1
|
Tridgett M, Mulet M, Johny SP, Ababi M, Raghunath M, Fustinoni C, Galabova B, Fernández-Díaz C, Mikalajūnaitė I, Tomás HA, Kucej M, Dunajová L, Zgrundo Z, Page E, McCall L, Parker-Manuel R, Payne T, Peckett M, Kent J, Holland L, Asatryan R, Montgomery L, Chow TL, Beveridge R, Salkauskaite I, Alam MT, Hollard D, Dowding S, Gabriel HB, Branciaroli C, Cawood R, Valenti W, Chang D, Patrício MI, Liu Q. Lentiviral vector packaging and producer cell lines yield titers equivalent to the industry-standard four-plasmid process. Mol Ther Methods Clin Dev 2024; 32:101315. [PMID: 39282073 PMCID: PMC11401174 DOI: 10.1016/j.omtm.2024.101315] [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: 02/13/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024]
Abstract
Lentiviral vector (LVV)-mediated cell and gene therapies have the potential to cure diseases that currently require lifelong intervention. However, the requirement for plasmid transfection hinders large-scale LVV manufacture. Moreover, large-scale plasmid production, testing, and transfection contribute to operational risk and the high cost associated with this therapeutic modality. Thus, we developed LVV packaging and producer cell lines, which reduce or eliminate the need for plasmid transfection during LVV manufacture. To develop a packaging cell line, lentiviral packaging genes were stably integrated by random integration of linearized plasmid DNA. Then, to develop EGFP- and anti-CD19 chimeric antigen receptor-encoding producer cell lines, transfer plasmids were integrated by transposase-mediated integration. Single-cell isolation and testing were performed to isolate the top-performing clonal packaging and producer cell lines. Production of LVVs that encode various cargo genes revealed consistency in the production performance of the packaging and producer cell lines compared to the industry-standard four-plasmid transfection method. By reducing or eliminating the requirement for plasmid transfection, while achieving production performance consistent with the current industry standard, the packaging and producer cell lines developed here can reduce costs and operational risks of LVV manufacture, thus increasing patient access to LVV-mediated cell and gene therapies.
Collapse
Affiliation(s)
- Matthew Tridgett
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Marie Mulet
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Sherin Parokkaran Johny
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Maria Ababi
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Meenakshi Raghunath
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Chloé Fustinoni
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Boryana Galabova
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Cristina Fernández-Díaz
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Iveta Mikalajūnaitė
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Hélio A Tomás
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Marek Kucej
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Lucia Dunajová
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Zofia Zgrundo
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Emma Page
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Lorna McCall
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Richard Parker-Manuel
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Tom Payne
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Matthew Peckett
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Jade Kent
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Louise Holland
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Robert Asatryan
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Louise Montgomery
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Tsz Lung Chow
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Ryan Beveridge
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Ieva Salkauskaite
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Mohine T Alam
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Daniel Hollard
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Sarah Dowding
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Heloísa Berti Gabriel
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Corinne Branciaroli
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Ryan Cawood
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Weimin Valenti
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
- WuXi Advanced Therapies, 4701 League Island Blvd, Philadelphia, PA 19112, USA
| | - David Chang
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
- WuXi Advanced Therapies, 4701 League Island Blvd, Philadelphia, PA 19112, USA
| | - Maria I Patrício
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| | - Qian Liu
- OXGENE, A WuXi Advanced Therapies Company, Medawar Centre, Robert Robinson Avenue, Oxford, Oxfordshire OX4 4HG, UK
| |
Collapse
|
2
|
Srivastava S, Singh S, Singh A. Augmenting the landscape of chimeric antigen receptor T-cell therapy. Expert Rev Anticancer Ther 2024; 24:755-773. [PMID: 38912754 DOI: 10.1080/14737140.2024.2372330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION The inception of recombinant DNA technology and live cell genomic alteration have paved the path for the excellence of cell and gene therapies and often provided the first curative treatment for many indications. The approval of the first Chimeric Antigen Receptor (CAR) T-cell therapy was one of the breakthrough innovations that became the headline in 2017. Currently, the therapy is primarily restricted to a few nations, and the market is growing at a CAGR (current annual growth rate) of 11.6% (2022-2032), as opposed to the established bio-therapeutic market at a CAGR of 15.9% (2023-2030). The limited technology democratization is attributed to its autologous nature, lack of awareness, therapy inclusion criteria, high infrastructure cost, trained personnel, complex manufacturing processes, regulatory challenges, recurrence of the disease, and long-term follow-ups. AREAS COVERED This review discusses the vision and strategies focusing on the CAR T-cell therapy democratization with mitigation plans. Further, it also covers the strategies to leverage the mRNA-based CAR T platform for building an ecosystem to ensure availability, accessibility, and affordability to the community. EXPERT OPINION mRNA-guided CAR T cell therapy is a rapidly growing area wherein a collaborative approach among the stakeholders is needed for its success.
Collapse
Affiliation(s)
| | - Sanjay Singh
- mRNA Department, Gennova Biopharmaceuticals Ltd. ITBT Park, Pune, India
| | - Ajay Singh
- mRNA Department, Gennova Biopharmaceuticals Ltd. ITBT Park, Pune, India
| |
Collapse
|
3
|
Metanat Y, Viktor P, Amajd A, Kaur I, Hamed AM, Abed Al-Abadi NK, Alwan NH, Chaitanya MVNL, Lakshmaiya N, Ghildiyal P, Khalaf OM, Ciongradi CI, Sârbu I. The paths toward non-viral CAR-T cell manufacturing: A comprehensive review of state-of-the-art methods. Life Sci 2024; 348:122683. [PMID: 38702027 DOI: 10.1016/j.lfs.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/11/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Although CAR-T cell therapy has emerged as a game-changer in cancer immunotherapy several bottlenecks limit its widespread use as a front-line therapy. Current protocols for the production of CAR-T cells rely mainly on the use of lentiviral/retroviral vectors. Nevertheless, according to the safety concerns around the use of viral vectors, there are several regulatory hurdles to their clinical use. Large-scale production of viral vectors under "Current Good Manufacturing Practice" (cGMP) involves rigorous quality control assessments and regulatory requirements that impose exorbitant costs on suppliers and as a result, lead to a significant increase in the cost of treatment. Pursuing an efficient non-viral method for genetic modification of immune cells is a hot topic in cell-based gene therapy. This study aims to investigate the current state-of-the-art in non-viral methods of CAR-T cell manufacturing. In the first part of this study, after reviewing the advantages and disadvantages of the clinical use of viral vectors, different non-viral vectors and the path of their clinical translation are discussed. These vectors include transposons (sleeping beauty, piggyBac, Tol2, and Tc Buster), programmable nucleases (ZFNs, TALENs, and CRISPR/Cas9), mRNA, plasmids, minicircles, and nanoplasmids. Afterward, various methods for efficient delivery of non-viral vectors into the cells are reviewed.
Collapse
Affiliation(s)
- Yekta Metanat
- Faculty of Medicine, Zahedan University of Medical Sciences, Sistan and Baluchestan Province, Iran
| | - Patrik Viktor
- Óbuda University, Karoly Keleti faculty, Tavaszmező u. 15-17, H-1084 Budapest, Hungary
| | - Ayesha Amajd
- Faculty of Transport and Aviation Engineering, Silesian University of Technology, Krasińskiego 8 Street, 40-019 Katowice, Poland
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bangalore, Karnataka, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | | | | | | | - M V N L Chaitanya
- School of pharmaceutical sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab - 144411, India
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Carmen Iulia Ciongradi
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Ioan Sârbu
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| |
Collapse
|
4
|
Williams-Fegredo T, Davies L, Knevelman C, Mitrophanous K, Miskin J, Rafiq QA. Degradation of specific glycosaminoglycans improves transfection efficiency and vector production in transient lentiviral vector manufacturing processes. Front Bioeng Biotechnol 2024; 12:1409203. [PMID: 38994127 PMCID: PMC11238175 DOI: 10.3389/fbioe.2024.1409203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Both cell surface and soluble extracellular glycosaminoglycans have been shown to interfere with the exogenous nucleic acid delivery efficiency of non-viral gene delivery, including lipoplex and polyplex-mediated transfection. Most gene therapy viral vectors used commercially and in clinical trials are currently manufactured using transient transfection-based bioprocesses. The growing demand for viral vector products, coupled with a global shortage in production capability, requires improved transfection technologies and processes to maximise process efficiency and productivity. Soluble extracellular glycosaminoglycans were found to accumulate in the conditioned cell culture medium of suspension adapted HEK293T cell cultures, compromising transfection performance and lentiviral vector production. The enzymatic degradation of specific, chondroitin sulphate-based, glycosaminoglycans with chondroitinase ABC was found to significantly enhance transfection performance. Additionally, we report significant improvements in functional lentiviral vector titre when cultivating cells at higher cell densities than those utilised in a control lentiviral vector bioprocess; an improvement that was further enhanced when cultures were supplemented with chondroitinase ABC prior to transfection. A 71.2% increase in functional lentiviral vector titre was calculated when doubling the cell density prior to transfection compared to the existing process and treatment of the high-density cell cultures with 0.1 U/mL chondroitinase ABC resulted in a further 18.6% increase in titre, presenting a method that can effectively enhance transfection performance.
Collapse
Affiliation(s)
- Thomas Williams-Fegredo
- Oxford Biomedica (UK) Limited, Oxford, United Kingdom
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Lee Davies
- Oxford Biomedica (UK) Limited, Oxford, United Kingdom
| | | | | | - James Miskin
- Oxford Biomedica (UK) Limited, Oxford, United Kingdom
| | - Qasim A. Rafiq
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
5
|
Mier NC, Roper DK. Effects of an indole derivative on cell proliferation, transfection, and alternative splicing in production of lentiviral vectors by transient co-transfection. PLoS One 2024; 19:e0297817. [PMID: 38833479 PMCID: PMC11149887 DOI: 10.1371/journal.pone.0297817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/12/2024] [Indexed: 06/06/2024] Open
Abstract
Lentiviral vectors derived from human immunodeficiency virus type I are widely used to deliver functional gene copies to mammalian cells for research and gene therapies. Post-transcriptional splicing of lentiviral vector transgene in transduced host and transfected producer cells presents barriers to widespread application of lentiviral vector-based therapies. The present study examined effects of indole derivative compound IDC16 on splicing of lentiviral vector transcripts in producer cells and corresponding yield of infectious lentiviral vectors. Indole IDC16 was shown previously to modify alternative splicing in human immunodeficiency virus type I. Human embryonic kidney 293T cells were transiently transfected by 3rd generation backbone and packaging plasmids using polyethyleneimine. Reverse transcription-quantitative polymerase chain reaction of the fraction of unspliced genomes in human embryonic kidney 293T cells increased up to 31% upon the indole's treatment at 2.5 uM. Corresponding yield of infectious lentiviral vectors decreased up to 4.5-fold in a cell transduction assay. Adjusting timing and duration of IDC16 treatment indicated that the indole's disruption of early stages of transfection and cell cycle had a greater effect on exponential time course of lentiviral vector production than its reduction of post-transcriptional splicing. Decrease in transfected human embryonic kidney 293T proliferation by IDC16 became significant at 10 uM. These findings indicated contributions by early-stage transfection, cell proliferation, and post-transcriptional splicing in transient transfection of human embryonic kidney 293T cells for lentiviral vector production.
