1
|
Kostenko A, Connon CJ, Swioklo S. Storable Cell-Laden Alginate Based Bioinks for 3D Biofabrication. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010023. [PMID: 36671596 PMCID: PMC9854877 DOI: 10.3390/bioengineering10010023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 12/06/2022] [Indexed: 12/28/2022]
Abstract
Over the last decade, progress in three dimensional (3D) bioprinting has advanced considerably. The ability to fabricate complex 3D structures containing live cells for drug discovery and tissue engineering has huge potential. To realise successful clinical translation, biologistics need to be considered. Refinements in the storage and transportation process from sites of manufacture to the clinic will enhance the success of future clinical translation. One of the most important components for successful 3D printing is the 'bioink', the cell-laden biomaterial used to create the printed structure. Hydrogels are favoured bioinks used in extrusion-based bioprinting. Alginate, a natural biopolymer, has been widely used due to its biocompatibility, tunable properties, rapid gelation, low cost, and easy modification to direct cell behaviour. Alginate has previously demonstrated the ability to preserve cell viability and function during controlled room temperature (CRT) storage and shipment. The novelty of this research lies in the development of a simple and cost-effective hermetic system whereby alginate-encapsulated cells can be stored at CRT before being reformulated into an extrudable bioink for on-demand 3D bioprinting of cell-laden constructs. To our knowledge the use of the same biomaterial (alginate) for storage and on-demand 3D bio-printing of cells has not been previously investigated. A straightforward four-step process was used where crosslinked alginate containing human adipose-derived stem cells was stored at CRT before degelation and subsequent mixing with a second alginate. The printability of the resulting bioink, using an extrusion-based bioprinter, was found to be dependent upon the concentration of the second alginate, with 4 and 5% (w/v) being optimal. Following storage at 15 °C for one week, alginate-encapsulated human adipose-derived stem cells exhibited a high viable cell recovery of 88 ± 18%. Stored cells subsequently printed within 3D lattice constructs, exhibited excellent post-print viability and even distribution. This represents a simple, adaptable method by which room temperature storage and biofabrication can be integrated for on-demand bioprinting.
Collapse
Affiliation(s)
- Anastassia Kostenko
- Atelerix Ltd, The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK
- International Centre for Life, Faculty of Medicine, Bioscience Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Che J. Connon
- Atelerix Ltd, The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK
- International Centre for Life, Faculty of Medicine, Bioscience Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
- Correspondence: ; Tel.: +44-(0)-191-241-8623
| | - Stephen Swioklo
- Atelerix Ltd, The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK
| |
Collapse
|
2
|
Lam C, Meinert E, Yang A, Cui Z. Impact of fast-track regulatory designations on strategic commercialization decisions for autologous cell therapies. Regen Med 2022; 17:155-174. [PMID: 35073729 DOI: 10.2217/rme-2021-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Regulatory authorities around the world have introduced incentives to improve the speed-to-market of innovative therapies. Aim & methods: To better understand the capacity and portfolio planning decisions of autologous cell therapies and particularly the impact of fast-tracking designations, this paper describes a mixed-integer linear programming approach for the optimization of capacity investment and portfolio selection decisions to maximize the net present value of a candidate portfolio of therapies under different regulatory programs. Results: The illustrative example shows that fast-track designations allow a 25% earlier breakeven, 42-86% higher net present value over a 20-year horizon with earlier upfront capital and reduce the portfolio's sensitivity to uncertainties. Conclusion: Fast-track designations are effective in providing commercialization incentives, but high capital risks given the compressed timeline should be better considered.
Collapse
Affiliation(s)
- Ching Lam
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, OX3 7DQ, UK
| | - Edward Meinert
- Department of Paediatrics, University of Oxford, Oxford, OX3 9DU, UK
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, Oxford, OX1 3PJ, UK
| | - Zhanfeng Cui
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
3
|
Rallapalli S, Guhathakurta S, Bishi DK, Subbarayan R, Mathapati S, Korrapati PS. A critical appraisal of humanized alternatives to fetal bovine serum for clinical applications of umbilical cord derived mesenchymal stromal cells. Biotechnol Lett 2021; 43:2067-2083. [PMID: 34499291 DOI: 10.1007/s10529-021-03180-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The study is aimed to verify the possibility of using humanized alternatives to fetal bovine serum (FBS) such as umbilical cord blood plasma (CBP) and AB+ plasma to support the long-term growth of mesenchymal stromal cells (MSCs) derived from the umbilical cord. We hypothesized that umbilical CBP would be a potential substitute to FBS, especially for small scale autologous clinical transplantations. METHODS The MSCs were cultured for six consecutive passages to evaluate xeno-free media's ability to support long-term growth. Cell proliferation rates, colony-forming-unit (CFU) efficiency and population doublings of expanded MSCs, were investigated. Ex vivo expanded MSCs were further characterized using flow cytometry and quantitative PCR. The impact of cryopreservation and composition of cryomedium on phenotype, viability of MSC was also assessed. RESULTS Our results on cell proliferation, colony-forming unit efficiency suggested that the expansion of the cells was successfully carried out in media supplemented with humanized alternatives. MSCs showed lower CFU counts in FBS (~ 25) than humanized alternatives (~ 35). The gene expression analysis revealed that transcripts showed significant differential expression by two to three folds in the FBS group compared with MSCs grown in medium with humanized alternatives (p < 0.05). In addition, MSCs grown in a medium with FBS had more osteogenic activity, a signature of unwanted differentiation. The majority of ex vivo expanded MSCs at early and late passages expressed CD44+, CD73+, CD105+, CD90+, and CD166+ in all the experimental groups tested (~ 90%). In contrast to the other MSC surface markers, expression levels of STRO-1+ (~ 21-10%) and TNAP+ (~ 29-11%) decreased with the increase in passage number for MSCs cultured in a FBS-supplemented medium (p < 0.05). CONCLUSION Our results established that CBP supported culture of umbilical cord tissue-derived MSCs and is a safer Xeno free replacement to FBS. The use of CBP also enables the storage of umbilical cord tissue derived MSCs in patient-specific conditions to minimize adverse events if cells are delivered directly to the patient.
Collapse
Affiliation(s)
- Suneel Rallapalli
- Biological Material Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, 600020, India
| | | | - Dillip Kumar Bishi
- Department of Biotechnology, Rama Devi Women's University, Bhubaneswar, India
| | | | - Santosh Mathapati
- Translational Health Science and Technology Institute, Faridabad, India
| | - Purna Sai Korrapati
- Biological Material Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, 600020, India.
| |
Collapse
|
4
|
The evolution of commercial drug delivery technologies. Nat Biomed Eng 2021; 5:951-967. [PMID: 33795852 DOI: 10.1038/s41551-021-00698-w] [Citation(s) in RCA: 498] [Impact Index Per Article: 166.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Drug delivery technologies have enabled the development of many pharmaceutical products that improve patient health by enhancing the delivery of a therapeutic to its target site, minimizing off-target accumulation and facilitating patient compliance. As therapeutic modalities expanded beyond small molecules to include nucleic acids, peptides, proteins and antibodies, drug delivery technologies were adapted to address the challenges that emerged. In this Review Article, we discuss seminal approaches that led to the development of successful therapeutic products involving small molecules and macromolecules, identify three drug delivery paradigms that form the basis of contemporary drug delivery and discuss how they have aided the initial clinical successes of each class of therapeutic. We also outline how the paradigms will contribute to the delivery of live-cell therapies.
