1
|
Colao IL, Corteling RL, Bracewell DG, Wall IB. Neural stem cell-derived extracellular vesicles purified by monolith chromatography retain stimulatory effect in in vitro scratch assay. Cytotherapy 2025; 27:365-377. [PMID: 39755977 DOI: 10.1016/j.jcyt.2024.11.007] [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: 03/11/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND AIMS Extracellular vesicles (EVs) have gained traction as potential cell-free therapeutic candidates. Development of purification methods that are scalable and robust is a major focus of EV research. Yet there is still little in the literature that evaluates purification methods against potency of the EV product. In the present study, we examined two monolith chromatography methods with a focus on assessing the ability of purified EVs to retain stimulatory effects on fibroblasts to connect scalable purification methods with product outputs. METHODS We characterized EVs recovered from CTX0E03 (CTX) neural stem cell-conditioned medium in terms of biomarker distribution, functional capacity and purity. We evaluated the ability of EVs to promote wound closure in an in vitro scratch assay prior to and following two monolith chromatography steps (anion exchange and hydrophobic interaction) to determine whether these options may better serve EV bioprocessing. RESULTS EVs from CTX cells were successful in initiating wound repair in a fibroblast scratch assay over 72 h with a single 20-μg dose. EV preparations presented the markers CD9, CD81 and CD63 but also contained culture albumin and DNA as process impurities. EVs recovered by tangential flow filtration could be successfully purified further by both monolith chromatography steps. Post-monolith EV stimulation was conserved. CONCLUSIONS The results indicate that monolith chromatography is a viable purification method for EVs derived from cell culture that does not detract from the product's ability to stimulate fibroblasts, suggesting that product functionality is conserved. Further work is needed in developing suitable downstream processes and analytics to achieve clinically relevant purities for injectable biologics.
Collapse
Affiliation(s)
- Ivano Luigi Colao
- Department of Biochemical Engineering, University College London, London, UK
| | | | - Daniel G Bracewell
- Department of Biochemical Engineering, University College London, London, UK.
| | - Ivan B Wall
- Institute of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
2
|
Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain 2024; 147:766-793. [PMID: 37975820 DOI: 10.1093/brain/awad392] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a leading cause of lifelong disabilities. Permanent sensory, motor and autonomic impairments after SCI are substantially attributed to degeneration of spinal cord neurons and axons, and disintegration of neural network. To date, minimal regenerative treatments are available for SCI with an unmet need for new therapies to reconstruct the damaged spinal cord neuron-glia network and restore connectivity with the supraspinal pathways. Multipotent neural precursor cells (NPCs) have a unique capacity to generate neurons, oligodendrocytes and astrocytes. Due to this capacity, NPCs have been an attractive cell source for cellular therapies for SCI. Transplantation of NPCs has been extensively tested in preclinical models of SCI in the past two decades. These studies have identified opportunities and challenges associated with NPC therapies. While NPCs have the potential to promote neuroregeneration through various mechanisms, their low long-term survival and integration within the host injured spinal cord limit the functional benefits of NPC-based therapies for SCI. To address this challenge, combinatorial strategies have been developed to optimize the outcomes of NPC therapies by enriching SCI microenvironment through biomaterials, genetic and pharmacological therapies. In this review, we will provide an in-depth discussion on recent advances in preclinical NPC-based therapies for SCI. We will discuss modes of actions and mechanism by which engrafted NPCs contribute to the repair process and functional recovery. We will also provide an update on current clinical trials and new technologies that have facilitated preparation of medical-grade human NPCs suitable for transplantation in clinical studies.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
| | - Ben Borys
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
3
|
Satani N, Parsha K, Savitz SI. Enhancing Stroke Recovery With Cellular Therapies. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00062-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
4
|
Adult Human Multipotent Neural Cells Could Be Distinguished from Other Cell Types by Proangiogenic Paracrine Effects via MCP-1 and GRO. Stem Cells Int 2021; 2021:6737288. [PMID: 34434240 PMCID: PMC8380502 DOI: 10.1155/2021/6737288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Adult human multipotent neural cells (ahMNCs) are unique cells derived from adult human temporal lobes. They show multipotent differentiation potentials into neurons and astrocytes. In addition, they possess proangiogenic capacities. The objective of this study was to characterize ahMNCs in terms of expression of cell type-specific markers, in vitro differentiation potentials, and paracrine factors compared with several other cell types including fetal neural stem cells (fNSCs) to provide detailed molecular and functional features of ahMNCs. Interestingly, the expression of cell type-specific markers of ahMNCs could not be differentiated from those of pericytes, mesenchymal stem cells (MSCs), or fNSCs. In contrast, differentiation potentials of ahMNCs and fNSCs into neural cells were higher than those of other cell types. Compared with MSCs, ahMNCs showed lower differentiation capacities into osteogenic and adipogenic cells. Moreover, ahMNCs uniquely expressed higher levels of MCP-1 and GRO family paracrine factors than fNSCs and MSCs. These high levels of MCP-1 and GRO family mediated in vivo proangiogenic effects of ahMNCs. These results indicate that ahMNCs have their own distinct characteristics that could distinguish ahMNCs from other cell types. Characteristics of ahMNCs could be utilized further in the preclinical and clinical development of ahMNCs for regenerative medicine. They could also be used as experimental references for other cell types including fNSCs.