Collapse
Affiliation(s)
- Nataly Carolina Mier
- Department of Biological Engineering, Utah State University, Logan, Utah, United States of America
| | - Donald Keith Roper
- Department of Biological Engineering, Utah State University, Logan, Utah, United States of America
| |
Collapse
|
6
|
Suleman S, Fawaz S, Roberts T, Ellison S, Bigger B, Themis M. Optimised protocols to generate high titre lentiviral vectors using a novel transfection agent enabling extended HEK293T culture following transient transfection and suspension culture. J Virol Methods 2024; 325:114884. [PMID: 38218417 DOI: 10.1016/j.jviromet.2024.114884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
HIV-1 based lentiviral viruses are considered powerful and versatile gene therapy vectors to deliver therapeutic genes to patients with hereditary or acquired diseases. These vectors can efficiently transduce a variety of cell types when dividing or non-dividing to provide permanent delivery and long-term gene expression. Demand for scalable manufacturing protocols able to generate enough high titre vector for widespread use of this technology is increasing and considerable efforts to improve vector production cost-effectively, is ongoing. Current methods for LV production mainly use transient transfection of producer cell lines. Cells can be grown at scale, either in 2D relying on culturing producer cells in multi-tray flask cell culture factories or in roller bottles or can be adapted to grow in 3D suspensions in large batch fermenters. This suits rapid production and testing of new vector constructs pre-clinically for their efficacy, particle titre and safety. In this study, we sought to improve lentiviral titre over time by testing two alternative commercially available transfection reagents Fugene® 6 and Genejuice® with the commonly used polycation, polyethyleneimine. Our aim was to identify less cytotoxic transfection reagents that could be used to generate LV particles at high titre past the often used 72 h period when vector is usually collected before producer cell death is caused due to post transfection cytotoxicity. We show that LV could be produced in extended culture using Genejuice® and even by transfected cells in glass flasks in suspension. Because this delivery agent is less toxic to 293 T producer cells, following optimisation of transfection we found that LV can be harvested for more than 10 days at high titre. Using our protocol, titres of 109 TU/ml and 108 TU/ml were routinely reached via traditional monolayer conditions or suspension cultures, respectively. We propose, this simple change in vector production enables large volumes of high titre vector to be produced, cost effectively for non-clinical in vivo and in vitro applications or for more stringent downstream clinical grade vector purification.
Collapse
Affiliation(s)
- Saqlain Suleman
- Department of Life Sciences, College of Health, Medicine & Life Sciences, Brunel University London, Uxbridge, UK
| | - Serena Fawaz
- Department of Life Sciences, College of Health, Medicine & Life Sciences, Brunel University London, Uxbridge, UK
| | - Terry Roberts
- Department of Life Sciences, College of Health, Medicine & Life Sciences, Brunel University London, Uxbridge, UK
| | - Stuart Ellison
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Brian Bigger
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Michael Themis
- Department of Life Sciences, College of Health, Medicine & Life Sciences, Brunel University London, Uxbridge, UK; Division of Ecology and Evolution, Department of Life Sciences, Imperial College London, London, UK; Testavec Ltd, Queensgate House, Maidenhead, UK.
| |
Collapse
|
7
|
Todesco HM, Gafuik C, John CM, Roberts EL, Borys BS, Pawluk A, Kallos MS, Potts KG, Mahoney DJ. High-titer manufacturing of SARS-CoV-2 Spike-pseudotyped VSV in stirred-tank bioreactors. Mol Ther Methods Clin Dev 2024; 32:101189. [PMID: 38327804 PMCID: PMC10847022 DOI: 10.1016/j.omtm.2024.101189] [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: 04/26/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic highlighted the importance of vaccine innovation in public health. Hundreds of vaccines built on numerous technology platforms have been rapidly developed against SARS-CoV-2 since 2020. Like all vaccine platforms, an important bottleneck to viral-vectored vaccine development is manufacturing. Here, we describe a scalable manufacturing protocol for replication-competent SARS-CoV-2 Spike-pseudotyped vesicular stomatitis virus (S-VSV)-vectored vaccines using Vero cells grown on microcarriers in a stirred-tank bioreactor. Using Cytodex 1 microcarriers over 6 days of fed-batch culture, Vero cells grew to a density of 3.95 ± 0.42 ×106 cells/mL in 1-L stirred-tank bioreactors. Ancestral strain S-VSV reached a peak titer of 2.05 ± 0.58 ×108 plaque-forming units (PFUs)/mL at 3 days postinfection. When compared to growth in plate-based cultures, this was a 29-fold increase in virus production, meaning a 1-L bioreactor produces the same amount of virus as 1,284 plates of 15 cm. In addition, the omicron BA.1 S-VSV reached a peak titer of 5.58 ± 0.35 × 106 PFU/mL. Quality control testing showed plate- and bioreactor-produced S-VSV had similar particle-to-PFU ratios and elicited comparable levels of neutralizing antibodies in immunized hamsters. This method should enhance preclinical and clinical development of pseudotyped VSV-vectored vaccines in future pandemics.
Collapse
Affiliation(s)
- Hayley M. Todesco
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Chris Gafuik
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cini M. John
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Erin L. Roberts
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Breanna S. Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Alexis Pawluk
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Michael S. Kallos
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Kyle G. Potts
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Douglas J. Mahoney
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
8
|
Mayani M, Nellimarla S, Mangalathillam R, Rao H, Patarroyo-White S, Ma J, Figueroa B. Depth filtration for clarification of intensified lentiviral vector suspension cell culture. Biotechnol Prog 2024; 40:e3409. [PMID: 37985144 DOI: 10.1002/btpr.3409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Depth filtration significantly impacts efficiency of lentiviral (LV) vector purification process. However, it is often deprioritized in the overall scope of viral vector manufacturing process optimization. The demand for LV vectors has increased with the rise in disease indications, making it crucial to improve current manufacturing processes. Upstream bioreactor process intensification has enabled cell densities of over 107 viable cells/mL, creating challenges for harvest unit operations. The larger size of LV vectors and their physiochemical similarity to host cell-DNA (HC-DNA) and poor clarification performance causes significant challenges for the subsequent chromatography-based purifications. As a result, a robust and scalable harvest of LV process is needed, especially for LV in vivo therapeutic quality needs. In this study, we systematically evaluated the overlooked yet important issue of depth filtration systems to improve enveloped LV functional vector recovery. We found that an established depth filtration system in process A that provided 94% (n = 6) LV functional recovery could not be translated to intensified Process B cell culture. Hence, the depth filtration process became a bottleneck for the purification performance in an intensified process. We demonstrated an improvement in LV functional vector recovery from 34% to 82% via filter train optimization for an intensified suspension cell culture system (>107 cells/mL with higher titer), while still maintaining a loading throughput of ≥82 L/m2 and turbidity ≤20 NTU. It was demonstrated that the two or three-stage depth filtration scheme is scalable and more suitable for high cell density culture for large scale for LV manufacturing process.
Collapse
Affiliation(s)
- Mukesh Mayani
- Genomic Medicine Unit CMC, Global CMC Development, Waltham, Massachusetts, USA
- Process and Analytical Development (PAD), Gene Therapy Franchise, National Resilience Inc., Waltham, Massachusetts, USA
| | - Srinivas Nellimarla
- Genomic Medicine Unit CMC, Global CMC Development, Waltham, Massachusetts, USA
| | | | - Hema Rao
- Genomic Medicine Unit CMC, Global CMC Development, Waltham, Massachusetts, USA
| | | | - Junfen Ma
- Genomic Medicine Unit CMC, Global CMC Development, Waltham, Massachusetts, USA
| | - Bruno Figueroa
- Genomic Medicine Unit CMC, Global CMC Development, Waltham, Massachusetts, USA
| |
Collapse
|
9
|
Vaz TA, Rodrigues AF, Coroadinha AS. Exploring nutrient supplementation and bioprocess optimization to improve the production of lentiviral vectors in serum-free medium suspension cultures. Biotechnol J 2024; 19:e2300212. [PMID: 37903159 DOI: 10.1002/biot.202300212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023]
Abstract
The use of lentiviral vectors (LV) in gene therapy has been growing in recent years. To meet the increasing clinical demand, LV production platforms will benefit from improved productivity and scalability to enable cost-effective manufacture of LV-based therapies. Here we report the adaptation of 293T cells to serum-free suspension cultures and the improvement of LV yields through transfection parameters optimization, process intensification and medium supplementation with nutrient boosters. Cells were sequentially adapted to different serum-free culture media, transfection parameters were optimized and the two best-performing conditions were selected to explore process intensification by increasing cell density at the time of transfection. LV production at higher cell densities increased volumetric titers up to 12-fold and lipid supplementation was the most efficient metabolic optimization strategy further enhancing LV productivity by 3-fold. Furthermore, cell concentration was identified and validated as an important source of transfection variability impairing cellular uptake of DNA polyplexes, impacting transfection efficiency and reducing LV titers down to 6-fold. This work contributes to improving LV-based gene therapy by establishing new scalable manufacturing platforms and providing key metabolic insights, unveiling important bioreaction parameters to improve vector yields.