Collapse
|
5
|
Grant R, Coopman K, Silva-Gomes S, Campbell JJ, Kara B, Braybrook J, Petzing J. Assessment of Protocol Impact on Subjectivity Uncertainty When Analyzing Peripheral Blood Mononuclear Cell Flow Cytometry Data Files. Methods Protoc 2021; 4:24. [PMID: 33808088 PMCID: PMC8103269 DOI: 10.3390/mps4020024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/21/2022] Open
Abstract
Measured variability of product within Cell and Gene Therapy (CGT) manufacturing arises from numerous sources across pre-analytical to post-analytical phases of testing. Operators are a function of the manufacturing process and are an important source of variability as a result of personal differences impacted by numerous factors. This research uses measurement uncertainty in comparison to Coefficient of Variation to quantify variation of participants when they complete Flow Cytometry data analysis through a 5-step gating sequence. Two study stages captured participants applying gates using their own judgement, and then following a diagrammatical protocol, respectively. Measurement uncertainty was quantified for each participant (and analysis phase) by following Guide to the Expression of Uncertainty in Measurement protocols, combining their standard deviations in quadrature from each gating step in the respective protocols. When participants followed a diagrammatical protocol, variation between participants reduced by 57%, increasing confidence in a more uniform reported cell count percentage. Measurement uncertainty provided greater resolution to the analysis processes, identifying that most variability contributed in the Flow Cytometry gating process is from the very first gate, where isolating target cells from dead or dying cells is required. This work has demonstrated the potential for greater usage of measurement uncertainty within CGT manufacturing scenarios, due to the resolution it provides for root cause analysis and continuous improvement.
Collapse
Affiliation(s)
- Rebecca Grant
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK;
| | - Karen Coopman
- Department of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK;
| | - Sandro Silva-Gomes
- GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK; (S.S.-G.); (B.K.)
| | | | - Bo Kara
- GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK; (S.S.-G.); (B.K.)
| | - Julian Braybrook
- LGC Group, Queen’s Road, Teddington, Middlesex TW11 0LY, UK; (J.J.C.); (J.B.)
| | - Jon Petzing
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK;
| |
Collapse
|
6
|
Lam C, Meinert E, Yang A, Cui Z. Comparison between centralized and decentralized supply chains of autologous chimeric antigen receptor T-cell therapies: a UK case study based on discrete event simulation. Cytotherapy 2021; 23:433-451. [PMID: 33674239 DOI: 10.1016/j.jcyt.2020.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/21/2020] [Accepted: 08/16/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND AIMS Decentralized, or distributed, manufacturing that takes place close to the point of care has been a manufacturing paradigm of heightened interest within the cell therapy domain because of the product's being living cell material as well as the need for a highly monitored and temperature-controlled supply chain that has the potential to benefit from close proximity between manufacturing and application. METHODS To compare the operational feasibility and cost implications of manufacturing autologous chimeric antigen receptor T (CAR T)-cell products between centralized and decentralized schemes, a discrete event simulation model was built using ExtendSIM 9 for simulating the patient-to-patient supply chain, from the collection of patient cells to the final administration of CAR T therapy in hospitals. Simulations were carried out for hypothetical systems in the UK using three demand levels-low (100 patients per annum), anticipated (200 patients per annum) and high (500 patients per annum)-to assess resource allocation, cost per treatment and system resilience to demand changes and to quantify the risks of mix-ups within the supply chain for the delivery of CAR T treatments. RESULTS The simulation results show that although centralized manufacturing offers better economies of scale, individual facilities in a decentralized system can spread facility costs across a greater number of treatments and better utilize resources at high demand levels (annual demand of 500 patients), allowing for an overall more comparable cost per treatment. In general, raw material and consumable costs have been shown to be one of the greatest cost drivers, and genetic modification-associated costs have been shown to account for over one third of raw material and consumable costs. Turnaround time per treatment for the decentralized scheme is shown to be consistently lower than its centralized counterpart, as there is no need for product freeze-thaw, packaging and transportation, although the time savings is shown to be insignificant in the UK case study because of its rather compact geographical setting with well-established transportation networks. In both schemes, sterility testing lies on the critical path for treatment delivery and is shown to be critical for treatment turnaround time reduction. CONCLUSIONS Considering both cost and treatment turnaround time, point-of-care manufacturing within the UK does not show great advantages over centralized manufacturing. However, further simulations using this model can be used to understand the feasibility of decentralized manufacturing in a larger geographical setting.
Collapse
Affiliation(s)
- Ching Lam
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Edward Meinert
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Estimation of manufacturing development costs of cell-based therapies: a feasibility study. Cytotherapy 2021; 23:730-739. [PMID: 33593688 DOI: 10.1016/j.jcyt.2020.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND AIMS Cell-based therapies (CBTs) provide opportunities to treat rare and high-burden diseases. Manufacturing development of these innovative products is said to be complex and costly. However, little research is available providing insight into resource use and cost drivers. Therefore, this study aimed to assess the feasibility of estimating the cost of manufacturing development of two cell-based therapy case studies using a CBT cost framework specifically designed for small-scale cell-based therapies. METHODS A retrospective costing study was conducted in which the cost of developing an adoptive immunotherapy of Epstein-Barr virus-specific cytotoxic T lymphocytes (CTLs) and a pluripotent stem cell (PSC) master cell bank was estimated. Manufacturing development was defined as products advancing from technology readiness level 3 to 6. The study was conducted in a Scottish facility. Development steps were recreated via developer focus groups. Data were collected from facility administrative and financial records and developer interviews. RESULTS Application of the manufacturing cost framework to retrospectively estimate the manufacturing design cost of two case studies in one Scottish facility appeared feasible. Manufacturing development cost was estimated at £1,201,016 for CTLs and £494,456 for PSCs. Most costs were accrued in the facility domain (56% and 51%), followed by personnel (20% and 32%), materials (19% and 15%) and equipment (4% and 2%). CONCLUSIONS Based on this study, it seems feasible to retrospectively estimate resources consumed in manufacturing development of cell-based therapies. This fosters inclusion of cost in the formulation and dissemination of best practices to facilitate early and sustainable patient access and inform future cost-conscious manufacturing design decisions.
Collapse
|
8
|
Lam C, van Velthoven MH, Meinert E. Developing a Blockchain-Based Supply Chain System for Advanced Therapies: Protocol for a Feasibility Study. JMIR Res Protoc 2020; 9:e17005. [PMID: 33315020 PMCID: PMC7769686 DOI: 10.2196/17005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 07/20/2020] [Accepted: 09/17/2020] [Indexed: 11/13/2022] Open
Abstract
Background Advanced therapies, including cell and gene therapies, have shown therapeutic promise in curing life-threatening diseases, such as leukemia and lymphoma. However, these therapies can be complicated and expensive to deliver due to their sensitivity to environment; troublesome tissue, cell, or genetic material sourcing; and complicated regulatory requirements. Objective This study aims to create a novel connected supply chain logistics and manufacturing management platform based on blockchain, with cell and gene therapy as a use case. Objectives are to define the requirements and perform feasibility evaluations on the use of blockchain for standardized manufacturing and establishment of a chain of custody for the needle-to-needle delivery of autologous cell and gene therapies. A way of lowering overall regulatory compliance costs for running a network of facilities operating similar or parallel processes will be evaluated by lowering the monitoring costs through publishing zero-knowledge proofs and product release by exception. Methods The study will use blockchain technologies to digitally connect and integrate supply chain with manufacturing to address the security, scheduling, and communication issues between advanced therapy treatment centers and manufacturing facilities in order to realize a transparent, secure, automated, and cost-effective solution to the delivery of these life-saving therapies. An agile software development methodology will be used to develop, implement, and evaluate the system. The system will adhere to the EU and US good manufacturing practices and regulatory requirements. Results This is a proposed study protocol, and upon acceptance, grant funding will be pursued for its execution in 2021. Conclusions The successful implementation of the integrated blockchain solution to supply chain and manufacturing of advanced therapies can push the industry standards toward a safer and more secure therapy delivery process. International Registered Report Identifier (IRRID) PRR1-10.2196/17005
Collapse
Affiliation(s)
- Ching Lam
- Digitally Enabled PrevenTative Health (DEPTH) Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Michelle Helena van Velthoven
- Digitally Enabled PrevenTative Health (DEPTH) Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Edward Meinert
- Digitally Enabled PrevenTative Health (DEPTH) Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,Department of Primary Care and Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
van Overbeeke E, Michelsen S, Toumi M, Stevens H, Trusheim M, Huys I, Simoens S. Market access of gene therapies across Europe, USA, and Canada: challenges, trends, and solutions. Drug Discov Today 2020; 26:399-415. [PMID: 33242695 DOI: 10.1016/j.drudis.2020.11.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 11/19/2020] [Indexed: 01/19/2023]
Abstract
This review can inform gene therapy developers on challenges that can be encountered when seeking market access. Moreover, it provides an overview of trends among challenges and potential solutions.