Collapse
|
5
|
Katsur M, He Z, Vinokur V, Corteling R, Yellon DM, Davidson SM. Exosomes from neuronal stem cells may protect the heart from ischaemia/reperfusion injury via JAK1/2 and gp130. J Cell Mol Med 2021; 25:4455-4465. [PMID: 33797200 PMCID: PMC8093960 DOI: 10.1111/jcmm.16515] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Myocardial infarction requires urgent reperfusion to salvage viable heart tissue. However, reperfusion increases infarct size further by promoting mitochondrial damage in cardiomyocytes. Exosomes from a wide range of different cell sources have been shown to activate cardioprotective pathways in cardiomyocytes, thereby reducing infarct size. Yet, it is currently challenging to obtain highly pure exosomes in quantities enough for clinical studies. To overcome this problem, we used exosomes isolated from CTX0E03 neuronal stem cells, which are genetically stable, conditionally inducible and can be produced on an industrial scale. However, it is unknown whether exosomes from neuronal stem cells may reduce cardiac ischaemia/reperfusion injury. In this study, we demonstrate that exosomes from differentiating CTX0E03 cells can reduce infarct size in mice. In an in vitro assay, these exosomes delayed cardiomyocyte mitochondrial permeability transition pore opening, which is responsible for cardiomyocyte death after reperfusion. The mechanism of MPTP inhibition was via gp130 signalling and the downstream JAK/STAT pathway. Our results support previous findings that exosomes from non‐cardiomyocyte‐related cells produce exosomes capable of protecting cardiomyocytes from myocardial infarction. We anticipate our findings may encourage scientists to use exosomes obtained from reproducible clinical‐grade stocks of cells for their ischaemia/reperfusion studies.
Collapse
Affiliation(s)
- Miroslava Katsur
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Vladimir Vinokur
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| |
Collapse
|
6
|
Jiao Y, Liu YW, Chen WG, Liu J. Neuroregeneration and functional recovery after stroke: advancing neural stem cell therapy toward clinical application. Neural Regen Res 2021; 16:80-92. [PMID: 32788451 PMCID: PMC7818886 DOI: 10.4103/1673-5374.286955] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stroke is a main cause of death and disability worldwide. The ability of the brain to self-repair in the acute and chronic phases after stroke is minimal; however, promising stem cell-based interventions are emerging that may give substantial and possibly complete recovery of brain function after stroke. Many animal models and clinical trials have demonstrated that neural stem cells (NSCs) in the central nervous system can orchestrate neurological repair through nerve regeneration, neuron polarization, axon pruning, neurite outgrowth, repair of myelin, and remodeling of the microenvironment and brain networks. Compared with other types of stem cells, NSCs have unique advantages in cell replacement, paracrine action, inflammatory regulation and neuroprotection. Our review summarizes NSC origins, characteristics, therapeutic mechanisms and repair processes, then highlights current research findings and clinical evidence for NSC therapy. These results may be helpful to inform the direction of future stroke research and to guide clinical decision-making.
Collapse
Affiliation(s)
- Yang Jiao
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University; Dalian Innovation Institute of Stem Cells and Precision Medicine, Dalian, Liaoning Province, China
| | - Yu-Wan Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wei-Gong Chen
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University; Dalian Innovation Institute of Stem Cells and Precision Medicine, Dalian, Liaoning Province, China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University; Dalian Innovation Institute of Stem Cells and Precision Medicine, Dalian, Liaoning Province, China
| |
Collapse
|
7
|
Castilla-Casadiego DA, Reyes-Ramos AM, Domenech M, Almodovar J. Effects of Physical, Chemical, and Biological Stimulus on h-MSC Expansion and Their Functional Characteristics. Ann Biomed Eng 2020; 48:519-535. [PMID: 31705365 PMCID: PMC6952531 DOI: 10.1007/s10439-019-02400-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/30/2019] [Indexed: 01/10/2023]
Abstract
Human adult mesenchymal stem or stromal cells (h-MSC) therapy has gained considerable attention due to the potential to treat or cure diseases given their immunosuppressive properties and tissue regeneration capabilities. Researchers have explored diverse strategies to promote high h-MSC production without losing functional characteristics or properties. Physical stimulus including stiffness, geometry, and topography, chemical stimulus, like varying the surface chemistry, and biochemical stimuli such as cytokines, hormones, small molecules, and herbal extracts have been studied but have yet to be translated to industrial manufacturing practice. In this review, we describe the role of those stimuli on h-MSC manufacturing, and how these stimuli positively promote h-MSC properties, impacting the cell manufacturing field for cell-based therapies. In addition, we discuss other process considerations such as bioreactor design, good manufacturing practice, and the importance of the cell donor and ethics factors for manufacturing potent h-MSC.