Collapse
Affiliation(s)
- Tiago A Vaz
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana F Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- LASIGE, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Ana S Coroadinha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
10
|
Ou J, Tang Y, Xu J, Tucci J, Borys MC, Khetan A. Recent advances in upstream process development for production of recombinant adeno-associated virus. Biotechnol Bioeng 2024; 121:53-70. [PMID: 37691172 DOI: 10.1002/bit.28545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/17/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Recombinant adeno-associated virus (rAAV) is rapidly emerging as the preferred delivery vehicle for gene therapies, with promising advantages in safety and efficacy. Key challenges in systemic in-vivo rAAV gene therapy applications are the gap in production capabilities versus potential market demand and complex production process. This review summarizes current available information on rAAV upstream manufacturing processes and proposed optimizations for production. The advancements in rAAV production media were reviewed with proposals to speed up the cell culture process development. Furthermore, major methods for genetic element delivery to host cells were summarized with their advantages, limitations, and future directions for optimization. In addition, culture vessel selection criteria were listed based on production cell system, scale, and development stage. Process control at the production step was also outlined with an in-depth understanding of production kinetics and quality control.
Collapse
Affiliation(s)
- Jianfa Ou
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Yawen Tang
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Jianlin Xu
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Julian Tucci
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Michael C Borys
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Anurag Khetan
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| |
Collapse
|
11
|
Triantafyllou N, Sarkis M, Krassakopoulou A, Shah N, Papathanasiou MM, Kontoravdi C. Uncertainty quantification for gene delivery methods: A roadmap for pDNA manufacturing from phase I clinical trials to commercialization. Biotechnol J 2024; 19:e2300103. [PMID: 37797343 DOI: 10.1002/biot.202300103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/01/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023]
Abstract
The fast-growing interest in cell and gene therapy (C>) products has led to a growing demand for the production of plasmid DNA (pDNA) and viral vectors for clinical and commercial use. Manufacturers, regulators, and suppliers need to develop strategies for establishing robust and agile supply chains in the otherwise empirical field of C>. A model-based methodology that has great potential to support the wider adoption of C> is presented, by ensuring efficient timelines, scalability, and cost-effectiveness in the production of key raw materials. Specifically, key process and economic parameters are identified for (1) the production of pDNA for the forward-looking scenario of non-viral-based Chimeric Antigen Receptor (CAR) T-cell therapies from clinical (200 doses) to commercial (40,000 doses) scale and (2) the commercial (40,000 doses) production of pDNA and lentiviral vectors for the current state-of-the-art viral vector-based CAR T-cell therapies. By applying a systematic global sensitivity analysis, we quantify uncertainty in the manufacturing process and apportion it to key process and economic parameters, highlighting cost drivers and limitations that steer decision-making. The results underline the cost-efficiency and operational flexibility of non-viral-based therapies in the overall C> supply chain, as well as the importance of economies-of-scale in the production of pDNA.
Collapse
Affiliation(s)
- Niki Triantafyllou
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Miriam Sarkis
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | | | - Nilay Shah
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Maria M Papathanasiou
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Cleo Kontoravdi
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| |
Collapse
|
12
|
Chua R, Ghosh S. An optimized method for gene knockdown in differentiating human and mouse adipocyte cultures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571780. [PMID: 38168248 PMCID: PMC10760114 DOI: 10.1101/2023.12.14.571780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Adipocyte cultures are a mainstay of metabolic disease research, yet loss-of-function studies in differentiating adipocytes is complicated by the refractoriness of lipid-containing adipocytes to standard siRNA transfections. Alternative methods, such as electroporation or adenovirus/lentivirus-based delivery systems are complex, expensive and often accompanied with unacceptable levels of cell death. To address this problem, we have tested two commercially available siRNA delivery systems in this study using a multi-parameter optimization approach. Our results identified a uniform siRNA transfection protocol that can be applied to human and mouse adipocyte cultures throughout the time course of differentiation, beginning with pre-differentiated cells and continuing up to lipid-accumulated differentiated adipocytes. Our findings allow for efficient transfection of human and mouse adipocyte cultures using standard and readily available methodologies, and should help significantly expand the scope of gene manipulation studies in these cell types.
Collapse
Affiliation(s)
- Ruiming Chua
- Program in Cardiovascular and Metabolic Diseases, Duke-NUS Medical School, Singapore
| | - Sujoy Ghosh
- Program in Cardiovascular and Metabolic Diseases, Duke-NUS Medical School, Singapore
- Laboratory of Computational Biology, Pennington Biomedical Research Center, LA, USA
| |
Collapse
|
13
|
Hein MD, Kazenmaier D, van Heuvel Y, Dogra T, Cattaneo M, Kupke SY, Stitz J, Genzel Y, Reichl U. Production of retroviral vectors in continuous high cell density culture. Appl Microbiol Biotechnol 2023; 107:5947-5961. [PMID: 37542575 PMCID: PMC10485120 DOI: 10.1007/s00253-023-12689-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 08/07/2023]
Abstract
Retroviral vectors derived from murine leukemia virus (MLV) are used in somatic gene therapy applications e.g. for genetic modification of hematopoietic stem cells. Recently, we reported on the establishment of a suspension viral packaging cell line (VPC) for the production of MLV vectors. Human embryonic kidney 293-F (HEK293-F) cells were genetically modified for this purpose using transposon vector technology. Here, we demonstrate the establishment of a continuous high cell density (HCD) process using this cell line. First, we compared different media regarding the maximum achievable viable cell concentration (VCC) in small scale. Next, we transferred this process to a stirred tank bioreactor before we applied intensification strategies. Specifically, we established a perfusion process using an alternating tangential flow filtration system. Here, VCCs up to 27.4E + 06 cells/mL and MLV vector titers up to 8.6E + 06 transducing units/mL were achieved. Finally, we established a continuous HCD process using a tubular membrane for cell retention and continuous viral vector harvesting. Here, the space-time yield was 18-fold higher compared to the respective batch cultivations. Overall, our results clearly demonstrate the feasibility of HCD cultivations for high yield production of viral vectors, especially when combined with continuous viral vector harvesting. KEY POINTS: • A continuous high cell density process for MLV vector production was established • The tubular cell retention membrane allowed for continuous vector harvesting • The established process had a 18-fold higher space time yield compared to a batch.
Collapse
Affiliation(s)
- Marc D Hein
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Daniel Kazenmaier
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Faculty of Biotechnology, University of Applied Sciences Mannheim, Mannheim, Germany
| | - Yasemin van Heuvel
- Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Leverkusen, Germany
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Tanya Dogra
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | | | - Sascha Y Kupke
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Jörn Stitz
- Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Leverkusen, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.
| | - Udo Reichl
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| |
Collapse
|
14
|
Fiol CR, Collignon ML, Welsh J, Rafiq QA. Optimizing and developing a scalable, chemically defined, animal component-free lentiviral vector production process in a fixed-bed bioreactor. Mol Ther Methods Clin Dev 2023; 30:221-234. [PMID: 37528866 PMCID: PMC10388200 DOI: 10.1016/j.omtm.2023.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
Lentiviral vectors (LVVs) play a critical role in gene delivery for ex vivo gene-modified cell therapies. However, the lack of scalable LVV production methods and the high cost associated with them may limit their use. In this work, we demonstrate the optimization and development of a scalable, chemically defined, animal component-free LVV production process using adherent human embryonic kidney 293T cells in a fixed-bed bioreactor. The initial studies focused on the optimization of the culture process in 2D static cultures. Process changes such as decreasing cell seeding density on day 0 from 2.5 × 104 to 5 × 103 cells/cm2, delaying the transient transfection from 24 to 120 h post-seeding, reducing plasmid DNA to 167 ng/cm2, and adding 5 mM sodium butyrate 6 h post-transfection improved functional LVV titers by 26.9-fold. The optimized animal component-free production process was then transferred to the iCELLis Nano bioreactor, a fixed-bed bioreactor, where titers of 1.2 × 106 TU/cm2 were achieved when it was operated in perfusion. In this work, comparable functional LVV titers were obtained with FreeStyle 293 Expression medium and the conventional Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum both at small and large scale.
Collapse
Affiliation(s)
- Carme Ripoll Fiol
- Department of Biochemical Engineering, University College London, Gower Street, WC1E 6BT London, UK
| | - Marie-Laure Collignon
- Department of Scientific and Laboratory Services (SLS), Pall Corporation, Reugelstraat 2, 3320 Hoegaarden, Belgium
| | - John Welsh
- Department of Research and Development (R&D), Pall Corporation, 5 Harbourgate Business Park, Southampton Road, PO6 4BQ Portsmouth, UK
| | - Qasim A. Rafiq
- Department of Biochemical Engineering, University College London, Gower Street, WC1E 6BT London, UK
| |
Collapse
|
15
|
Broussau S, Lytvyn V, Simoneau M, Guilbault C, Leclerc M, Nazemi-Moghaddam N, Coulombe N, Elahi SM, McComb S, Gilbert R. Packaging cells for lentiviral vectors generated using the cumate and coumermycin gene induction systems and nanowell single-cell cloning. Mol Ther Methods Clin Dev 2023; 29:40-57. [PMID: 36936448 PMCID: PMC10018046 DOI: 10.1016/j.omtm.2023.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Lentiviral vectors (LVs) are important for cell therapy because of their capacity to stably modify the genome after integration. This study describes a novel and relatively simple approach to generate packaging cells and producer clones for self-inactivating (SIN) LVs pseudotyped with the vesicular stomatitis virus glycoprotein (VSV-G). A novel gene regulation system, based on the combination of the cumate and coumermycin induction systems, was developed to ensure tight control for the expression of cytotoxic packaging elements. To accelerate clone isolation and ensure monoclonality, the packaging genes were transfected simultaneously into human embryonic kidney cells (293SF-3F6) previously engineered with the induction system, and clones were isolated after limiting dilution into nanowell arrays using a robotic cell picking instrument with scanning capability. The method's effectiveness to isolate colonies derived from single cells was demonstrated using mixed populations of cells labeled with two different fluorescent markers. Because the recipient cell line grew in suspension culture, and all the procedures were performed without serum, the resulting clones were readily adaptable to serum-free suspension culture. The best producer clone produced LVs expressing GFP at a titer of 2.3 × 108 transduction units (TU)/mL in the culture medium under batch mode without concentration.