Collapse
Affiliation(s)
- Eline van Overbeeke
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium.
| | - Sissel Michelsen
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium; Healthcare Management Centre, Vlerick Business School, Reep 1, 9000 Ghent, Belgium
| | - Mondher Toumi
- Public Health Department, Aix Marseille University, 27 bd Jean Moulin, Marseille, France
| | - Hilde Stevens
- Institute for Interdisciplinary Innovation in Healthcare (I(3)h), Université libre de Bruxelles, Route de Lennik 808, Brussels, Belgium
| | - Mark Trusheim
- Massachusetts Institute of Technology, 100 Main Street, Cambridge, MA 02139, USA
| | - Isabelle Huys
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium
| | - Steven Simoens
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium
| |
Collapse
|
10
|
Sanicola HW, Stewart CE, Mueller M, Ahmadi F, Wang D, Powell SK, Sarkar K, Cutbush K, Woodruff MA, Brafman DA. Guidelines for establishing a 3-D printing biofabrication laboratory. Biotechnol Adv 2020; 45:107652. [PMID: 33122013 DOI: 10.1016/j.biotechadv.2020.107652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022]
Abstract
Advanced manufacturing and 3D printing are transformative technologies currently undergoing rapid adoption in healthcare, a traditionally non-manufacturing sector. Recent development in this field, largely enabled by merging different disciplines, has led to important clinical applications from anatomical models to regenerative bioscaffolding and devices. Although much research to-date has focussed on materials, designs, processes, and products, little attention has been given to the design and requirements of facilities for enabling clinically relevant biofabrication solutions. These facilities are critical to overcoming the major hurdles to clinical translation, including solving important issues such as reproducibility, quality control, regulations, and commercialization. To improve process uniformity and ensure consistent development and production, large-scale manufacturing of engineered tissues and organs will require standardized facilities, equipment, qualification processes, automation, and information systems. This review presents current and forward-thinking guidelines to help design biofabrication laboratories engaged in engineering model and tissue constructs for therapeutic and non-therapeutic applications.
Collapse
Affiliation(s)
- Henry W Sanicola
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Caleb E Stewart
- Department of Neurosurgery, Louisiana State Health Sciences Center, Shreveport, LA 71103, USA.
| | | | - Farzad Ahmadi
- Department of Electrical and Computer Engineering, Youngstown State University, Youngstown, OH 44555, USA
| | - Dadong Wang
- Quantitative Imaging Research Team, Data61, Commonwealth Scientific and Industrial Research Organization, Marsfield, NSW 2122, Australia
| | - Sean K Powell
- Science and Engineering Faculty, Queensland University of Technology, Brisbane 4029, Australia
| | - Korak Sarkar
- M3D Laboratory, Ochsner Health System, New Orleans, LA 70121, USA
| | - Kenneth Cutbush
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Maria A Woodruff
- Science and Engineering Faculty, Queensland University of Technology, Brisbane 4029, Australia.
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
11
|
Emerson J, Kara B, Glassey J. Multivariate data analysis in cell gene therapy manufacturing. Biotechnol Adv 2020; 45:107637. [PMID: 32980438 DOI: 10.1016/j.biotechadv.2020.107637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/27/2020] [Accepted: 09/22/2020] [Indexed: 01/26/2023]
Abstract
The emergence of cell gene therapy (CGT) as a safe and efficacious treatment for numerous severe inherited and acquired human diseases has led to growing interest and investment in new CGT products. The most successful of these have been autologous viral vector-based treatments. The development of viral vector manufacturing processes and ex vivo patient cell processing capabilities is a pressing issue in the advancement of autologous viral vector-based CGT treatments. In viral vector production, scale-up is a critical task due to the limited scalability of traditional laboratory systems and the demand for high volumes of viral vector manufactured in accordance with current good manufacturing practice. Ex vivo cell processing methods require optimisation and automation before they can be scaled out, and several other manufacturing challenges are prevalent such as high levels of raw material and process variability, difficulty characterising complex materials, and a lack of knowledge of critical process parameters and their effect on critical quality attributes of the viral vector and cell drug products. Multivariate data analysis (MVDA) has been leveraged successfully in a variety of applications in the chemical and biochemical industries, including for tasks such as bioprocess monitoring, identification of critical process parameters and assessment of process variability and comparability during process development, scale-up and technology transfer. Henceforth, MVDA is reviewed here as a suitable tool for tackling some of the challenges faced in the development of CGT manufacturing processes. A summary of some key CGT manufacturing challenges is provided along with a review of MVDA applications to mammalian and microbial processes, and an exploration of the potential benefits, requirements and pre-requisites of MVDA applications in the development of CGT manufacturing processes.
Collapse
Affiliation(s)
- Joseph Emerson
- School of Engineering, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| | - Bo Kara
- Currently, Evox Therapeutics, Medawar Centre, Oxford OX4 4HG, UK.
| | - Jarka Glassey
- School of Engineering, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
12
|
Childs PG, Reid S, Salmeron-Sanchez M, Dalby MJ. Hurdles to uptake of mesenchymal stem cells and their progenitors in therapeutic products. Biochem J 2020; 477:3349-3366. [PMID: 32941644 PMCID: PMC7505558 DOI: 10.1042/bcj20190382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022]
Abstract
Twenty-five years have passed since the first clinical trial utilising mesenchymal stomal/stem cells (MSCs) in 1995. In this time academic research has grown our understanding of MSC biochemistry and our ability to manipulate these cells in vitro using chemical, biomaterial, and mechanical methods. Research has been emboldened by the promise that MSCs can treat illness and repair damaged tissues through their capacity for immunomodulation and differentiation. Since 1995, 31 therapeutic products containing MSCs and/or progenitors have reached the market with the level of in vitro manipulation varying significantly. In this review, we summarise existing therapeutic products containing MSCs or mesenchymal progenitor cells and examine the challenges faced when developing new therapeutic products. Successful progression to clinical trial, and ultimately market, requires a thorough understanding of these hurdles at the earliest stages of in vitro pre-clinical development. It is beneficial to understand the health economic benefit for a new product and the reimbursement potential within various healthcare systems. Pre-clinical studies should be selected to demonstrate efficacy and safety for the specific clinical indication in humans, to avoid duplication of effort and minimise animal usage. Early consideration should also be given to manufacturing: how cell manipulation methods will integrate into highly controlled workflows and how they will be scaled up to produce clinically relevant quantities of cells. Finally, we summarise the main regulatory pathways for these clinical products, which can help shape early therapeutic design and testing.