Collapse
Affiliation(s)
- David A Castilla-Casadiego
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Ana M Reyes-Ramos
- Department of Chemical Engineering, University of Puerto Rico Mayagüez, Call Box 9000, Mayagüez, PR, 00681-9000, USA
| | - Maribella Domenech
- Department of Chemical Engineering, University of Puerto Rico Mayagüez, Call Box 9000, Mayagüez, PR, 00681-9000, USA
| | - Jorge Almodovar
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA.
| |
Collapse
|
8
|
Moutsatsou P, Ochs J, Schmitt RH, Hewitt CJ, Hanga MP. Automation in cell and gene therapy manufacturing: from past to future. Biotechnol Lett 2019; 41:1245-1253. [PMID: 31541330 PMCID: PMC6811377 DOI: 10.1007/s10529-019-02732-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/12/2019] [Indexed: 01/19/2023]
Abstract
As more and more cell and gene therapies are being developed and with the increasing number of regulatory approvals being obtained, there is an emerging and pressing need for industrial translation. Process efficiency, associated cost drivers and regulatory requirements are issues that need to be addressed before industrialisation of cell and gene therapies can be established. Automation has the potential to address these issues and pave the way towards commercialisation and mass production as it has been the case for 'classical' production industries. This review provides an insight into how automation can help address the manufacturing issues arising from the development of large-scale manufacturing processes for modern cell and gene therapy. The existing automated technologies with applicability in cell and gene therapy manufacturing are summarized and evaluated here.
Collapse
Affiliation(s)
- P Moutsatsou
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B7 4ET, UK
| | - J Ochs
- Fraunhofer Institut für Produktionstechnologie IPT, Steinbachstrasse 17, 52074, Aachen, Germany
| | - R H Schmitt
- Fraunhofer Institut für Produktionstechnologie IPT, Steinbachstrasse 17, 52074, Aachen, Germany.,Laboratory for Machine Tools and Production Engineering (WZL), RWTH, Aachen, Germany
| | - C J Hewitt
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B7 4ET, UK
| | - M P Hanga
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B7 4ET, UK.
| |
Collapse
|
9
|
Dabrowski A, Robinson TJ, Felling RJ. Promoting Brain Repair and Regeneration After Stroke: a Plea for Cell-Based Therapies. Curr Neurol Neurosci Rep 2019; 19:5. [PMID: 30712068 DOI: 10.1007/s11910-019-0920-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW After decades of hype, cell-based therapies are emerging into the clinical arena for the purposes of promoting recovery after stroke. In this review, we discuss the most recent science behind the role of cell-based therapies in ischemic stroke and the efforts to translate these therapies into human clinical trials. RECENT FINDINGS Preclinical data support numerous beneficial effects of cell-based therapies in both small and large animal models of ischemic stroke. These benefits are driven by multifaceted mechanisms promoting brain repair through immunomodulation, trophic support, circuit reorganization, and cell replacement. Cell-based therapies offer tremendous potential for improving outcomes after stroke through multimodal support of brain repair. Based on recent clinical trials, cell-based therapies appear both feasible and safe in all phases of stroke. Ongoing translational research and clinical trials will further refine these therapies and have the potential to transform the approach to stroke recovery and rehabilitation.
Collapse
Affiliation(s)
- Ania Dabrowski
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA
| | - Thomas J Robinson
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA
| | - Ryan J Felling
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA.
| |
Collapse
|
10
|
O'Rourke C, Day AGE, Murray-Dunning C, Thanabalasundaram L, Cowan J, Stevanato L, Grace N, Cameron G, Drake RAL, Sinden J, Phillips JB. An allogeneic 'off the shelf' therapeutic strategy for peripheral nerve tissue engineering using clinical grade human neural stem cells. Sci Rep 2018; 8:2951. [PMID: 29440680 PMCID: PMC5811594 DOI: 10.1038/s41598-018-20927-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
Artificial tissues constructed from therapeutic cells offer a promising approach for improving the treatment of severe peripheral nerve injuries. In this study the effectiveness of using CTX0E03, a conditionally immortalised human neural stem cell line, as a source of allogeneic cells for constructing living artificial nerve repair tissue was tested. CTX0E03 cells were differentiated then combined with collagen to form engineered neural tissue (EngNT-CTX), stable aligned sheets of cellular hydrogel. EngNT-CTX sheets were delivered within collagen tubes to repair a 12 mm sciatic nerve injury model in athymic nude rats. Autologous nerve grafts (autografts) and empty tubes were used for comparison. After 8 weeks functional repair was assessed using electrophysiology. Further, detailed histological and electron microscopic analysis of the repaired nerves was performed. Results indicated that EngNT-CTX supported growth of neurites and vasculature through the injury site and facilitated reinnervation of the target muscle. These findings indicate for the first time that a clinically validated allogeneic neural stem cell line can be used to construct EngNT. This provides a potential 'off the shelf' tissue engineering solution for the treatment of nerve injury, overcoming the limitations associated with nerve autografts or the reliance on autologous cells for populating repair constructs.