Collapse
Affiliation(s)
- Sophie Broussau
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Viktoria Lytvyn
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Mélanie Simoneau
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Claire Guilbault
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Mélanie Leclerc
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Nazila Nazemi-Moghaddam
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Nathalie Coulombe
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Seyyed Mehdy Elahi
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
| | - Scott McComb
- Department of Immunology, Human Health Therapeutics Research Centre, National Research Council, Canada, Ottawa, ON K1A 0R6, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rénald Gilbert
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC H4P 2R2, Canada
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
- Département de Génie chimique, Université Laval, Québec, QC G1V 0A6, Canada
- Corresponding author: Rénald Gilbert, National Research Council Canada, Building Montreal, 6100 Avenue Royalmount, Montreal, QC H4P 2R2, Canada.
| |
Collapse
|
16
|
Syzdykova L, Zauatbayeva G, Keyer V, Ramanculov Y, Arsienko R, Shustov AV. Process for production of chimeric antigen receptor-transducing lentivirus particles using infection with replicon particles containing self-replicating RNAs. Biochem Eng J 2023. [DOI: 10.1016/j.bej.2023.108814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
17
|
Nardo D, Pitts MG, Kaur R, Venditto VJ. In vivo assessment of triazine lipid nanoparticles as transfection agents for plasmid DNA. Biomater Sci 2022; 10:6968-6979. [PMID: 36222485 PMCID: PMC9729407 DOI: 10.1039/d2bm01289h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-viral vectors for in vivo delivery of plasmid DNA rely on optimized formulations to achieve robust transgene expression. Several cationic lipids have been developed to deliver nucleic acids, but most recent literature has focused on mRNA due to its increased expression profile and excluded plasmid DNA, which may have the advantage of being less immunogenic. In this study, we describe the in vivo evaluation of cationic triazine based lipids, previously prepared by our group. We identify one lipid with limited in vivo toxicity for studies to optimize the lipid formulations, which include an evaluation of the influence of PEG and helper lipids on transgene expression. We then demonstrate that lipoplexes, but not lipid nanoparticles, formed from triazine lipids achieve similar transgene expression levels as AAV vectors and offer enhanced expression as compared to a commercially available cationic lipid, DOTAP. Importantly, the lipid nanoparticles and lipoplexes induce minimal antibody profiles toward the expressed protein, while serving as a platform to induce robust antibody responses when directly delivering the protein. Collectively, these data demonstrate the potential for triazine based lipids as non-viral vectors for gene delivery, and highlights the need to optimize each formulation based on the exact contents to achieve enhanced transgene expression with plasmid DNA constructs.
Collapse
Affiliation(s)
- David Nardo
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| | - Michelle G Pitts
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| | - Rupinder Kaur
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| | - Vincent J Venditto
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| |
Collapse
|
18
|
Lavado-García J, Pérez-Rubio P, Cervera L, Gòdia F. The cell density effect in animal cell-based bioprocessing: Questions, insights and perspectives. Biotechnol Adv 2022; 60:108017. [PMID: 35809763 DOI: 10.1016/j.biotechadv.2022.108017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/31/2022] [Accepted: 07/01/2022] [Indexed: 11/28/2022]
Abstract
One of the main challenges in the development of bioprocesses based on cell transient expression is the commonly reported reduction of cell specific productivity at increasing cell densities. This is generally known as the cell density effect (CDE). Many efforts have been devoted to understanding the cell metabolic implications to this phenomenon in an attempt to design operational strategies to overcome it. A comprehensive analysis of the main studies regarding the CDE is provided in this work to better define the elements comprising its cause and impact. Then, examples of methodologies and approaches employed to achieve successful transient expression at high cell densities (HCD) are thoroughly reviewed. A critical assessment of the limitations of the reported studies in the understanding of the CDE is presented, covering the leading hypothesis of the molecular implications. The overall analysis of previous work on CDE may offer useful insights for further research into manufacturing of biologics.
Collapse
Affiliation(s)
- Jesús Lavado-García
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Pol Pérez-Rubio
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Laura Cervera
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Francesc Gòdia
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| |
Collapse
|
19
|
Tran MY, Kamen AA. Production of Lentiviral Vectors Using a HEK-293 Producer Cell Line and Advanced Perfusion Processing. Front Bioeng Biotechnol 2022; 10:887716. [PMID: 35774066 PMCID: PMC9237754 DOI: 10.3389/fbioe.2022.887716] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/24/2022] [Indexed: 12/03/2022] Open
Abstract
The field of lentiviral vector (LV) production continues to face challenges in large-scale manufacturing, specifically regarding producing enough vectors to meet the demand for treating patients as well as producing high and consistent quality of vectors for efficient dosing. Two areas of interest are the use of stable producer cell lines, which facilitates the scalability of LV production processes as well as making the process more reproducible and robust for clinical applications, and the search of a cell retention device scalable to industrial-size bioreactors. This manuscript investigates a stable producer cell line for producing LVs with GFP as the transgene at shake flask scale and demonstrates LV production at 3L bioreactor scale using the Tangential Flow Depth Filtration (TFDF) as a cell retention device in perfusion mode. Cumulative functional yields of 3.3 x 1011 and 3.9 x 1011 transducing units were achieved; the former over 6 days of LV production with 16.3 L of perfused media and the latter over 4 days with 16 L. In comparing to a previously published value that was achieved using the same stable producer cell line and the acoustic filter as the perfusion device at the same bioreactor scale, the TFDF perfusion run produced 1.5-fold higher cumulative functional yield. Given its scale-up potential, the TFDF is an excellent candidate to be further evaluated to determine optimized conditions that can ultimately support continuous manufacturing of LVs at large scale.
Collapse
|
20
|
Yadav M, Atala A, Lu B. Developing all-in-one virus-like particles for Cas9 mRNA/single guide RNA co-delivery and aptamer-containing lentiviral vectors for improved gene expression. Int J Biol Macromol 2022; 209:1260-1270. [PMID: 35461863 DOI: 10.1016/j.ijbiomac.2022.04.114] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023]
Abstract
Lentiviral vectors (LVs) are widely used for delivering foreign genes for long-term expression. Recently, virus-like particles (VLPs) were developed for mRNA or ribonucleoprotein (RNP) delivery for short-term endonuclease expression. Generating large amount of LVs or VLPs is challenging. On the other hand, methods for using VLPs to co-deliver Cas9 mRNA and single guide RNA (sgRNA) are limited. Fusing aptamer-binding protein (ABP) to the N-terminus of HIV Gag protein is currently the successful way to develop hybrid particles for co-delivering Cas9 mRNA and sgRNA. The effects of modifying Gag protein this way on particle assembly are unknown. Previously we found that adding an ABP after the second zinc finger domain of nucleocapsid (NC) protein had minimal effects on particle assembly. Based on these observations, here we developed hybrid particles for Cas9 mRNA and sgRNA co-delivery with normal capsid assembly efficiency. We further improved LVs for integrated gene expression by including an aptamer sequence in lentiviral genomic RNA, which improved lentiviral particle production and enhanced LV genomic RNA packaging. In summary, here we describe the development of new all-in-one VLPs for co-delivery of Cas9 mRNA and sgRNA, and new LVs for enhanced vector production and gene expression.
Collapse
Affiliation(s)
- Manish Yadav
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
21
|
Shi R, Jia S, Liu H, Nie H. Clinical grade lentiviral vector purification and quality control requirements. J Sep Sci 2022; 45:2093-2101. [PMID: 35247228 DOI: 10.1002/jssc.202100937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 11/10/2022]
Abstract
Lentiviral vectors have been proven to be a powerful tool in gene therapies that includes the ability to perform long-term gene editing in both dividing and non-dividing cells. In order to meet the rising demand of clinical grade lentiviral vectors for future clinical trials and requirements by regulatory agencies, new methods and technologies were developed, including the rapid optimization of production and purification processes. However, gaps still exist in achieving ideal yields and recovery rates in large-scale manufacturing process steps. The downstream purification process is a critical step required to obtain sufficient quantity and high-quality lentiviral vectors products, which is challenged by the low stability of the LV particles and large production volumes associated with the manufacturing process. This review summarizes the most recent and promising technologies and enhancements used in the large-scale purification process step of LV manufacturing and aims to provide a significant contribution towards the achievement of providing sufficient quantity and quality of LVs in scalable processes. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ruina Shi
- Immunochina Pharmaceutical Co., Ltd., Beijing, China
| | - Shenghua Jia
- Immunochina Pharmaceutical Co., Ltd., Beijing, China
| | - Huwei Liu
- College of Life Sciences, Wuchang University of Technology, Wuhan, China.,Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Honggang Nie
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Analytical Instrumental Center, Peking University, Beijing, China
| |
Collapse
|
22
|
Tan E, Chin CSH, Lim ZFS, Ng SK. HEK293 Cell Line as a Platform to Produce Recombinant Proteins and Viral Vectors. Front Bioeng Biotechnol 2021; 9:796991. [PMID: 34966729 PMCID: PMC8711270 DOI: 10.3389/fbioe.2021.796991] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/25/2021] [Indexed: 01/04/2023] Open
Abstract
Animal cell-based expression platforms enable the production of complex biomolecules such as recombinant proteins and viral vectors. Although most biotherapeutics are produced in animal cell lines, production in human cell lines is expanding. One important advantage of using human cell lines is the increased potential that the resulting biotherapeutics would carry more “human-like” post-translational modifications. Among the human cell lines, HEK293 is widely utilized due to its high transfectivity, rapid growth rate, and ability to grow in a serum-free, suspension culture. In this review, we discuss the use of HEK293 cells and its subtypes in the production of biotherapeutics. We also compare their usage against other commonly used host cell lines in each category of biotherapeutics and summarise the factors influencing the choice of host cell lines used.
Collapse
Affiliation(s)
- Evan Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Cara Sze Hui Chin
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Zhi Feng Sherman Lim
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| |
Collapse
|
23
|
Abstract
INTRODUCTION Lentiviral vectors have emerged as powerful vectors for vaccination, due to their high efficiency to transduce dendritic cells and to induce long-lasting humoral immunity, CD8+ T cells, and effective protection in numerous preclinical animal models of infection and oncology. AREAS COVERED Here, we reviewed the literature, highlighting the relevance of lentiviral vectors in vaccinology. We recapitulated both their virological and immunological aspects of lentiviral vectors. We compared lentiviral vectors to the gold standard viral vaccine vectors, i.e. adenoviral vectors, and updated the latest results in lentiviral vector-based vaccination in preclinical models. EXPERT OPINION Lentiviral vectors are non-replicative, negligibly inflammatory, and not targets of preexisting immunity in human populations. These are major characteristics to consider in vaccine development. The potential of lentiviral vectors to transduce non-dividing cells, including dendritic cells, is determinant in their strong immunogenicity. Notably, lentiviral vectors can be engineered to target antigen expression to specific host cells. The very weak inflammatory properties of these vectors allow their use in mucosal vaccination, with particular interest in infectious diseases that affect the lungs or brain, including COVID-19. Recent results in various preclinical models have reinforced the interest of these vectors in prophylaxis against infectious diseases and in onco-immunotherapy.