Collapse
Affiliation(s)
- Peter G. Childs
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, U.K
- Centre for the Cellular Microenvironment, SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, U.K
| | - Stuart Reid
- Centre for the Cellular Microenvironment, SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, U.K
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Matthew J. Dalby
- Centre for the Cellular Microenvironment, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| |
Collapse
|
13
|
Dosta P, Ferber S, Zhang Y, Wang K, Ros A, Uth N, Levinson Y, Abraham E, Artzi N. Scale-up manufacturing of gelatin-based microcarriers for cell therapy. J Biomed Mater Res B Appl Biomater 2020; 108:2937-2949. [PMID: 32356942 DOI: 10.1002/jbm.b.34624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/11/2020] [Accepted: 04/13/2020] [Indexed: 12/20/2022]
Abstract
Microcarriers, including crosslinked porous gelatin beads (Cultispher G) are widely used as cell carriers for cell therapy applications. Microcarriers can support a range of adherent cell types in stirred tank bioreactor culture, which is scalable up to several thousands of liters. Cultispher G in particular is advantageous for cell therapy applications because it can be dissolved enzymatically, and thus cells can be harvested without the need to perform a large-scale cell-bead filtration step. This enzymatic dissolution, however, is challenged by the slow degradation of the carriers in the presence of enzymes as new extracellular matrix is being deposited by the proliferating cells. This extended dissolution timelimits the yield of cell recovery while compromising cellular viability. We report herein the development of crosslinked porous gelatin beads that afford rapid, stimuli-triggered dissolution for facile cell removal using human mesenchymal stem cells (hMSC) as a model system. We successfully fabricated redox-sensitive beads (RS beads) and studied their cell growth, dissolution time and cell yield, compared to regular gelatin-based beads (Reg beads). We have shown that RS beads allow for much faster dissolution compared to Reg beads, supporting better hMSC detachment and recovery following 8 days of culture in spinner flasks, or in 3L bioreactors. These newly synthesized RS beads show promise as cellular microcarriers and can be used for scale-up manufacturing of different cell types while providing on-demand degradation for facile cell retrieval.
Collapse
Affiliation(s)
- Pere Dosta
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shiran Ferber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi Zhang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kui Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Albert Ros
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas Uth
- Research and Technology, Walkersville, Maryland, USA
| | | | - Eytan Abraham
- Research and Technology, Walkersville, Maryland, USA
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
14
|
Shariatzadeh M, Chandra A, Wilson SL, McCall MJ, Morizur L, Lesueur L, Chose O, Gepp MM, Schulz A, Neubauer JC, Zimmermann H, Abranches E, Man J, O’Shea O, Stacey G, Hewitt Z, Williams DJ. Distributed automated manufacturing of pluripotent stem cell products. THE INTERNATIONAL JOURNAL, ADVANCED MANUFACTURING TECHNOLOGY 2020; 106:1085-1103. [PMID: 31983799 PMCID: PMC6954896 DOI: 10.1007/s00170-019-04516-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/27/2019] [Indexed: 05/04/2023]
Abstract
Establishing how to effectively manufacture cell therapies is an industry-level problem. Decentralised manufacturing is of increasing importance, and its challenges are recognised by healthcare regulators with deviations and comparability issues receiving specific attention from them. This paper is the first to report the deviations and other risks encountered when implementing the expansion of human pluripotent stem cells (hPSCs) in an automated three international site-decentralised manufacturing setting. An experimental demonstrator project expanded a human embryonal carcinoma cell line (2102Ep) at three development sites in France, Germany and the UK using the CompacT SelecT (Sartorius Stedim, Royston, UK) automated cell culture platform. Anticipated variations between sites spanned material input, features of the process itself and production system details including different quality management systems and personnel. Where possible, these were pre-addressed by implementing strategies including standardisation, cell bank mycoplasma testing and specific engineering and process improvements. However, despite such measures, unexpected deviations occurred between sites including software incompatibility and machine/process errors together with uncharacteristic contaminations. Many only became apparent during process proving or during the process run. Further, parameters including growth rate and viability discrepancies could only be determined post-run, preventing 'live' corrective measures. The work confirms the critical nature of approaches usually taken in Good Manufacturing Practice (GMP) manufacturing settings and especially emphasises the requirement for monitoring steps to be included within the production system. Real-time process monitoring coupled with carefully structured quality systems is essential for multiple site working including clarity of decision-making roles. Additionally, an over-reliance upon post-process visual microscopic comparisons has major limitations; it is difficult for non-experts to detect deleterious culture changes and such detection is slow.
Collapse
Affiliation(s)
- Maryam Shariatzadeh
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU UK
| | - Amit Chandra
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU UK
- Present Address: Yposkesi, 26, rue Henri Auguste-Desbruères, 91100 Corbeil-Essonnes, France
| | - Samantha L Wilson
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU UK
| | - Mark J McCall
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU UK
| | - Lise Morizur
- CECS/I-STEM, 28, rue Henri Auguste-Desbruères, 91100 Corbeil-Essonnes, France
| | - Léa Lesueur
- CECS/I-STEM, 28, rue Henri Auguste-Desbruères, 91100 Corbeil-Essonnes, France
| | - Olivier Chose
- CECS/I-STEM, 28, rue Henri Auguste-Desbruères, 91100 Corbeil-Essonnes, France
| | - Michael M. Gepp
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
- Fraunhofer Project Center for Stem Cell Process Engineering, Neunerplatz 2, 97082 Würzburg, Germany
| | - André Schulz
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
- Present Address: Knappschaft Eye Clinic Sulzbach, An der Klinik 10, 66280 Sulzbach, Germany
| | - Julia C. Neubauer
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
- Fraunhofer Project Center for Stem Cell Process Engineering, Neunerplatz 2, 97082 Würzburg, Germany
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
- Fraunhofer Project Center for Stem Cell Process Engineering, Neunerplatz 2, 97082 Würzburg, Germany
- Saarland University, 66123 Saarbruecken, Germany
- Universidad Católica del Norte, Coquimbo, Chile
| | - Elsa Abranches
- NISBC, Blanche Lane, South Mimms, Potters Bar, EN6 3QG UK
| | - Jennifer Man
- NISBC, Blanche Lane, South Mimms, Potters Bar, EN6 3QG UK
- Present Address: Oxfordshire, UK
| | - Orla O’Shea
- NISBC, Blanche Lane, South Mimms, Potters Bar, EN6 3QG UK
| | - Glyn Stacey
- NISBC, Blanche Lane, South Mimms, Potters Bar, EN6 3QG UK
- Present Address: Adaptimmune, 60 Jubilee Avenue, Milton Park, Abingdon, Oxfordshire OX14 4RX UK
| | - Zoe Hewitt
- Centre for Stem Cell Biology (CSCB), University of Sheffield, Western Bank, Sheffield, S10 2TN UK
| | - David J Williams
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU UK
| |
Collapse
|
15
|
Das R, Roosloot R, van Pel M, Schepers K, Driessen M, Fibbe WE, de Bruijn JD, Roelofs H. Preparing for cell culture scale-out: establishing parity of bioreactor- and flask-expanded mesenchymal stromal cell cultures. J Transl Med 2019; 17:241. [PMID: 31340829 PMCID: PMC6657181 DOI: 10.1186/s12967-019-1989-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Cell-based therapies have the potential to become treatment options for many diseases, but efficient scale-out of these therapies has proven to be a major hurdle. Bioreactors can be used to overcome this hurdle, but changing the culture method can introduce unwanted changes to the cell product. Therefore, it is important to establish parity between products generated using traditional methods versus those generated using a bioreactor. Methods Mesenchymal stromal cells (MSCs) are cultured in parallel using either traditional culture flasks, spinner vessels or a new bioreactor system. To investigate parity between the cells obtained from different methods, harvested cells are compared in terms of yield, phenotype and functionality. Results Bioreactor-based expansion yielded high cell numbers (222–510 million cells). Highest cell expansion was observed upon culture in flasks [average 5.0 population doublings (PDL)], followed by bioreactor (4.0 PDL) and spinner flasks (3.3 PDL). Flow cytometry confirmed MSC identity (CD73+, CD90+ and CD105+) and lack of contaminating hematopoietic cell populations. Cultured MSCs did not display genetic aberrations and no difference in differentiation and immunomodulatory capacity was observed between culture conditions. The response to IFNγ stimulation was similar for cells obtained from all culture conditions, as was the capacity to inhibit T cell proliferation. Conclusions The new bioreactor technology can be used to culture large amounts of cells with characteristics equivalent to those cultured using traditional, flask based, methods. Electronic supplementary material The online version of this article (10.1186/s12967-019-1989-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruud Das
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands.