Collapse
Affiliation(s)
- C O'Rourke
- Department of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
- UCL Centre for Nerve Engineering, London, UK
| | - A G E Day
- Department of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
- UCL Centre for Nerve Engineering, London, UK
| | - C Murray-Dunning
- Department of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - L Thanabalasundaram
- UCL Centre for Nerve Engineering, London, UK
- ReNeuron, Pencoed, Bridgend, Wales, UK
| | - J Cowan
- Royal National Orthopaedic Hospital, Stanmore, UK
| | | | - N Grace
- Sartorius Stedim Biotech, Royston, UK
| | - G Cameron
- Sartorius Stedim Biotech, Royston, UK
| | | | - J Sinden
- UCL Centre for Nerve Engineering, London, UK
- ReNeuron, Pencoed, Bridgend, Wales, UK
| | - J B Phillips
- Department of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK.
- UCL Centre for Nerve Engineering, London, UK.
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, UK.
| |
Collapse
|
11
|
Daniszewski M, Crombie DE, Henderson R, Liang HH, Wong RCB, Hewitt AW, Pébay A. Automated Cell Culture Systems and Their Applications to Human Pluripotent Stem Cell Studies. SLAS Technol 2017; 23:315-325. [PMID: 28574793 DOI: 10.1177/2472630317712220] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Pluripotent stem cells are an extremely powerful tool in modeling human diseases and hold much promise for personalized regenerative or cell replacement therapies. There is an increasing need for reproducible large-scale stem cell and differentiated progeny production, with minimal variation, rendering manual approaches impracticable. Here, we provide an overview of systems currently available for automated stem cell culture, and undertake a review of their capacities, capabilities, and relative limitations. With the merging of modern technology and stem cell biology, an increased demand and implementation of automated platforms for stem cell studies is anticipated.
Collapse
Affiliation(s)
- Maciej Daniszewski
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Duncan E Crombie
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Rachael Henderson
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Helena H Liang
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Raymond C B Wong
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Alex W Hewitt
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia.,3 School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Alice Pébay
- 1 Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,2 Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Sinden JD, Hicks C, Stroemer P, Vishnubhatla I, Corteling R. Human Neural Stem Cell Therapy for Chronic Ischemic Stroke: Charting Progress from Laboratory to Patients. Stem Cells Dev 2017; 26:933-947. [PMID: 28446071 PMCID: PMC5510676 DOI: 10.1089/scd.2017.0009] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic disability after stroke represents a major unmet neurologic need. ReNeuron's development of a human neural stem cell (hNSC) therapy for chronic disability after stroke is progressing through early clinical studies. A Phase I trial has recently been published, showing no safety concerns and some promising signs of efficacy. A single-arm Phase II multicenter trial in patients with stable upper-limb paresis has recently completed recruitment. The hNSCs administrated are from a manufactured, conditionally immortalized hNSC line (ReNeuron's CTX0E03 or CTX), generated with c-mycERTAM technology. This technology has enabled CTX to be manufactured at large scale under cGMP conditions, ensuring sufficient supply to meets the demands of research, clinical development, and, eventually, the market. CTX has key pro-angiogenic, pro-neurogenic, and immunomodulatory characteristics that are mechanistically important in functional recovery poststroke. This review covers the progress of CTX cell therapy from its laboratory origins to the clinic, concluding with a look into the late stage clinical future.
Collapse
|
13
|
Irion S, Zabierowski SE, Tomishima MJ. Bringing Neural Cell Therapies to the Clinic: Past and Future Strategies. Mol Ther Methods Clin Dev 2017; 4:72-82. [PMID: 28344993 PMCID: PMC5363320 DOI: 10.1016/j.omtm.2016.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023]
Abstract
Cell replacement therapy in the nervous system has a rich history, with ∼40 years of research and ∼30 years of clinical experience. There is compelling evidence that appropriate cells can integrate and function in the dysfunctioning human nervous system, but the clinical results are mixed in practice. A number of factors conspire to vary patient outcome: the indication, cell source, patient selection, and team performing transplantation are all variables that can affect efficacy. Most early clinical trials have used fetal cells, a limited cell source that resists scale and standardization. Direct fetal cell transplantation creates significant challenges to commercialization that is the ultimate goal of an effective cell therapy. One approach to help scale and standardize fetal cell preparations is the expansion of neural cells in vitro. Expansion is achieved by transformation or through the application of mitogens before cryopreservation. Recently, neural cells derived from pluripotent stem cells have provided a scalable alternative. Pluripotent stem cells are desirable for manufacturing but present alternative concerns and manufacturing obstacles. All cell sources require robust and reproducible manufacturing to make nervous system cell replacement therapy an option for patients. Here, we discuss the challenges and opportunities for cell replacement in the nervous system. In this review, we give an overview of completed and ongoing neural cell transplantation clinical trials, and we discuss the challenges and opportunities for future cell replacement trials with a particular focus on pluripotent stem cell-derived therapies.