Collapse
Affiliation(s)
- Min-Wen Ku
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| | - Pierre Charneau
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laleh Majlessi
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| |
Collapse
|
24
|
Budde D, Jurkiewicz E. Risk analysis of leachables in cell and gene therapy using a CAR-T model process. Int J Pharm 2021; 607:121015. [PMID: 34411651 DOI: 10.1016/j.ijpharm.2021.121015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/17/2022]
Abstract
With the rapidly emerging field of autologous therapies, Single-Use (SU) technologies are increasingly used in personalized medicine due to their manifold advantages. Although qualification of the starting material of autologous therapies such as the CAR-T process has been highlighted, little attention has been paid to the effect of leachables on cell-based therapies, even if recent studies indicate interactions of leachables with cells. To close this gap, this study presents a risk-analysis of SU-material on a CAR-T process and identifies hazards imposed by tubing materials and leachables thereof. In order to represent a CAR-T process in its entirety, two test systems, namely a lentivirus production process and primary T-cells, were used. While the effects on lentivirus production are comparable to those reported for antibody production processes in CHO cells, we found that PVC material and corresponding leachables, i.e. plasticizer, inhibit cell growth of primary T-cells to a great extent. Additionally, our results indicate that critical quality attributes are affected by the PVC material.
Collapse
Affiliation(s)
- Dana Budde
- Sartorius Stedim Biotech GmbH, August Spindler-Str. 11, 37079 Goettingen, Germany; Universität Bielefeld, Universitätsstraße 25, 33615 Bielefeld, Germany.
| | - Elke Jurkiewicz
- Sartorius Stedim Biotech GmbH, August Spindler-Str. 11, 37079 Goettingen, Germany
| |
Collapse
|
25
|
Leinonen HM, Lepola S, Lipponen EM, Heikura T, Koponen T, Parker N, Ylä-Herttuala S, Lesch HP. Benchmarking of Scale-X Bioreactor System in Lentiviral and Adenoviral Vector Production. Hum Gene Ther 2021; 31:376-384. [PMID: 32075423 PMCID: PMC7087403 DOI: 10.1089/hum.2019.247] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We have previously produced viral vectors (lentiviral vector, adenoviral vector, and adeno-associated viral vector) in small and in commercial scale in adherent cells using Pall fixed-bed iCELLis® bioreactor. Recently, a company called Univercells has launched a new fixed-bed bioreactor with the same cell growth surface matrix material, but with different fixed-bed structure than is used in iCELLis bioreactor. We sought to compare the new scale-X™ hydro bioreactor (2.4 m2) and iCELLis Nano system (2.67 m2) to see if the difference has any effect on cell growth or lentiviral vector and adenoviral vector productivity. Runs were performed using parameters optimized for viral vector production in iCELLis Nano bioreactor. Cell growth was monitored by counting nuclei, as well as by following glucose consumption and lactate production. In both bioreactor systems, cells grew well, and the cell distribution was found quite homogeneous in scale-X bioreactor. Univercells scale-X bioreactor was proven to be at least equally efficient or even improved in both lentiviral vector and adenoviral vector production. Based on the results, the same protocol and parameters used in viral vector production in iCELLis bioreactor can also be successfully used for the production in scale-X bioreactor system.
Collapse
Affiliation(s)
- Hanna M Leinonen
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland; and
| | - Saana Lepola
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland; and
| | - Eevi M Lipponen
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland; and
| | - Tommi Heikura
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tiina Koponen
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nigel Parker
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hanna P Lesch
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland; and
| |
Collapse
|
26
|
Abaandou L, Quan D, Shiloach J. Affecting HEK293 Cell Growth and Production Performance by Modifying the Expression of Specific Genes. Cells 2021; 10:cells10071667. [PMID: 34359846 PMCID: PMC8304725 DOI: 10.3390/cells10071667] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
The HEK293 cell line has earned its place as a producer of biotherapeutics. In addition to its ease of growth in serum-free suspension culture and its amenability to transfection, this cell line’s most important attribute is its human origin, which makes it suitable to produce biologics intended for human use. At the present time, the growth and production properties of the HEK293 cell line are inferior to those of non-human cell lines, such as the Chinese hamster ovary (CHO) and the murine myeloma NSO cell lines. However, the modification of genes involved in cellular processes, such as cell proliferation, apoptosis, metabolism, glycosylation, secretion, and protein folding, in addition to bioprocess, media, and vector optimization, have greatly improved the performance of this cell line. This review provides a comprehensive summary of important achievements in HEK293 cell line engineering and on the global engineering approaches and functional genomic tools that have been employed to identify relevant genes for targeted engineering.
Collapse
Affiliation(s)
- Laura Abaandou
- Biotechnology Core Laboratory National Institutes of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; (L.A.); (D.Q.)
- Department of Chemistry and Biochemistry, College of Science, George Mason University, Fairfax, VA 22030, USA
| | - David Quan
- Biotechnology Core Laboratory National Institutes of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; (L.A.); (D.Q.)
| | - Joseph Shiloach
- Biotechnology Core Laboratory National Institutes of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; (L.A.); (D.Q.)
- Correspondence:
| |
Collapse
|
27
|
Dias MM, Vidigal J, Sequeira DP, Alves PM, Teixeira AP, Roldão A. Insect High FiveTM cell line development using site-specific flipase recombination technology. G3-GENES GENOMES GENETICS 2021; 11:6274903. [PMID: 33982066 PMCID: PMC8763235 DOI: 10.1093/g3journal/jkab166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 05/24/2021] [Indexed: 11/14/2022]
Abstract
Insect Trichoplusia ni High FiveTM (Hi5) cells have been widely explored for production of heterologous proteins, traditionally mostly using the lytic baculovirus expression vector system (BEVS), and more recently using virus-free transient gene expression systems. Stable expression in such host cells would circumvent the drawbacks associated with both systems when it comes to scale-up and implementation of more efficient high-cell density process modes for the manufacturing of biologics. In this work, we combined Flipase (Flp) recombinase-mediated cassette exchange (RMCE) with fluorescence-activated cell sorting (FACS) for generating a stable master clonal Hi5 cell line with the flexibility to express single or multiple proteins of interest from a tagged genomic locus. The 3-step protocol herein implemented consisted of (i) introducing the RMCE docking cassette into the cell genome by random integration followed by selection in Hygromycin B and FACS (Hi5-tagging population), (ii) eliminating cells tagged in loci with low recombination efficiency by transfecting the tagging population with an eGFP-containing target cassette followed by selection in G418 and FACS (Hi5-RMCE population), and (iii) isolation of pure eGFP-expressing cells by FACS and expansion to suspension cultures (Hi5-RMCE master clone). Exchangeability of the locus in the master clone was demonstrated in small-scale suspension cultures by replacing the target cassette by one containing a single protein (i.e. iCherry, as an intracellular protein model) or two proteins (i.e. influenza HA and M1 for virus-like particles production, as an extracellular protein model). Overall, the stable insect Hi5 cell platform herein assembled has the potential to assist and accelerate biologics development.
Collapse
Affiliation(s)
- Mafalda M Dias
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| | - João Vidigal
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| | - Daniela P Sequeira
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal.,Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
| | - Paula M Alves
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| | - Ana P Teixeira
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal.,ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 - Basel, Switzerland
| | - António Roldão
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| |
Collapse
|
28
|
Cellular pathways of recombinant adeno-associated virus production for gene therapy. Biotechnol Adv 2021; 49:107764. [PMID: 33957276 DOI: 10.1016/j.biotechadv.2021.107764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/10/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022]
Abstract
Recombinant adeno-associated viruses (rAAVs) are among the most important vectors for in vivo gene therapies. With the rapid development of gene therapy, current rAAV manufacturing capacity faces a challenge to meet the emerging demand for these therapies in the future. To examine the bottlenecks in rAAV production during cell culture, we focus here on an analysis of cellular pathways of rAAV production, based on an overview of assembly mechanisms first in the wild-type (wt) AAV replication and then in the common methods of rAAV production. The differences analyzed between the wild-type and recombinant systems provide insights into the mechanistic differences that may correlate with viral productivity. Based on these analyses, we identify potential barriers to high productivity of rAAV and discuss future directions for improvement to meet the emerging needs set by the growth of rAAV-based therapy and the needs of patients.
Collapse
|
29
|
Characterization of Extracellular Vesicles Secreted in Lentiviral Producing HEK293SF Cell Cultures. Viruses 2021; 13:v13050797. [PMID: 33946875 PMCID: PMC8145507 DOI: 10.3390/v13050797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022] Open
Abstract
Lentiviral vectors (LVs) are a powerful tool for gene and cell therapy and human embryonic kidney cells (HEK293) have been extensively used as a platform for production of these vectors. Like most cells and cellular tissues, HEK293 cells release extracellular vesicles (EVs). EVs released by cells share similar size, biophysical characteristics and even a biogenesis pathway with cell-produced enveloped viruses, making it a challenge to efficiently separate EVs from LVs. Thus, EVs co-purified with LVs during downstream processing, are considered “impurities” in the context of gene and cell therapy. A greater understanding of EVs co-purifying with LVs is needed to enable improved downstream processing. To that end, EVs from an inducible lentivirus producing cell line were studied under two conditions: non-induced and induced. EVs were identified in both conditions, with their presence confirmed by transmission electron microscopy and Western blot. EV cargos from each condition were then further characterized by a multi-omic approach. Nineteen proteins were identified by mass spectrometry as potential EV markers to differentiate EVs in LV preparations. Lipid composition of EV preparations before and after LV induction showed similar enrichment in phosphatidylserine. RNA cargos in EVs showed enrichment in transcripts involved in viral processes and binding functions. These findings provide insights on the product profile of lentiviral preparations and could support the development of improved separation strategies aimed at removing co-produced EVs.