| | - Rens Roosloot
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands
| | - Melissa van Pel
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Koen Schepers
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Marijn Driessen
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands
| | - Willem E Fibbe
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Joost Dick de Bruijn
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands.,Twente University, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.,Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Helene Roelofs
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
16
|
Lin H, Li Q, Du Q, Wang O, Wang Z, Akert L, Carlson MA, Zhang C, Subramanian A, Zhang C, Lunning M, Li M, Lei Y. Integrated generation of induced pluripotent stem cells in a low-cost device. Biomaterials 2019; 189:23-36. [DOI: 10.1016/j.biomaterials.2018.10.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
|
17
|
Bandeiras C, Cabral JM, Finkelstein SN, Ferreira FC. Modeling biological and economic uncertainty on cell therapy manufacturing: the choice of culture media supplementation. Regen Med 2018; 13:917-933. [PMID: 30488770 DOI: 10.2217/rme-2018-0034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM To evaluate the cost-effectiveness of autologous cell therapy manufacturing in xeno-free conditions. MATERIALS & METHODS Published data on the isolation and expansion of mesenchymal stem/stromal cells introduced donor, multipassage and culture media variability on cell yields and process times on adherent culture flasks to drive cost simulation of a scale-out campaign of 1000 doses of 75 million cells each in a 400 square meter Good Manufacturing Practices facility. RESULTS & CONCLUSION Passage numbers in the expansion step are strongly associated with isolation cell yield and drive cost increases per donor of $1970 and 2802 for fetal bovine serum and human platelet lysate. Human platelet lysate decreases passage numbers and process costs in 94.5 and 97% of donors through lower facility and labor costs. Cost savings are maintained with full equipment depreciation and higher numbers of cells per dose, highlighting the number of cells per passage step as the key cost driver.
Collapse
Affiliation(s)
- Cátia Bandeiras
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal.,Institute for Data, Systems & Society, Massachusetts Institute of Technology, 50 Ames Street, Cambridge MA 02139, USA.,Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston MA 02215, USA
| | - Joaquim Ms Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal
| | - Stan N Finkelstein
- Institute for Data, Systems & Society, Massachusetts Institute of Technology, 50 Ames Street, Cambridge MA 02139, USA.,Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston MA 02215, USA
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal
| |
Collapse
|
18
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med 2018; 23:720-730. [PMID: 30484934 PMCID: PMC6349184 DOI: 10.1111/jcmm.14035] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/20/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is a common, severe emergency case in clinics, with high incidence, significant mortality and increased costs. Despite development in the understanding of its pathophysiology, the therapeutic choices are still confined to dialysis and renal transplantation. Considering their antiapoptotic, immunomodulatory, antioxidative and pro‐angiogenic effects, mesenchymal stem cells (MSCs) may be a promising candidate for AKI management. Based on these findings, some clinical trials have been performed, but the results are contradictory (NCT00733876, NCT01602328). The low engraftment, poor survival rate, impaired paracrine ability and delayed administration of MSCs are the four main reasons for the limited clinical efficacy. Investigators have developed a series of preconditioning strategies to improve MSC survival rates and paracrine ability. In this review, by summarizing these encouraging studies, we intend to provide a comprehensive understanding of various preconditioning strategies on AKI therapy and improve the prognosis of AKI patients by regenerative medicine.
Collapse
Affiliation(s)
- Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ping Zhang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hua Jiang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jianghua Chen
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
19
|
Hunsberger JG, Shupe T, Atala A. An Industry-Driven Roadmap for Manufacturing in Regenerative Medicine. Stem Cells Transl Med 2018; 7:564-568. [PMID: 30009571 PMCID: PMC6090514 DOI: 10.1002/sctm.18-0060] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/19/2018] [Indexed: 01/24/2023] Open
Abstract
Regenerative medicine is poised to become a significant industry within the medical field. As such, the development of strategies and technologies for standardized and automated regenerative medicine clinical manufacturing has become a priority. An industry‐driven roadmap toward industrial scale clinical manufacturing was developed over a 3‐year period by a consortium of companies with significant investment in the field of regenerative medicine. Additionally, this same group identified critical roadblocks that stand in the way of advanced, large‐scale regenerative medicine clinical manufacturing. This perspective article details efforts to reach a consensus among industry stakeholders on the shortest pathway for providing access to regenerative medicine therapies for those in need, both within the United States and around the world. Stem Cells Translational Medicine2018;7:564–568
Collapse
Affiliation(s)
- Joshua G Hunsberger
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Thomas Shupe
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| |
Collapse
|
20
|
Vilsbøll AW, Mouritsen JM, Jensen LP, Bødker N, Holst AW, Pennisi CP, Ehlers L. Cell-based therapy for the treatment of female stress urinary incontinence: an early cost-effectiveness analysis. Regen Med 2018; 13:321-330. [PMID: 29715070 DOI: 10.2217/rme-2017-0124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To perform an early cost-effectiveness analysis of in vitro expanded myoblasts (IVM) and minced myofibers versus midurethral slings (MUS) for surgical treatment of female stress urinary incontinence. METHODS Cost-effectiveness and sensitivity analyses were performed using a decision tree comprising previously published data and expert opinions. RESULTS & CONCLUSION In the base case scenario, MUS was the cost-effective strategy with a negative incremental cost-effectiveness ratio compared with IVM and a positive incremental cost-effectiveness ratio compared with minced myofibers. However, the sensitivity analysis indicates that IVM may become an alternative providing greater effect at a higher cost. With the possibility of becoming more effective, IVM treatment would be advantageous over MUS given its reduced invasiveness and lower risks of complications.
Collapse
Affiliation(s)
- Andreas West Vilsbøll
- Department of Health Science & Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Jakob Munk Mouritsen
- Department of Health Science & Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Line Park Jensen
- Department of Health Science & Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Nikolaj Bødker
- Department of Health Science & Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | | | - Cristian P Pennisi
- Department of Health Science & Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Lars Ehlers
- Danish Center for Healthcare Improvements, Aalborg University, Aalborg, Denmark
| |
Collapse
|
21
|
Hourd P, Williams DJ. Scanning the horizon for high value-add manufacturing science: Accelerating manufacturing readiness for the next generation of disruptive, high-value curative cell therapeutics. Cytotherapy 2018; 20:759-767. [DOI: 10.1016/j.jcyt.2018.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/11/2022]
|
22
|
Calmels B, Mfarrej B, Chabannon C. From clinical proof-of-concept to commercialization of CAR T cells. Drug Discov Today 2018; 23:758-762. [PMID: 29317339 DOI: 10.1016/j.drudis.2018.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/09/2017] [Accepted: 01/04/2018] [Indexed: 01/01/2023]
Abstract
The development of CAR T cells currently represents an exciting opportunity to convert the already published clinical successes observed in clinical trials into commercially available efficient therapies. However, the path toward successful commercialization is still hindered by many hurdles. Here, we review such issues as: the need for structured collaborations between hospital collection and clinical facilities and industry manufacturing facilities to streamline the supply chain; necessity for uniform and efficient medical procedures to cope with severe toxicities associated with CAR T cells; and absolute need to define an economical and sustainable model for manufacturers and payers. The fast pace at which the field is evolving requires careful assessments for the benefit of patients.
