that virus recovery after concentration by ultracentrifugation depends on the size of the packaged inserts, heavily decreasing for large packaged inserts. We describe a fast (4 h) centrifugation protocol at reduced speed allowing high virus recovery even for large and fragile lentivirus vectors. The protocols outlined in the current report should be useful for many labs interested in producing and concentrating high titer lentiviral stocks.
Collapse
Affiliation(s)
- Nadya al Yacoub
- Department of Dermatology, Charité University Hospital, Berlin, Germany
| | | | | | | |
Collapse
|
35
|
Barat B, Wu AM. Metabolic biotinylation of recombinant antibody by biotin ligase retained in the endoplasmic reticulum. BIOMOLECULAR ENGINEERING 2007; 24:283-91. [PMID: 17379573 PMCID: PMC2682619 DOI: 10.1016/j.bioeng.2007.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 02/01/2007] [Indexed: 11/23/2022]
Abstract
Due to its strength and specificity, the interaction between avidin and biotin has been used in a variety of scientific and medical applications ranging from immunohistochemistry to drug targeting. The present study describes two methods for biotinylation of proteins secreted from eukaryotic cells using the Escherichia coli biotin protein ligase. In one system the biotin ligase was co-secreted from the cells along with substrate protein enabling extracellular biotinylation of the tagged protein. In the other system, biotin ligase was engineered to be retained in the endoplasmic reticulum (ER) and metabolically biotinylates the secretory protein as it passes through the ER. An engineered antibody fragment, a diabody with specificity for carcinoembryonic antigen (CEA) was fused to the biotin acceptor domain (123 amino acid) of Propionibacterium shermanii. Coexpression of the fusion protein with ER retained biotin ligase showed higher biotinylation efficiency than biotinylation by co-secreted ligase. Biotinylation of the anti-CEA diabody tagged with a short (15 amino acid, Biotin Avitag) biotin acceptor peptide was also successful. Utilization of ER retained biotin ligase for biotinylation of protein is an attractive alternative for efficiently producing uniformly biotinylated recombinant proteins for a variety of avidin-biotin technologies.
Collapse
Affiliation(s)
- Bhaswati Barat
- Crump Institute for Molecular Imaging, Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, 700 Westwood Plaza, Los Angeles, CA 90095
| | - Anna M. Wu
- Crump Institute for Molecular Imaging, Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, 700 Westwood Plaza, Los Angeles, CA 90095
| |
Collapse
|
36
|
Guinn BA, Norris JS, Farzaneh F, Deisseroth AB. International Society for Cell and Gene Therapy of Cancer: 2005 meeting in Shenzhen, China. Cancer Gene Ther 2006; 14:128-38. [PMID: 17041563 DOI: 10.1038/sj.cgt.7700996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The 2005 International Society for Cell and Gene Therapy of Cancer (ISCGT) Congress was held in Shenzhen, China (www.iscgtchina2005.com) from December 9th-11th 2005. Here, we describe a representation of the most seminal presentations providing an overview of the progress in the field of cancer gene therapy including the successful introduction of the first approved gene therapy drug.
Collapse
Affiliation(s)
- B A Guinn
- King's College London School of Medicine, Department of Haematological Medicine, The Rayne Institute, 123 Coldharbour Lane, London, UK.
| | | | | | | |
Collapse
|
37
|
Chan L, Hardwick NR, Guinn BA, Darling D, Gäken J, Galea-Lauri J, Ho AY, Mufti GJ, Farzaneh F. An immune edited tumour versus a tumour edited immune system: Prospects for immune therapy of acute myeloid leukaemia. Cancer Immunol Immunother 2006; 55:1017-24. [PMID: 16450142 PMCID: PMC11030980 DOI: 10.1007/s00262-006-0129-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2005] [Accepted: 01/04/2006] [Indexed: 10/25/2022]
Abstract
Cell based therapies for acute myeloid leukaemia (AML) have made significant progress in the last decade benefiting the prognosis and survival of patients with this aggressive form of leukaemia. Due to advances in haematopoietic stem cell transplantation (HSCT) and particularly the advent of reduced intensity conditioning (RIC), the scope of transplantation has now extended to those patients previously ineligible due to age and health restrictions and has been associated with a decrease in transplant related mortality. The apparent graft versus leukaemia (GvL) effect observed following HSCT demonstrates the potential of the immune system to target and eradicate AML cells. Building on previously published pre-clinical studies by ourselves and others, we are now initiating a Phase I clinical study in which lentiviral vectors are used to genetically modify AML cells to express B7.1 (CD80) and IL-2. By combining allogeneic HSCT with immunisation, using the autologous AML cells expressing B7.1 and IL-2, we hope to stimulate immune eradication of residual AML cells in poor prognosis patients that have achieved donor chimerism. In this report we describe the background to cell therapy based approaches for AML, and discuss difficulties associated with the deployment of a chronically stimulated, hence exhausted/depleted immune system to eradicate tumour cells that have already escaped immune surveillance.
Collapse
Affiliation(s)
- Lucas Chan
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Nicola R. Hardwick
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Barbara-ann Guinn
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Dave Darling
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Joop Gäken
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Joanna Galea-Lauri
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Aloysius Y. Ho
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Ghulam J. Mufti
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Farzin Farzaneh
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| |
Collapse
|