Collapse
|
30
|
Jiang MQ, Yu SP, Wei ZZ, Zhong W, Cao W, Gu X, Wu A, McCrary MR, Berglund K, Wei L. Conversion of Reactive Astrocytes to Induced Neurons Enhances Neuronal Repair and Functional Recovery After Ischemic Stroke. Front Aging Neurosci 2021; 13:612856. [PMID: 33841125 PMCID: PMC8032905 DOI: 10.3389/fnagi.2021.612856] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
The master neuronal transcription factor NeuroD1 can directly reprogram astrocytes into induced neurons (iNeurons) after stroke. Using viral vectors to drive ectopic ND1 expression in gliotic astrocytes after brain injury presents an autologous form of cell therapy for neurodegenerative disease. Cultured astrocytes transfected with ND1 exhibited reduced proliferation and adopted neuronal morphology within 2-3 weeks later, expressed neuronal/synaptic markers, and extended processes. Whole-cell recordings detected the firing of evoked action potentials in converted iNeurons. Focal ischemic stroke was induced in adult GFAP-Cre-Rosa-YFP mice that then received ND1 lentivirus injections into the peri-infarct region 7 days after stroke. Reprogrammed cells did not express stemness genes, while 2-6 weeks later converted cells were co-labeled with YFP (constitutively activated in astrocytes), mCherry (ND1 infection marker), and NeuN (mature neuronal marker). Approximately 66% of infected cells became NeuN-positive neurons. The majority (~80%) of converted cells expressed the vascular glutamate transporter (vGLUT) of glutamatergic neurons. ND1 treatment reduced astrogliosis, and some iNeurons located/survived inside of the savaged ischemic core. Western blotting detected higher levels of BDNF, FGF, and PSD-95 in ND1-treated mice. MultiElectrode Array (MEA) recordings in brain slices revealed that the ND1-induced reprogramming restored interrupted cortical circuits and synaptic plasticity. Furthermore, ND1 treatment significantly improved locomotor, sensorimotor, and psychological functions. Thus, conversion of endogenous astrocytes to neurons represents a plausible, on-site regenerative therapy for stroke.
Collapse
Affiliation(s)
- Michael Qize Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, United States
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, United States
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, United States
| | - Weiwei Zhong
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, United States
| | - Wenyuan Cao
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, United States
| | - Anika Wu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Myles Randolph McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Ken Berglund
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, United States
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
31
|
Abstract
Gene therapy is an innovative treatment for Primary Immune Deficiencies (PIDs) that uses autologous hematopoietic stem cell transplantation to deliver stem cells with added or edited versions of the missing or malfunctioning gene that causes the PID. Initial studies of gene therapy for PIDs in the 1990-2000's used integrating murine gamma-retroviral vectors. While these studies showed clinical efficacy in many cases, especially with the administration of marrow cytoreductive conditioning before cell re-infusion, these vectors caused genotoxicity and development of leukoproliferative disorders in several patients. More recent studies used lentiviral vectors in which the enhancer elements of the long terminal repeats self-inactivate during reverse transcription ("SIN" vectors). These SIN vectors have excellent safety profiles and have not been reported to cause any clinically significant genotoxicity. Gene therapy has successfully treated several PIDs including Adenosine Deaminase Severe Combined Immunodeficiency (SCID), X-linked SCID, Artemis SCID, Wiskott-Aldrich Syndrome, X-linked Chronic Granulomatous Disease and Leukocyte Adhesion Deficiency-I. In all, gene therapy for PIDs has progressed over the recent decades to be equal or better than allogeneic HSCT in terms of efficacy and safety. Further improvements in methods should lead to more consistent and reliable efficacy from gene therapy for a growing list of PIDs.
Collapse
Affiliation(s)
- Lisa A. Kohn
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Donald B. Kohn
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
32
|
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
|
33
|
Martínez-Molina E, Chocarro-Wrona C, Martínez-Moreno D, Marchal JA, Boulaiz H. Large-Scale Production of Lentiviral Vectors: Current Perspectives and Challenges. Pharmaceutics 2020; 12:pharmaceutics12111051. [PMID: 33153183 PMCID: PMC7693937 DOI: 10.3390/pharmaceutics12111051] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/20/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023] Open
Abstract
Lentiviral vectors (LVs) have gained value over recent years as gene carriers in gene therapy. These viral vectors are safer than what was previously being used for gene transfer and are capable of infecting both dividing and nondividing cells with a long-term expression. This characteristic makes LVs ideal for clinical research, as has been demonstrated with the approval of lentivirus-based gene therapies from the Food and Drug Administration and the European Agency for Medicine. A large number of functional lentiviral particles are required for clinical trials, and large-scale production has been challenging. Therefore, efforts are focused on solving the drawbacks associated with the production and purification of LVsunder current good manufacturing practice. In recent years, we have witnessed the development and optimization of new protocols, packaging cell lines, and culture devices that are very close to reaching the target production level. Here, we review the most recent, efficient, and promising methods for the clinical-scale production ofLVs.
Collapse
Affiliation(s)
- Eduardo Martínez-Molina
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
| | - Carlos Chocarro-Wrona
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
| | - Daniel Martínez-Moreno
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
| | - Juan A. Marchal
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
| | - Houria Boulaiz
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-958-241-271
| |
Collapse
|
34
|
Soldi M, Sergi Sergi L, Unali G, Kerzel T, Cuccovillo I, Capasso P, Annoni A, Biffi M, Rancoita PMV, Cantore A, Lombardo A, Naldini L, Squadrito ML, Kajaste-Rudnitski A. Laboratory-Scale Lentiviral Vector Production and Purification for Enhanced Ex Vivo and In Vivo Genetic Engineering. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:411-425. [PMID: 33294490 PMCID: PMC7683235 DOI: 10.1016/j.omtm.2020.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/13/2020] [Indexed: 12/31/2022]
Abstract
Lentiviral vectors (LVs) are increasingly employed in gene and cell therapy. Standard laboratory production of LVs is not easily scalable, and research-grade LVs often contain contaminants that can interfere with downstream applications. Moreover, purified LV production pipelines have been developed mainly for costly, large-scale, clinical-grade settings. Therefore, a standardized and cost-effective process is still needed to obtain efficient, reproducible, and properly executed experimental studies and preclinical development of ex vivo and in vivo gene therapies, as high infectivity and limited adverse reactions are important factors potentially influencing experimental outcomes also in preclinical settings. We describe here an optimized laboratory-scale workflow whereby an LV-containing supernatant is purified and concentrated by sequential chromatographic steps, obtaining biologically active LVs with an infectious titer and specific activity in the order of 109 transducing unit (TU)/mL and 5 × 104 TU/ng of HIV Gag p24, respectively. The purification workflow removes >99% of the starting plasmid, DNA, and protein impurities, resulting in higher gene transfer and editing efficiency in severe combined immunodeficiency (SCID)-repopulating hematopoietic stem and progenitor cells (HSPCs) ex vivo, as well as reduced activation of inflammatory responses ex vivo and in vivo as compared to TU-matched, laboratory-grade vectors. Our results highlight the value of accessible purified LV production for experimental studies and preclinical testing.
Collapse
Affiliation(s)
- Monica Soldi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Lucia Sergi Sergi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Giulia Unali
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Thomas Kerzel
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Paola Capasso
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Mauro Biffi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Paola Maria Vittoria Rancoita
- CUSSB-University Center for Statistics and the Biomedical Statistics, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Mario Leonardo Squadrito
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
35
|
Ferreira MV, Cabral ET, Coroadinha AS. Progress and Perspectives in the Development of Lentiviral Vector Producer Cells. Biotechnol J 2020; 16:e2000017. [PMID: 32686901 DOI: 10.1002/biot.202000017] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/07/2020] [Indexed: 12/12/2022]
Abstract
After two decades of clinical trials, gene therapy demonstrated effectiveness in the treatment of a series of diseases. Currently, several gene therapy products are approved and used in the clinic. Lentiviral vectors (LVs) are one of the most used transfer vehicles to deliver genetic material and the vector of choice to modify hematopoietic cells to correct primary immunodeficiencies, hemoglobinopathies, and leukodystrophies. LVs are also widely used to modify T cells to treat cancers in immunotherapies (e.g., chimeric antigen receptors T cell therapies, CAR-T). In genome editing, LVs are used to deliver sequence-specific designer nucleases and DNA templates. The approval LV gene therapy products (e.g., Kymriah, for B-cell Acute lymphoblastic leukemia treatment; LentiGlobin, for β-thalassemia treatment) reinforced the need to improve their bioprocess manufacturing. The production has been mostly dependent on transient transfection. Production from stable cell lines facilitate GMP compliant processes, providing an easier scale-up, reproducibility and cost-effectiveness. The establishment of stable LV producer cell lines presents, however, several difficulties, with the cytotoxicity of some of the vector proteins being a major challenge. Genome editing technologies pose additional challenges to LV producer cells. Herein the major bottlenecks, recent achievements, and perspectives in the development of LV stable cell lines are revised.
Collapse
Affiliation(s)
- Mariana V Ferreira
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Elisa T Cabral
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Ana Sofia Coroadinha
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal.,The Discoveries centre for Regenerative and Precision Medicine, Nova University Lisbon, Oeiras Campus, Av. da República, 2780-157, Oeiras, Portugal
| |
Collapse
|
36
|
Sharon DM, Nesdoly S, Yang HJ, Gélinas JF, Xia Y, Ansorge S, Kamen AA. A pooled genome-wide screening strategy to identify and rank influenza host restriction factors in cell-based vaccine production platforms. Sci Rep 2020; 10:12166. [PMID: 32699298 PMCID: PMC7376217 DOI: 10.1038/s41598-020-68934-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/30/2020] [Indexed: 12/26/2022] Open
Abstract
Cell-derived influenza vaccines provide better protection and a host of other advantages compared to the egg-derived vaccines that currently dominate the market, but their widespread use is hampered by a lack of high yield, low cost production platforms. Identification and knockout of innate immune and metabolic restriction factors within relevant host cell lines used to grow the virus could offer a means to substantially increase vaccine yield. In this paper, we describe and validate a novel genome-wide pooled CRISPR/Cas9 screening strategy that incorporates a reporter virus and a FACS selection step to identify and rank restriction factors in a given vaccine production cell line. Using the HEK-293SF cell line and A/PuertoRico/8/1934 H1N1 influenza as a model, we identify 64 putative influenza restriction factors to direct the creation of high yield knockout cell lines. In addition, gene ontology and protein complex enrichment analysis of this list of putative restriction factors offers broader insights into the primary host cell determinants of viral yield in cell-based vaccine production systems. Overall, this work will advance efforts to address the public health burden posed by influenza.