Collapse
Affiliation(s)
- Boris Calmels
- CBT-1409: INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, AP-HM, Marseille, France; Cell Processing and Cell Collection Facility/Tissue Establishment, Institut Paoli-Calmettes, Marseille, France
| | - Bechara Mfarrej
- CBT-1409: INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, AP-HM, Marseille, France; Cell Processing and Cell Collection Facility/Tissue Establishment, Institut Paoli-Calmettes, Marseille, France
| | - Christian Chabannon
- CBT-1409: INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, AP-HM, Marseille, France; Cell Processing and Cell Collection Facility/Tissue Establishment, Institut Paoli-Calmettes, Marseille, France; EBMT Cell Therapy & Immunobiology Working Party.
| |
Collapse
|
23
|
Harrison RP, Ruck S, Rafiq QA, Medcalf N. Decentralised manufacturing of cell and gene therapy products: Learning from other healthcare sectors. Biotechnol Adv 2017; 36:345-357. [PMID: 29278756 DOI: 10.1016/j.biotechadv.2017.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/13/2022]
Abstract
Decentralised or 'redistributed' manufacturing represents an attractive choice for production of some cell and gene therapies (CGTs), in particular personalised therapies. Decentralised manufacturing splits production into various locations or regions and in doing so, imposes organisational changes on the structure of a company. This confers a significant advantage by democratising supply, creating jobs without geographical restriction to the central hub and allowing a more flexible response to external pressures and demands. This comes with challenges that need to be addressed including, a reduction in oversight, decision making and control by central management which can be critical in maintaining quality in healthcare product manufacturing. The unwitting adoption of poor business strategies at an early stage in development has the potential to undermine the market success of otherwise promising products. To maximise the probability of realising the benefits that decentralised manufacturing of CGTs has to offer, it is important to examine alternative operational paradigms to learn from their successes and to avoid their failures. Whilst no other situation is quite the same as CGTs, some illustrative examples of established manufacturing paradigms are described. Each of these shares a unique attribute with CGTs which aids understanding of how decentralised manufacturing might be implemented for CGTs in a similar manner. In this paper we present a collection of paradigms that can be drawn on in formulating a roadmap to success for decentralised production of CGTs.
Collapse
Affiliation(s)
- Richard P Harrison
- Centre for Biological Engineering, Holywell Park, Loughborough University, Loughborough LE11 3TU, UK.
| | - Steven Ruck
- Centre for Biological Engineering, Holywell Park, Loughborough University, Loughborough LE11 3TU, UK
| | - Qasim A Rafiq
- Department of Biochemical Engineering, Faculty of Engineering Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Nicholas Medcalf
- Centre for Biological Engineering, Holywell Park, Loughborough University, Loughborough LE11 3TU, UK
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Organ donation in the United States registered 9079 deceased organ donors in 2015. This high percentage of donations allowed organ transplantation in 29 851 recipients. Despite increasing numbers of transplants performed in comparison with previous years, the numbers of patients that are in need for a transplant increase every year at a higher rate. This reveals that the discrepancy between the demand and availability of organs remains fundamental problem in organ transplantation. RECENT FINDINGS Development of bioengineered organs represents a promising approach to increase the pool of organs for transplantation. The technology involves obtaining complex three-dimensional scaffolds that support cellular activity and functional remodeling though tissue recellularization protocols using progenitor cells. This innovative approach integrates cross-thematic approaches from specific areas of transplant immunology, tissue engineering and stem cell biology, to potentially manufacture an unlimited source of donor organs for transplantation. SUMMARY Although bioengineered organs are thought to escape immune recognition, the potential immune reactivity toward each of its components has not been studied in detail. Here, we summarize the host immune response toward different progenitor cells and discuss the potential implications of using nonself biological scaffolds to develop bioengineered organs.
Collapse
|
25
|
Morrow D, Ussi A, Migliaccio G. Addressing Pressing Needs in the Development of Advanced Therapies. Front Bioeng Biotechnol 2017; 5:55. [PMID: 28993805 PMCID: PMC5622199 DOI: 10.3389/fbioe.2017.00055] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/06/2017] [Indexed: 12/16/2022] Open
Abstract
The commercial development of advanced therapy medicinal products (ATMPs) represents great opportunity for therapeutic innovation but is beset by many challenges for its developers. Although the ATMP field continues to progress at a rapid pace, evidenced by the increasing number of clinical trials conducted over the past few years, several factors continue to complicate the introduction of ATMPs as a curative treatment for multiple disease types, by blocking their translational pathway from research to the patient. While several recent publications (Trounson and McDonald, 2015; Abou-El-Enein et al., 2016a,b) as well as an Innovative Medicines Initiative consultation (IMI, 2016) this year have highlighted the major gaps in ATMP development, with manufacturing, regulatory, and reimbursement issues at the forefront, there remains to be formulated a coherent strategy to address these by bringing the relevant stakeholders to a single forum, whose task it would be to design and execute a delta plan to alleviate the most pressing bottlenecks. This article focuses on two of the most urgent areas in need of attention in ATMP development, namely manufacturing and reimbursement, and promotes the concept of innovation-dedicated research infrastructures to support a multi-sector approach for ensuring the successful development, entry, and ensuing survival of ATMPs in the healthcare market.
Collapse
Affiliation(s)
- David Morrow
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, Netherlands
| | - Anton Ussi
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, Netherlands
| | - Giovanni Migliaccio
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, Netherlands
| |
Collapse
|
26
|
A roadmap for cost-of-goods planning to guide economic production of cell therapy products. Cytotherapy 2017; 19:1383-1391. [PMID: 28935190 DOI: 10.1016/j.jcyt.2017.06.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/29/2017] [Indexed: 12/31/2022]
Abstract
Cell therapy products are frequently developed and produced without incorporating cost considerations into process development, contributing to prohibitively costly products. Herein we contextualize individual process development decisions within a broad framework for cost-efficient therapeutic manufacturing. This roadmap guides the analysis of cost of goods (COG) arising from tissue procurement, material acquisition, facility operation, production, and storage. We present the specific COG considerations related to each of these elements as identified through a 2013 International Society for Cellular Therapy COG survey, highlighting the differences between autologous and allogeneic products. Planning and accounting for COG at each step in the production process could reduce costs, allowing for more affordable market pricing to improve the long-term viability of the cell therapy product and facilitate broader patient access to novel and transformative cell therapies.