Collapse
Affiliation(s)
- Stefan Irion
- Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Susan E. Zabierowski
- Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA
- SKI Stem Cell Research Facility and Cell Therapy and Cell Engineering Facility, Sloan Kettering Institute, New York, NY 10065, USA
| | - Mark J. Tomishima
- Center for Stem Cell Biology, Sloan Kettering Institute, New York, NY 10065, USA
- SKI Stem Cell Research Facility, Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| |
Collapse
|
14
|
Kalladka D, Sinden J, Pollock K, Haig C, McLean J, Smith W, McConnachie A, Santosh C, Bath PM, Dunn L, Muir KW. Human neural stem cells in patients with chronic ischaemic stroke (PISCES): a phase 1, first-in-man study. Lancet 2016; 388:787-96. [PMID: 27497862 DOI: 10.1016/s0140-6736(16)30513-x] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND CTX0E03 is an immortalised human neural stem-cell line from which a drug product (CTX-DP) was developed for allogeneic therapy. Dose-dependent improvement in sensorimotor function in rats implanted with CTX-DP 4 weeks after middle cerebral artery occlusion stroke prompted investigation of the safety and tolerability of this treatment in stroke patients. METHODS We did an open-label, single-site, dose-escalation study. Men aged 60 years or older with stable disability (National Institutes of Health Stroke Scale [NIHSS] score ≥6 and modified Rankin Scale score 2-4) 6-60 months after ischaemic stroke were implanted with single doses of 2 million, 5 million, 10 million, or 20 million cells by stereotactic ipsilateral putamen injection. Clinical and brain imaging data were collected over 2 years. The primary endpoint was safety (adverse events and neurological change). This trial is registered with ClinicalTrials.gov, number NCT01151124. FINDINGS 13 men were recruited between September, 2010, and January, 2013, of whom 11 (mean age 69 years, range 60-82) received CTX-DP. Median NIHSS score before implantation was 7 (IQR 6-8) and the mean time from stroke was 29 (SD 14) months. Three men had subcortical infarcts only and seven had right-hemisphere infarcts. No immunological or cell-related adverse events were seen. Other adverse events were related to the procedure or comorbidities. Hyperintensity around the injection tracts on T2-weighted fluid-attenuation inversion recovery MRI was seen in five patients. At 2 years, improvement in NIHSS score ranged from 0 to 5 (median 2) points. INTERPRETATION Single intracerebral doses of CTX-DP up to 20 million cells induced no adverse events and were associated with improved neurological function. Our observations support further investigation of CTX-DP in stroke patients. FUNDING ReNeuron Limited.
Collapse
Affiliation(s)
- Dheeraj Kalladka
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - John Sinden
- ReNeuron Ltd, Surrey Research Park, Guildford, Surrey, UK
| | | | - Caroline Haig
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - John McLean
- Department of Neuroradiology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Wilma Smith
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Celestine Santosh
- Department of Neuroradiology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Laurence Dunn
- Neurosurgery, Institute of Neurological Sciences, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Keith W Muir
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK.
| |
Collapse
|
15
|
Savitz SI, Parsha K. Enhancing Stroke Recovery with Cellular Therapies. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
16
|
Potential of Neural Stem Cell-Based Therapy for Parkinson's Disease. PARKINSONS DISEASE 2015; 2015:571475. [PMID: 26664823 PMCID: PMC4664819 DOI: 10.1155/2015/571475] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/03/2015] [Indexed: 12/14/2022]
Abstract
Neural stem cell (NSC) transplantation is an emerging strategy for restoring neuronal function in neurological disorders, such as Parkinson's disease (PD), which is characterized by a profound and selective loss of nigrostriatal dopaminergic (DA) neurons. Adult neurogenesis generates newborn neurons that can be observed at specialized niches where endothelial cells (ECs) play a significant role in regulating the behavior of NSCs, including self-renewal and differentiating into all neural lineage cells. In this minireview, we highlight the importance of establishing an appropriate microenvironment at the target site of NSC transplantation, where grafted cells integrate into the surroundings in order to enhance DA neurotransmission. Using a novel model of NSC-EC coculture, it is possible to combine ECs with NSCs, to generate such a neurovascular microenvironment. With appropriate NSCs selected, the composition of the transplant can be investigated through paracrine and juxtacrine signaling within the neurovascular unit (NVU). With target site cellular and acellular compartments of the microenvironment recognized, guided DA differentiation of NSCs can be achieved. As differentiated DA neurons integrate into the existing nigrostriatal DA pathway, the symptoms of PD can potentially be alleviated by reversing characteristic neurodegeneration.
Collapse
|
17
|
Akiyama H, Kobayashi A, Ichimura M, Tone H, Nakatani M, Inoue M, Tojo A, Kagami H. Comparison of manual and automated cultures of bone marrow stromal cells for bone tissue engineering. J Biosci Bioeng 2015; 120:570-6. [DOI: 10.1016/j.jbiosc.2015.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 02/24/2015] [Accepted: 03/16/2015] [Indexed: 01/05/2023]
|
18
|
Heathman TRJ, Nienow AW, McCall MJ, Coopman K, Kara B, Hewitt CJ. The translation of cell-based therapies: clinical landscape and manufacturing challenges. Regen Med 2015; 10:49-64. [PMID: 25562352 DOI: 10.2217/rme.14.73] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapies have the potential to make a large contribution toward currently unmet patient need and thus effective manufacture of these products is essential. Many challenges must be overcome before this can become a reality and a better definition of the manufacturing requirements for cell-based products must be obtained. The aim of this study is to inform industry and academia of current cell-based therapy clinical development and to identify gaps in their manufacturing requirements. A total of 1342 active cell-based therapy clinical trials have been identified and characterized based on cell type, target indication and trial phase. Multiple technologies have been assessed for the manufacture of these cell types in order to facilitate product translation and future process development.