Collapse
MESH Headings
- CRISPR-Cas Systems/genetics
- Cell Survival
- Gene Editing
- Gene Ontology
- Genes, Reporter
- Genetic Vectors/genetics
- Genetic Vectors/metabolism
- Genome, Viral
- HEK293 Cells
- Humans
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/metabolism
- Influenza, Human/pathology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- RNA, Guide, CRISPR-Cas Systems/metabolism
- Virus Replication
Collapse
Affiliation(s)
- David M. Sharon
- Department of Bioengineering, McGill University, McConnell Engineering Building, Room 363, 3480 Rue University, Montreal, QC H3A 2K6 Canada
| | - Sean Nesdoly
- Department of Bioengineering, McGill University, McConnell Engineering Building, Room 363, 3480 Rue University, Montreal, QC H3A 2K6 Canada
| | - Hsin J. Yang
- Department of Bioengineering, McGill University, McConnell Engineering Building, Room 363, 3480 Rue University, Montreal, QC H3A 2K6 Canada
| | - Jean-François Gélinas
- Department of Bioengineering, McGill University, McConnell Engineering Building, Room 363, 3480 Rue University, Montreal, QC H3A 2K6 Canada
| | - Yu Xia
- Department of Bioengineering, McGill University, McConnell Engineering Building, Room 363, 3480 Rue University, Montreal, QC H3A 2K6 Canada
| | - Sven Ansorge
- Human Health Therapeutics, National Research Council of Canada, Montreal, QC Canada
| | - Amine A. Kamen
- Department of Bioengineering, McGill University, McConnell Engineering Building, Room 363, 3480 Rue University, Montreal, QC H3A 2K6 Canada
- Human Health Therapeutics, National Research Council of Canada, Montreal, QC Canada
| |
Collapse
|
37
|
Concise review on optimized methods in production and transduction of lentiviral vectors in order to facilitate immunotherapy and gene therapy. Biomed Pharmacother 2020; 128:110276. [PMID: 32502836 DOI: 10.1016/j.biopha.2020.110276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Lentiviral vectors (LVs) have provided an efficient way to integrate our gene of interest into eukaryote cells. Human immunodeficiency virus (HIV)-derived LVs have been vastly studied to become an invaluable asset in gene delivery. This abled LVs to be used in both research laboratories and gene therapy. Pseudotyping HIV-1 based LVs, abled it to transduce different types of cells, especially hematopoietic stem cells. A wide range of tropism, plus to the ability to integrate genes into target cells, made LVs an armamentarium in gene therapy. The third and fourth generations of self-inactivating LVs are being used to achieve safe gene therapy. Not only advanced methods enabled the clinical-grade LV production on a large scale, but also considerably heightened transduction efficiency. One of which is microfluidic systems that revolutionized gene delivery approaches. Since gene therapy using LVs attracted lots of attention to itself, we provided a brief review of LV structure and life-cycle along with methods for improving both LV production and transduction. Also, we mentioned some of their utilization in immunotherapy and gene therapy.
Collapse
|
38
|
Development of a laboratory scalable process for enhancing lentivirus production by transient transfection of HEK293 adherent cultures. Gene Ther 2020; 27:482-494. [DOI: 10.1038/s41434-020-0152-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/25/2022]
|
39
|
Do Minh A, Tran MY, Kamen AA. Lentiviral Vector Production in Suspension Culture Using Serum-Free Medium for the Transduction of CAR-T Cells. Methods Mol Biol 2020; 2086:77-83. [PMID: 31707669 DOI: 10.1007/978-1-0716-0146-4_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The production of lentiviral vectors (LVs) in human embryonic kidney 293 (HEK293) cells using serum-free medium in a suspension culture for the transduction of chimeric antigen receptor T-cells (CAR-T) can be achieved by different methods. This chapter describes LV production by transient transfection, induction of stable packaging cell lines, and induction of stable producer cell lines.
Collapse
Affiliation(s)
- Aline Do Minh
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Michelle Yen Tran
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montreal, QC, Canada.
| |
Collapse
|
40
|
Durous L, Rosa-Calatrava M, Petiot E. Advances in influenza virus-like particles bioprocesses. Expert Rev Vaccines 2019; 18:1285-1300. [DOI: 10.1080/14760584.2019.1704262] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Laurent Durous
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Emma Petiot
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| |
Collapse
|
41
|
Moleirinho MG, Silva RJS, Alves PM, Carrondo MJT, Peixoto C. Current challenges in biotherapeutic particles manufacturing. Expert Opin Biol Ther 2019; 20:451-465. [PMID: 31773998 DOI: 10.1080/14712598.2020.1693541] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: The development of novel complex biotherapeutics led to new challenges in biopharmaceutical industry. The potential of these particles has been demonstrated by the approval of several products, in the different fields of gene therapy, oncolytic therapy, and tumor vaccines. However, their manufacturing still presents challenges related to the high dosages and purity required.Areas covered: The main challenges that biopharmaceutical industry faces today and the most recent developments in the manufacturing of different biotherapeutic particles are reported here. Several unit operations and downstream trains to purify virus, virus-like particles and extracellular vesicles are described. Innovations on the different purification steps are also highlighted with an eye on the implementation of continuous and integrated processes.Expert opinion: Manufacturing platforms that consist of a low number of unit operations, with higher-yielding processes and reduced costs will be highly appreciated by the industry. The pipeline of complex therapeutic particles is expanding and there is a clear need for advanced tools and manufacturing capacity. The use of single-use technologies, as well as continuous integrated operations, are gaining ground in the biopharmaceutical industry and should be supported by more accurate and faster analytical methods.
Collapse
Affiliation(s)
- Mafalda G Moleirinho
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal.,ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Ricardo J S Silva
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal
| | - Paula M Alves
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal.,ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Manuel J T Carrondo
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal
| | - Cristina Peixoto
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal.,ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| |
Collapse
|
42
|
Bauler M, Roberts JK, Wu CC, Fan B, Ferrara F, Yip BH, Diao S, Kim YI, Moore J, Zhou S, Wielgosz MM, Ryu B, Throm RE. Production of Lentiviral Vectors Using Suspension Cells Grown in Serum-free Media. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:58-68. [PMID: 31890741 PMCID: PMC6931067 DOI: 10.1016/j.omtm.2019.11.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 11/15/2019] [Indexed: 02/09/2023]
Abstract
Lentiviral vectors are increasingly utilized in cell and gene therapy applications because they efficiently transduce target cells such as hematopoietic stem cells and T cells. Large-scale production of current Good Manufacturing Practices-grade lentiviral vectors is limited because of the adherent, serum-dependent nature of HEK293T cells used in the manufacturing process. To optimize large-scale clinical-grade lentiviral vector production, we developed an improved production scheme by adapting HEK293T cells to grow in suspension using commercially available and chemically defined serum-free media. Lentiviral vectors with titers equivalent to those of HEK293T cells were produced from SJ293TS cells using optimized transfection conditions that reduced the required amount of plasmid DNA by 50%. Furthermore, purification of SJ293TS-derived lentiviral vectors at 1 L yielded a recovery of 55% ± 14% (n = 138) of transducing units in the starting material, more than a 2-fold increase over historical yields from adherent HEK293T serum-dependent lentiviral vector preparations. SJ293TS cells were stable to produce lentiviral vectors over 4 months of continuous culture. SJ293TS-derived lentiviral vectors efficiently transduced primary hematopoietic stem cells and T cells from healthy donors. Overall, our SJ293TS cell line enables high-titer vector production in serum-free conditions while reducing the amount of input DNA required, resulting in a highly efficient manufacturing option.
Collapse
Affiliation(s)
- Matthew Bauler
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jessica K Roberts
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chang-Chih Wu
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Baochang Fan
- Therapeutics Production and Quality, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Francesca Ferrara
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Bon Ham Yip
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shiyong Diao
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Young-In Kim
- Experimental Cell Therapeutics Lab, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer Moore
- Experimental Cell Therapeutics Lab, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sheng Zhou
- Experimental Cell Therapeutics Lab, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Matthew M Wielgosz
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Byoung Ryu
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Robert E Throm
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
43
|
Curing Hemoglobinopathies: Challenges and Advances of Conventional and New Gene Therapy Approaches. Mediterr J Hematol Infect Dis 2019; 11:e2019067. [PMID: 31700592 PMCID: PMC6827604 DOI: 10.4084/mjhid.2019.067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022] Open
Abstract
Inherited hemoglobin disorders, including beta-thalassemia (BT) and sickle-cell disease (SCD), are the most common monogenic diseases worldwide, with a global carrier frequency of over 5%.1 With migration, they are becoming more common worldwide, making their management and care an increasing concern for health care systems. BT is characterized by an imbalance in the α/β-globin chain ratio, ineffective erythropoiesis, chronic hemolytic anemia, and compensatory hemopoietic expansion.1 Globally, there are over 25,000 births each year with transfusion-dependent thalassemia (TDT). The currently available treatment for TDT is lifelong transfusions and iron chelation therapy or allogenic bone marrow transplantation as a curative option. SCD affects 300 million people worldwide2 and severely impacts the quality of life of patients who experience unpredictable, recurrent acute and chronic severe pain, stroke, infections, pulmonary disease, kidney disease, retinopathy, and other complications. While survival has been dramatically extended, quality of life is markedly reduced by disease- and treatment-associated morbidity. The development of safe, tissue-specific and efficient vectors, and efficient gene-editing technologies have led to the development of several gene therapy trials for BT and SCD. However, the complexity of the approach presents its hurdles. Fundamental factors at play include the requirement for myeloablation on a patient with benign disease, the age of the patient, and the consequent bone marrow microenvironment. A successful path from proof-ofconcept studies to commercialization must render gene therapy a sustainable and accessible approach for a large number of patients. Furthermore, the cost of these therapies is a considerable challenge for the health care system. While new promising therapeutic options are emerging,3,4 and many others are on the pipeline,5 gene therapy can potentially cure patients. We herein provide an overview of the most recent, likely potentially curative therapies for hemoglobinopathies and a summary of the challenges that these approaches entail.