Collapse
|
27
|
Swioklo S, Ding P, Pacek AW, Connon CJ. Process parameters for the high-scale production of alginate-encapsulated stem cells for storage and distribution throughout the cell therapy supply chain. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
28
|
Abbasalizadeh S, Pakzad M, Cabral JMS, Baharvand H. Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin Biol Ther 2017; 17:1201-1219. [PMID: 28699788 DOI: 10.1080/14712598.2017.1354982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Currently, promising outcomes from clinical trials of allogeneic cells, especially allogeneic mesenchymal stromal cells, fibroblasts, keratinocytes, and human cardiac stem cells, have encouraged research institutions, small and medium enterprises (SMEs), and big pharmaceutical companies to invest and focus on developing allogeneic cell therapy products. Commercial and large-scale production of allogeneic cell therapy products requires unique capabilities to develop technologies that generate safe and effective allogeneic cells/cell lines and their fully characterized master/working banks. In addition, it is necessary to design robust upstream and downstream manufacturing processes, and establish integrated, well-designed manufacturing facilities to produce high quality affordable products in accordance with current GMP regulations for the production of cell therapy products. Areas covered: The authors highlight: the recent advances in the development of allogeneic products, the available options to develop robust manufacturing processes, and facility design considerations. Expert opinion: Currently, there are multiple challenges in development of allogeneic cell therapy products. Indeed, the field is still in its infancy; with technologies and regulations still under development, as is our understanding of the mechanisms of action in the body and their interaction with the host immune system. Their characterization and testing is also an emerging and very complex area.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran.,b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Mohammad Pakzad
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Joaquim M S Cabral
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Hossein Baharvand
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran.,c Department of Developmental Biology , University of Science and Culture , Tehran , Iran
| |
Collapse
|
29
|
Stacey GN, Connon CJ, Coopman K, Dickson AJ, Fuller B, Hunt CJ, Kemp P, Kerby J, Man J, Matejtschuk P, Moore H, Morris J, Oreffo ROC, Slater N, Ward S, Wiggins C, Zimmermann H. Preservation and stability of cell therapy products: recommendations from an expert workshop. Regen Med 2017; 12:553-564. [DOI: 10.2217/rme-2017-0073] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
If the field of regenerative medicine is to deliver therapies, rapid expansion and delivery over considerable distances to large numbers of patients is needed. This will demand efficient stabilization and shipment of cell products. However, cryopreservation science is poorly understood by life-scientists in general and in recent decades only limited progress has been made in the technology of preservation and storage of cells. Rapid translation of new developments to a broader range of cell types will be vital, as will assuring a deeper knowledge of the fundamental cell biology relating to successful preservation and recovery of cell cultures. This report presents expert consensus on these and other issues which need to be addressed for more efficient delivery of cell therapies.
Collapse
Affiliation(s)
- Glyn N Stacey
- UK Stem Cell Bank, Division of Advanced Therapies, NIBSC, South Mimms, Hertfordshire, UK
| | - Che J Connon
- Institute of Genetic Medicine, University of Newcastle, Newcastle upon Tyne, UK
| | - Karen Coopman
- Chemical Engineering, Loughborough University, Loughborough, UK
| | - Alan J Dickson
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Barry Fuller
- Department of Surgery, University College London, London, UK
| | - Charles J Hunt
- UK Stem Cell Bank, Division of Advanced Therapies, NIBSC, South Mimms, Hertfordshire, UK
| | - Paul Kemp
- Intercytex Ltd & HairClone, Manchester, UK
| | - Julie Kerby
- Cell Therapy Manufacturing Development, Pfizer, Cambridge, UK
| | - Jennifer Man
- UK Stem Cell Bank, Division of Advanced Therapies, NIBSC, South Mimms, Hertfordshire, UK
| | - Paul Matejtschuk
- Standardisation Science, National Institute for Biological Standards and Control (NIBSC) a centre of the MHRA, South Mimms, Hertfordshire, UK
| | - Harry Moore
- Department of Biomedical Sciences, University of Sheffield, Sheffield, UK
| | | | - Richard OC Oreffo
- Centre for Human Development, Stem Cells & Regeneration, University of Southampton, Southampton, UK
| | - Nigel Slater
- The Bioscience Engineering Group, University of Cambridge, Cambridge, UK
| | | | - Claire Wiggins
- National Health Service – Blood & Transplant, Watford, UK
| | - Heiko Zimmermann
- Fraunhofer-Institute for Biomedical Engineering, Sulzbach, Germany
- Department of Molecular & Cellular Biotechnology/Nanotechnology, Saarland University, Saarbrücken, Germany
- Department of Marine Sciences, Universidad Católica del Norte, Antafogasta/Coquimbo, Chile
| |
Collapse
|
30
|
Papadaki M. Adaptation through Collaboration: Developing Novel Platforms to Advance the Delivery of Advanced Therapies to Patients. Front Med (Lausanne) 2017; 4:56. [PMID: 28611985 PMCID: PMC5447030 DOI: 10.3389/fmed.2017.00056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 04/27/2017] [Indexed: 12/23/2022] Open
Abstract
For the nascent field of advanced therapies, collaboration will be a game-changer, turning scientific progress that was once unimaginable into transformative medical practice. Despite promise for lifelong management and even cure of disease, skepticism remains about the feasibility of their delivery to patients, fueling investment risks. With the potential for long-term effectiveness in need of frequent reassessment, current approaches to predict real-life drug performance bear little relevance, necessitating novel and iterative schemes to monitoring the benefit–risk profiles throughout the life span of advanced therapies. This work explains that reinventing an adoption route for Advanced Therapy Medicinal Products is as much about the scientific and clinical components, as it is about the organizational structures, requiring an unprecedented level of interactions between stakeholders not traditionally connected; from developers and regulators, to payers, patients, and funders. By reflecting on the successes and lessons learned from the growing space of global precompetitive consortia and public–private partnerships, as well as a number of emerging accelerated development pathways, this work aims to inform the foundations for a future roadmap that can smooth the path to approval, reimbursement, and access, while delivering value to all stakeholders. Echoing the growing demands to bring these transformative products to patients, it provides critical insights to enhance our capacity in three fundamental domains: deploying the operational flexibilities offered by the growing space of collaborations, utilizing emerging flexible and accelerated pathways to tackle challenges in quantifying long-term effectiveness, and building the necessary digital and clinical infrastructure for knowledge development.