Collapse
Affiliation(s)
- Thomas R J Heathman
- Centre for Biological Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | | | | | | | | | | |
Collapse
|
19
|
Soares FAC, Chandra A, Thomas RJ, Pedersen RA, Vallier L, Williams DJ. Investigating the feasibility of scale up and automation of human induced pluripotent stem cells cultured in aggregates in feeder free conditions. J Biotechnol 2014; 173:53-8. [PMID: 24440272 PMCID: PMC3969287 DOI: 10.1016/j.jbiotec.2013.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/23/2013] [Accepted: 12/16/2013] [Indexed: 11/09/2022]
Abstract
First published protocol for scalable automation of hiPSC in feeder-free conditions. Successful transfer of hiPSC between sites representative of research and manufacture. Comparability between manual and automated expansion protocols for hiPSC.
The transfer of a laboratory process into a manufacturing facility is one of the most critical steps required for the large scale production of cell-based therapy products. This study describes the first published protocol for scalable automated expansion of human induced pluripotent stem cell lines growing in aggregates in feeder-free and chemically defined medium. Cells were successfully transferred between different sites representative of research and manufacturing settings; and passaged manually and using the CompacT SelecT automation platform. Modified protocols were developed for the automated system and the management of cells aggregates (clumps) was identified as the critical step. Cellular morphology, pluripotency gene expression and differentiation into the three germ layers have been used compare the outcomes of manual and automated processes.
Collapse
Affiliation(s)
- Filipa A C Soares
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine and Department of Surgery, University of Cambridge, UK; Centre for Biological Engineering, Loughborough University, UK.
| | - Amit Chandra
- Centre for Biological Engineering, Loughborough University, UK
| | - Robert J Thomas
- Centre for Biological Engineering, Loughborough University, UK
| | - Roger A Pedersen
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine and Department of Surgery, University of Cambridge, UK
| | - Ludovic Vallier
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine and Department of Surgery, University of Cambridge, UK; Wellcome Trust Sanger Institute, Hinxton, UK
| | | |
Collapse
|
20
|
Mondragon-Teran P, Tostoes R, Mason C, Lye GJ, Veraitch FS. Oxygen-controlled automated neural differentiation of mouse embryonic stem cells. Regen Med 2013; 8:171-82. [PMID: 23477397 DOI: 10.2217/rme.13.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Automation and oxygen tension control are two tools that provide significant improvements to the reproducibility and efficiency of stem cell production processes. AIM the aim of this study was to establish a novel automation platform capable of controlling oxygen tension during both the cell-culture and liquid-handling steps of neural differentiation processes. MATERIALS & METHODS We built a bespoke automation platform, which enclosed a liquid-handling platform in a sterile, oxygen-controlled environment. An airtight connection was used to transfer cell culture plates to and from an automated oxygen-controlled incubator. RESULTS Our results demonstrate that our system yielded comparable cell numbers, viabilities, metabolism profiles and differentiation efficiencies when compared with traditional manual processes. Interestingly, eliminating exposure to ambient conditions during the liquid-handling stage resulted in significant improvements in the yield of MAP2-positive neural cells, indicating that this level of control can improve differentiation processes. CONCLUSION This article describes, for the first time, an automation platform capable of maintaining oxygen tension control during both the cell-culture and liquid-handling stages of a 2D embryonic stem cell differentiation process.
Collapse
Affiliation(s)
- Paul Mondragon-Teran
- Biomedical Research Division, Centro Medico Nacional '20 de Noviembre' - ISSSTE. San Lorenzo 502, Del Valle, Benito Juarez, México City, 03229 México
| | | | | | | | | |
Collapse
|
21
|
Adams CF, Pickard MR, Chari DM. Magnetic nanoparticle mediated transfection of neural stem cell suspension cultures is enhanced by applied oscillating magnetic fields. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:737-41. [PMID: 23751375 DOI: 10.1016/j.nano.2013.05.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 12/18/2022]
Abstract
UNLABELLED Safe genetic modification of neural stem cell (NSC) transplant populations is a key goal for regenerative neurology. We describe a technically simple and safe method to increase transfection in NSCs propagated in the neurosphere (suspension culture) model, using magnetic nanoparticles deployed with applied oscillating magnetic fields ('magnetofection technology'). We show that transfection efficiency was enhanced over two-fold by oscillating magnetic fields (frequency=4 Hz). The protocols had no effect on cell viability, cell number, stem cell marker expression and differentiation profiles of 'magnetofected' cultures, highlighting the safety of the technique. As far as we are aware, this is the first successful application of magnetofection technology to suspension cultures of neural cells. The procedures described offer a means to augment the therapeutic potential of NSCs propagated as neurospheres - a culture model of high clinical translational relevance - by safe genetic manipulation, with further potential for incorporation into 'magneto-multifection' (repeat transfection) protocols. FROM THE CLINICAL EDITOR This team of investigators describe a simple and safe method to increase transfection in neural stem cells using magnetic nanoparticles deployed with oscillating magnetic fields, demonstrating a greater than two-fold transfection efficiency increase by applying low frequency magnetic oscillation.