Collapse
|
44
|
Gélinas JF, Azizi H, Kiesslich S, Lanthier S, Perdersen J, Chahal PS, Ansorge S, Kobinger G, Gilbert R, Kamen AA. Production of rVSV-ZEBOV in serum-free suspension culture of HEK 293SF cells. Vaccine 2019; 37:6624-6632. [DOI: 10.1016/j.vaccine.2019.09.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022]
|
45
|
Chen P, Demirji J, Ivleva VB, Horwitz J, Schwartz R, Arnold F. The transient expression of CHIKV VLP in large stirred tank bioreactors. Cytotechnology 2019; 71:1079-1093. [PMID: 31560090 DOI: 10.1007/s10616-019-00346-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/19/2019] [Indexed: 01/06/2023] Open
Abstract
Transient gene expression (TGE) bioprocesses have been difficult to scale up in large stirred tank bioreactors with volumes of more than 1.5 L. Low production levels are often observed, but the causes have not been investigated (Gutierrez-Granados et al. in Crit Rev Biotechnol 38:918-940, 2018). Chikungunya Virus-like particle (VLP), expressed by DNA-PEI transient transfection, is a representative case study for these difficulties. Clinical materials were produced in shake flasks, but the process suffered when transferred to large stirred tank bioreactors. The resulting process was not operationally friendly nor cost effective. In this study, a systematic approach was used to investigate the root causes of the poor scale up performance. The transfection conditions were first screened in ambr® 15 high throughput mini bioreactors then examined in 3 L stirred-tank systems. The studies found that production level was negatively correlated with inoculum cell growth status (P < 0.05). The pH range, DNA and PEI levels, order of the reagent addition, and gas-sparging systems were also studied and found to affect process performance. Further hydromechanical characterizations (Re, energy dissipation rates, and P/V, etc.) of shake flasks, ambr® 15, and 3-L stirred tank systems were performed. Overall, the study discovered that the shear stress (caused by a microsparger) and PEI toxicity together were the root causes of scale-up failure. Once the microsparger was replaced by a macrosparger, the scale-up was successful.
Collapse
Affiliation(s)
- Peifeng Chen
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd, Gaithersburg, MD, 20878, USA.
| | - Jacob Demirji
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd, Gaithersburg, MD, 20878, USA
| | - Vera B Ivleva
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd, Gaithersburg, MD, 20878, USA
| | - Joe Horwitz
- Amicus Therapeutics, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | | | - Frank Arnold
- Tunnell Consulting, 900 E. 8th Ave, King of Prussia, PA, 19406, USA
| |
Collapse
|
46
|
Leinonen HM, Lipponen EM, Valkama AJ, Hynynen H, Oruetxebarria I, Turkki V, Olsson V, Kurkipuro J, Samaranayake H, Määttä AM, Parker NR, Ylä-Herttuala S, Lesch HP. Preclinical Proof-of-Concept, Analytical Development, and Commercial Scale Production of Lentiviral Vector in Adherent Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:63-71. [PMID: 31649956 PMCID: PMC6804948 DOI: 10.1016/j.omtm.2019.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/20/2019] [Indexed: 12/22/2022]
Abstract
The therapeutic efficacy of a lentiviral vector (LV) expressing the herpes simplex virus thymidine kinase (HSV-TK) was studied in an immunocompetent rat glioblastoma model. Intraperitoneal ganciclovir injections (50 mg/kg/day) were administered for 14 consecutive days, resulting in reduced tumor volumes as monitored by MRI. Survival analyses revealed a significant improvement among the LV-expressing HSV-TK (LV-TK)/ganciclovir-treated animals when compared to non-treated control rats. However, a limiting factor in the use of LV has been the suboptimal small-scale production in flasks. Our aim during the translation phase, prior to entering the final pre-clinical and early clinical phases, was to develop a scalable, robust, and disposable manufacturing process for LV-TKs. We also aimed to minimize future process changes and enable production upscaling to make the process suitable for larger patient populations. The upstream process relies on fixed-bed iCELLis technology and transient plasmid transfection. This is the first time iCELLis 500 commercial-scale bioreactor was used for LV production. A testing strategy to determine the pharmacological activity of LV-TK drug product by measuring cell viability was developed, and the specificity of the potency assay was also proven. In this paper we focus on upstream process development while showing analytical development and the proof-of-concept of LV-TK functionality.
Collapse
Affiliation(s)
- Hanna M Leinonen
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland
| | - Eevi M Lipponen
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland
| | - Anniina J Valkama
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland.,Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Heidi Hynynen
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland
| | - Igor Oruetxebarria
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland
| | - Vesa Turkki
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland
| | - Venla Olsson
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | - Ann-Marie Määttä
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nigel R Parker
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hanna P Lesch
- Kuopio Center for Gene and Cell Therapy, Kuopio, Finland.,FinVector, Kuopio, Finland
| |
Collapse
|
47
|
Binz RL, Tian E, Sadhukhan R, Zhou D, Hauer-Jensen M, Pathak R. Identification of novel breakpoints for locus- and region-specific translocations in 293 cells by molecular cytogenetics before and after irradiation. Sci Rep 2019; 9:10554. [PMID: 31332273 PMCID: PMC6646394 DOI: 10.1038/s41598-019-47002-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022] Open
Abstract
The human kidney embryonic 293 cell line (293 cells) is extensively used in biomedical and pharmaceutical research. These cells exhibit a number of numerical and structural chromosomal anomalies. However, the breakpoints responsible for these structural chromosomal rearrangements have not been comprehensively characterized. In addition, it is not known whether chromosomes with structural rearrangement are more sensitive to external toxic agents, such as ionizing radiation. We used G-banding, spectral karyotyping (SKY), and locus- and region-specific fluorescence in situ hybridization (FISH) probes designed in our lab or obtained from commercial vendor to address this gap. Our G-banding analysis revealed that the chromosome number varies from 66 to 71, with multiple rearrangements and partial additions and deletions. SKY analysis confirmed 3 consistent rearrangements, two simple and one complex in nature. Multicolor FISH analysis identified an array of breakpoints responsible for locus- and region-specific translocations. Finally, SKY analysis revealed that radio-sensitivity of structurally rearranged chromosomes is dependent on radiation dose. These findings will advance our knowledge in 293 cell biology and will enrich the understanding of radiation biology studies.
Collapse
Affiliation(s)
- Regina L Binz
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Erming Tian
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ratan Sadhukhan
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Pharmacodynamics, College of Pharmacy, Gainesville, FL, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
48
|
Poorebrahim M, Sadeghi S, Fakhr E, Abazari MF, Poortahmasebi V, Kheirollahi A, Askari H, Rajabzadeh A, Rastegarpanah M, Linē A, Cid-Arregui A. Production of CAR T-cells by GMP-grade lentiviral vectors: latest advances and future prospects. Crit Rev Clin Lab Sci 2019; 56:393-419. [PMID: 31314617 DOI: 10.1080/10408363.2019.1633512] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR) T-cells represent a paradigm shift in cancer immunotherapy and a new milestone in the history of oncology. In 2017, the Food and Drug Administration approved two CD19-targeted CAR T-cell therapies (Kymriah™, Novartis, and Yescarta™, Kite Pharma/Gilead Sciences) that have remarkable efficacy in some B-cell malignancies. The CAR approach is currently being evaluated in multiple pivotal trials designed for the immunotherapy of hematological malignancies as well as solid tumors. To generate CAR T-cells ex vivo, lentiviral vectors (LVs) are particularly appealing due to their ability to stably integrate relatively large DNA inserts, and to efficiently transduce both dividing and nondividing cells. This review discusses the latest advances and challenges in the design and production of CAR T-cells, and the good manufacturing practices (GMP)-grade production process of LVs used as a gene transfer vehicle. New developments in the application of CAR T-cell therapy are also outlined with particular emphasis on next-generation allogeneic CAR T-cells.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Solmaz Sadeghi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR , Tehran , Iran
| | - Elham Fakhr
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) , Heidelberg , Germany
| | - Mohammad Foad Abazari
- Research Center for Clinical Virology, Tehran University of Medical Sciences , Tehran , Iran
| | - Vahdat Poortahmasebi
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,Faculty of Medicine, Department of Bacteriology and Virology, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran , Tehran , Iran
| | - Hassan Askari
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Rajabzadeh
- Applied Cell Sciences and Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Malihe Rastegarpanah
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Aija Linē
- Latvian Biomedical Research and Study Centre , Riga , Latvia
| | - Angel Cid-Arregui
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR , Tehran , Iran.,Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
49
|
Garg U, Chauhan S, Nagaich U, Jain N. Current Advances in Chitosan Nanoparticles Based Drug Delivery and Targeting. Adv Pharm Bull 2019; 9:195-204. [PMID: 31380245 PMCID: PMC6664124 DOI: 10.15171/apb.2019.023] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/11/2019] [Accepted: 04/13/2019] [Indexed: 01/08/2023] Open
Abstract
Nanoparticles (NPs) have been found to be potential targeted and controlled release drug delivery systems. Various drugs can be loaded in the NPs to achieve targeted delivery. Chitosan NPs being biodegradable, biocompatible, less toxic and easy to prepare, are an effective and potential tool for drug delivery. Chitosan is natural biopolymer which can be easily functionalized to obtain the desired targeted results and is also approved by GRAS (Generally Recognized as Safe by the United States Food and Drug Administration [US FDA]). Various methods for preparation of chitosan NPs include, ionic cross-linking, covalent cross-linking, reverse micellar method, precipitation and emulsion-droplet coalescence method. Chitosan NPs are found to have plethora of applications in drug delivery diagnosis and other biological applications. The key applications include ocular drug delivery, per-oral delivery, pulmonary drug delivery, nasal drug delivery, mucosal drug delivery, gene delivery, buccal drug delivery, vaccine delivery, vaginal drug delivery and cancer therapy. The present review describes the formation of chitosan, synthesis of chitosan NPs and their various applications in drug delivery.
Collapse
Affiliation(s)
| | | | | | - Neha Jain
- Amity Institute of Pharmacy, Amity University, Sector-125, Noida, Uttar Pradesh-201303
| |
Collapse
|
50
|
McCarron A, Donnelley M, McIntyre C, Parsons D. Transient Lentiviral Vector Production Using a Packed-Bed Bioreactor System. Hum Gene Ther Methods 2019; 30:93-101. [DOI: 10.1089/hgtb.2019.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Alexandra McCarron
- Adelaide Medical School, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Respiratory and Sleep Medicine, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
| | - Martin Donnelley
- Adelaide Medical School, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Respiratory and Sleep Medicine, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
| | - Chantelle McIntyre
- Adelaide Medical School, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
- Genetics and Molecular Pathology, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
| | - David Parsons
- Adelaide Medical School, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Respiratory and Sleep Medicine, SA Pathology, Women's and Children's Hospital, Adelaide, Australia
| |
Collapse
|