Collapse
Affiliation(s)
- Chris F Adams
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, United Kingdom
| | | | | |
Collapse
|
22
|
Gorba T, Conti L. Neural stem cells as tools for drug discovery: novel platforms and approaches. Expert Opin Drug Discov 2013; 8:1083-94. [DOI: 10.1517/17460441.2013.805199] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
23
|
Mandenius CF. Biomechatronics for designing bioprocess monitoring and control systems: Application to stem cell production. J Biotechnol 2012; 162:430-40. [DOI: 10.1016/j.jbiotec.2012.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 07/23/2012] [Accepted: 09/03/2012] [Indexed: 12/26/2022]
|
24
|
McLaren D, Gorba T, Marguerie de Rotrou A, Pillai G, Chappell C, Stacey A, Lingard S, Falk A, Smith A, Koch P, Brüstle O, Vickers R, Tinsley J, Flanders D, Bello P, Craig S. Automated large-scale culture and medium-throughput chemical screen for modulators of proliferation and viability of human induced pluripotent stem cell-derived neuroepithelial-like stem cells. ACTA ACUST UNITED AC 2012; 18:258-68. [PMID: 23042076 DOI: 10.1177/1087057112461446] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The aim of this study was to demonstrate proof-of-concept feasibility for the use of human neural stem cells (NSCs) for high-throughput screening (HTS) applications. For this study, an adherent human induced pluripotent stem (iPS) cell-derived long-term, self-renewing, neuroepithelial-like stem (lt-NES) cell line was selected as a representative NSC. Here, we describe the automated large-scale serum-free culture ("scale-up") of human lt-NES cells on the CompacT SelecT cell culture robotic platform, followed by their subsequent automated "scale-out" into a microwell plate format. We also report a medium-throughput screen of 1000 compounds to identify modulators of neural stem cell proliferation and/or survival. The screen was performed on two independent occasions using a cell viability assay with end-point reading resulting in the identification of 24 potential hit compounds, 5 of which were found to increase the proliferation and/or survival of human lt-NES on both occasions. Follow-up studies confirmed a dose-dependent effect of one of the hit compounds, which was a Cdk-2 modulator. This approach could be further developed as part of a strategy to screen compounds to either improve the procedures for the in vitro expansion of neural stem cells or to potentially modulate endogenous neural stem cell behavior in the diseased nervous system.
Collapse
|
25
|
Williams DJ, Thomas RJ, Hourd PC, Chandra A, Ratcliffe E, Liu Y, Rayment EA, Archer JR. Precision manufacturing for clinical-quality regenerative medicines. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2012; 370:3924-3949. [PMID: 22802496 DOI: 10.1098/rsta.2011.0049] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Innovations in engineering applied to healthcare make a significant difference to people's lives. Market growth is guaranteed by demographics. Regulation and requirements for good manufacturing practice-extreme levels of repeatability and reliability-demand high-precision process and measurement solutions. Emerging technologies using living biological materials add complexity. This paper presents some results of work demonstrating the precision automated manufacture of living materials, particularly the expansion of populations of human stem cells for therapeutic use as regenerative medicines. The paper also describes quality engineering techniques for precision process design and improvement, and identifies the requirements for manufacturing technology and measurement systems evolution for such therapies.
Collapse
Affiliation(s)
- David J Williams
- Healthcare Engineering Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Stroke, for some years now the neglected major indication in the pharmaceutical development cupboard, has recently become one of the hot areas for stem cell therapy development. This is driven by better understanding of potential therapeutic opportunities both in the acute and chronic phases and the launch of a series of new early phase clinical trials in a number of countries, driven by positive data in relevant animal models. In addition, the impetus for stem cell product development is motivated by patient demand, with thousands of victims seeking unproven treatments abroad. This article looks at the many challenges facing the development of a stem cell therapy for stroke. These range from product characterization and banking, through nonclinical safety and efficacy to the regulatory requirements for starting patient trials and beyond to maximizing value from carefully designed efficacy trials.
Collapse
Affiliation(s)
| | - Keith W. Muir
- Institute of Neuroscience and Psychology, University of Glasgow, Scotland, UK
| |
Collapse
|
27
|
Feline Neural Progenitor Cells I: Long-Term Expansion under Defined Culture Conditions. Stem Cells Int 2012; 2012:108340. [PMID: 22577394 PMCID: PMC3347782 DOI: 10.1155/2012/108340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 02/03/2012] [Indexed: 11/23/2022] Open
Abstract
Neural progenitor cells (NPCs) of feline origin (cNPCs) have demonstrated utility in transplantation experiments, yet are difficult to grow in culture beyond the 1 month time frame. Here we use an enriched, serum-free base medium (Ultraculture) and report the successful long-term propagation of these cells. Primary cultures were derived from fetal brain tissue and passaged in DMEM/F12-based or Ultraculture-based proliferation media, both in the presence of EGF + bFGF. Cells in standard DMEM/F12-based medium ceased to proliferate by 1-month, whereas the cells in the Ultraculture-based medium continued to grow for at least 5 months (end of study) with no evidence of senescence. The Ultraculture-based cultures expressed lower levels of progenitor and lineage-associated markers under proliferation conditions but retained multipotency as evidenced by the ability to differentiate into neurons and glia following growth factor removal in the presence of FBS. Importantly, later passage cNPCs did not develop chromosomal aberrations.
Collapse
|
28
|
Smith EJ, Stroemer RP, Gorenkova N, Nakajima M, Crum WR, Tang E, Stevanato L, Sinden JD, Modo M. Implantation Site and Lesion Topology Determine Efficacy of a Human Neural Stem Cell Line in a Rat Model of Chronic Stroke. Stem Cells 2012; 30:785-96. [DOI: 10.1002/stem.1024] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
29
|
Abstract
Human cell culture processes developed at research laboratory scale need to be translated to large-scale production processes to achieve commercial application to a large market. To allow this transition of scale with consistent process performance and control of costs, it will be necessary to reduce manual processing and increase automation. There are a number of commercially available platforms that will reduce manual process intervention and improve process control for different culture formats. However, in many human cell-based applications, there is currently a need to remain close to the development format, usually adherent culture on cell culture plastic or matrix-coated wells or flasks due to deterioration of cell quality in other environments, such as suspension. This chapter presents an example method for adherent automated human stem cell culture using a specific automated flask handling platform, the CompacT SelecT.
Collapse
Affiliation(s)
- Robert Thomas
- Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Leicestershire, UK.
| | | |
Collapse
|
30
|
Hunt CJ. Cryopreservation of Human Stem Cells for Clinical Application: A Review. Transfus Med Hemother 2011; 38:107-123. [PMID: 21566712 PMCID: PMC3088734 DOI: 10.1159/000326623] [Citation(s) in RCA: 226] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 01/26/2011] [Indexed: 12/13/2022] Open
Abstract
SUMMARY: Stem cells have been used in a clinical setting for many years. Haematopoietic stem cells have been used for the treatment of both haematological and non-haematological disease; while more recently mesenchymal stem cells derived from bone marrow have been the subject of both laboratory and early clinical studies. Whilst these cells show both multipotency and expansion potential, they nonetheless do not form stable cell lines in culture which is likely to limit the breadth of their application in the field of regenerative medicine. Human embryonic stem cells are pluripotent cells, capable of forming stable cell lines which retain the capacity to differentiate into cells from all three germ layers. This makes them of special significance in both regenerative medicine and toxicology. Induced pluripotent stem (iPS) cells may also provide a similar breadth of utility without some of the confounding ethical issues surrounding embryonic stem cells. An essential pre-requisite to the commercial and clinical application of stem cells are suitable cryopreservation protocols for long-term storage. Whilst effective methods for cryopreservation and storage have been developed for haematopoietic and mesenchymal stem cells, embryonic cells and iPS cells have proved more refractory. This paper reviews the current state of cryopreservation as it pertains to stem cells and in particular the embryonic and iPS cell.
Collapse
Affiliation(s)
- Charles J. Hunt
- UK Stem Cell Bank, National Institute for Biological Standards and Control, Health Protection Agency, South Mimms, Potters Bar, UK
| |
Collapse
|
31
|
Ratcliffe E, Thomas RJ, Williams DJ. Current understanding and challenges in bioprocessing of stem cell-based therapies for regenerative medicine. Br Med Bull 2011; 100:137-55. [PMID: 21852279 DOI: 10.1093/bmb/ldr037] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND A novel manufacturing industry is emerging to translate unique cellular therapy bioprocesses to robust, scaled manufacturing production for successful clinical translation. SOURCE OF DATA This review summarizes key translational issues, and current and future perspectives to improve translation of cell-based therapy bioprocessing, based on literature search and author research. AREAS OF AGREEMENT It is widely recognized that cell-based therapies could revolutionize health care for a range of diseases, and that there are gaps in the overarching framework and technologies to generate clinical success. AREAS OF CONTROVERSY There is limited understanding of how to fulfil requirements as regulatory and manufacturing guidelines are incomplete and few have achieved commercialization. GROWING POINTS Recent developments are encouraging adoption of automation and quality engineering approaches for bioprocessing of cell-based therapies. AREAS TIMELY FOR DEVELOPING RESEARCH Include technology development to improve the cost and purity of manufacture and final product quality.
Collapse
Affiliation(s)
- Elizabeth Ratcliffe
- Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, LE11 3TU, UK.
| | | | | |
Collapse
|