Collapse
Affiliation(s)
- Magdalini Papadaki
- Association of the British Pharmaceutical Industry, London, United Kingdom
| |
Collapse
|
31
|
Joswig AJ, Mitchell A, Cummings KJ, Levine GJ, Gregory CA, Smith R, Watts AE. Repeated intra-articular injection of allogeneic mesenchymal stem cells causes an adverse response compared to autologous cells in the equine model. Stem Cell Res Ther 2017; 8:42. [PMID: 28241885 PMCID: PMC5329965 DOI: 10.1186/s13287-017-0503-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/30/2017] [Accepted: 02/09/2017] [Indexed: 12/11/2022] Open
Abstract
Background Intra-articular injection of mesenchymal stem cells (MSCs) is efficacious in osteoarthritis therapy. A direct comparison of the response of the synovial joint to intra-articular injection of autologous versus allogeneic MSCs has not been performed. The objective of this study was to assess the clinical response to repeated intra-articular injection of allogeneic versus autologous MSCs prepared in a way to minimize xeno-contaminants in a large animal model. Methods Intra-articular injections of bone marrow-derived, culture-expanded MSCs to a forelimb metacarpophalangeal joint were performed at week 0 and week 4 (six autologous; six autologous with xeno-contamination; six allogeneic). In the week following each injection, clinical and synovial cytology evaluations were performed. Results Following the first intra-articular injection, there were no differences in clinical parameters over time. Following the second intra-articular injection, there was a significant adverse response of the joint to allogeneic MSCs and autologous MSCs with xeno-contamination with elevated synovial total nucleated cell counts. There was also significantly increased pain from joints injected with autologous MSCs with xeno-contamination. Conclusions Repeated intra-articular injection of allogeneic MSCs results in an adverse clinical response, suggesting there is immune recognition of allogeneic MSCs upon a second exposure. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0503-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amanda-Jo Joswig
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Alexis Mitchell
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Kevin J Cummings
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Gwendolyn J Levine
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843, USA
| | - Carl A Gregory
- Institute for Regenerative Medicine and Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, Temple, TX, 76502, USA
| | - Roger Smith
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843, USA
| | - Ashlee E Watts
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
32
|
Lin H, Li Q, Lei Y. An Integrated Miniature Bioprocessing for Personalized Human Induced Pluripotent Stem Cell Expansion and Differentiation into Neural Stem Cells. Sci Rep 2017; 7:40191. [PMID: 28057917 PMCID: PMC5216399 DOI: 10.1038/srep40191] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/01/2016] [Indexed: 01/01/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) are ideal cell sources for personalized cell therapies since they can be expanded to generate large numbers of cells and differentiated into presumably all the cell types of the human body in vitro. In addition, patient specific iPSC-derived cells induce minimal or no immune response in vivo. However, with current cell culture technologies and bioprocessing, the cost for biomanufacturing clinical-grade patient specific iPSCs and their derivatives are very high and not affordable for majority of patients. In this paper, we explored the use of closed and miniature cell culture device for biomanufacturing patient specific neural stem cells (NSCs) from iPSCs. We demonstrated that, with the assist of a thermoreversible hydrogel scaffold, the bioprocessing including iPSC expansion, iPSC differentiation into NSCs, the subsequent depletion of undifferentiated iPSCs from the NSCs, and concentrating and transporting the purified NSCs to the surgery room, could be integrated and completed within two closed 15 ml conical tubes.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Fred &Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
33
|
Thurman-Newell JA, Petzing JN, Williams DJ. A meta-analysis of biological variation in blood-based therapy as a precursor to bio-manufacturing. Cytotherapy 2016; 18:686-94. [PMID: 27059205 DOI: 10.1016/j.jcyt.2016.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 01/04/2016] [Accepted: 01/23/2016] [Indexed: 11/17/2022]
Abstract
Currently cellular therapies, such as hematopoietic stem cell transplantation (HSCT), are produced at a small scale on a case-by-case basis, usually in a clinical or near-clinical setting. Meeting the demand for future cellular therapies will require a robust and scalable manufacturing process that is either designed around or controls the variation associated with biological starting materials. Understanding variation requires both a measure of the allowable variation (that does not negatively affect patient outcome) and the achievable variation (with current technology). The prevalence of HSCT makes it an ideal case study to prepare for more complex biological manufacturing with more challenging regulatory classifications. A systematic meta-analysis of the medical literature surrounding HSCT has been completed of which the key outcomes are the following: (i) the range of transplanted CD34+ cells/kg can be up to six orders of magnitude around the median for allogeneic procedures and four orders of magnitude for autologous procedures, (ii) there is no improvement in variation encountered over a period of 30 years and (iii) as study size increases, the amount of variation encountered also increases. A more detailed, stratified source from a controlled single-site clinical center is required to further define a control strategy for the manufacture of biologics.
Collapse
Affiliation(s)
- Jamie A Thurman-Newell
- Healthcare Engineering Group, Centre for Biological Engineering, Holywell Park, Loughborough University, Loughborough, UK.
| | - Jon N Petzing
- Healthcare Engineering Group, Centre for Biological Engineering, Holywell Park, Loughborough University, Loughborough, UK
| | - David J Williams
- Healthcare Engineering Group, Centre for Biological Engineering, Holywell Park, Loughborough University, Loughborough, UK
| |
Collapse
|
34
|
A relativity concept in mesenchymal stromal cell manufacturing. Cytotherapy 2016; 18:613-20. [PMID: 27059199 DOI: 10.1016/j.jcyt.2016.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/12/2016] [Accepted: 02/05/2016] [Indexed: 02/07/2023]
Abstract
Mesenchymal stromal cells (MSCs) are being experimentally tested in several biological systems and clinical settings with the aim of verifying possible therapeutic effects for a variety of indications. MSCs are also known to be heterogeneous populations, with phenotypic and functional features that depend heavily on the individual donor, the harvest site, and the culture conditions. In the context of this multidimensional complexity, a recurrent question is whether it is feasible to produce MSC batches as "standard" therapeutics, possibly within scalable manufacturing systems. Here, we provide a short overview of the literature on different culture methods for MSCs, including those employing innovative technologies, and of some typically assessed functional features (e.g., growth, senescence, genomic stability, clonogenicity, etc.). We then offer our perspective of a roadmap on how to identify and refine manufacturing systems for MSCs intended for specific clinical indications. We submit that the vision of producing MSCs according to a unique standard, although commercially attractive, cannot yet be scientifically substantiated. Instead, efforts should be concentrated on standardizing methods for characterization of MSCs generated by different groups, possibly covering a vast gamut of functionalities. Such assessments, combined with hypotheses on the therapeutic mode of action and associated clinical data, should ultimately allow definition of in-process controls and measurable release criteria for MSC manufacturing. These will have to be validated as predictive of potency in suitable pre-clinical models and of therapeutic efficacy in patients.
Collapse
|
35
|
Lambrechts T, Sonnaert M, Schrooten J, Luyten FP, Aerts JM, Papantoniou I. Large-Scale Mesenchymal Stem/Stromal Cell Expansion: A Visualization Tool for Bioprocess Comparison. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:485-498. [DOI: 10.1089/ten.teb.2016.0111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Toon Lambrechts
- M3-BIORES: Measure, Model and Manage Bioresponses, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Sonnaert
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Department of Metallurgy and Materials Engineering, KU Leuven, Leuven, Belgium
| | - Jan Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Antleron, Leuven, Belgium
| | - Frank P. Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Jean-Marie Aerts
- M3-BIORES: Measure, Model and Manage Bioresponses, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
36
|
Large-scale progenitor cell expansion for multiple donors in a monitored hollow fibre bioreactor. Cytotherapy 2016; 18:1219-33. [PMID: 27421744 DOI: 10.1016/j.jcyt.2016.05.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/29/2016] [Accepted: 05/20/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND AIMS With the increasing scale in stem cell production, a robust and controlled cell expansion process becomes essential for the clinical application of cell-based therapies. The objective of this work was the assessment of a hollow fiber bioreactor (Quantum Cell Expansion System from Terumo BCT) as a cell production unit for the clinical-scale production of human periosteum derived stem cells (hPDCs). METHODS We aimed to demonstrate comparability of bioreactor production to standard culture flask production based on a product characterization in line with the International Society of Cell Therapy in vitro benchmarks and supplemented with a compelling quantitative in vivo bone-forming potency assay. Multiple process read-outs were implemented to track process performance and deal with donor-to-donor-related variation in nutrient needs and harvest timing. RESULTS The data show that the hollow fiber bioreactor is capable of robustly expanding autologous hPDCs on a clinical scale (yield between 316 million and 444 million cells starting from 20 million after ± 8 days of culture) while maintaining their in vitro quality attributes compared with the standard flask-based culture. The in vivo bone-forming assay on average resulted in 10.3 ± 3.7% and 11.0 ± 3.8% newly formed bone for the bioreactor and standard culture flask respectively. The analysis showed that the Quantum system provides a reproducible cell expansion process in terms of yields and culture conditions for multiple donors.
Collapse
|
37
|
Abou-El-Enein M, Bauer G, Medcalf N, Volk HD, Reinke P. Putting a price tag on novel autologous cellular therapies. Cytotherapy 2016; 18:1056-1061. [PMID: 27288308 DOI: 10.1016/j.jcyt.2016.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 12/24/2022]
Abstract
Cell therapies, especially autologous therapies, pose significant challenges to researchers who wish to move from small, probably academic, methods of manufacture to full commercial scale. There is a dearth of reliable information about the costs of operation, and this makes it difficult to predict with confidence the investment needed to translate the innovations to the clinic, other than as small-scale, clinician-led prescriptions. Here, we provide an example of the results of a cost model that takes into account the fixed and variable costs of manufacture of one such therapy. We also highlight the different factors that influence the product final pricing strategy. Our findings illustrate the need for cooperative and collective action by the research community in pre-competitive research to generate the operational models that are much needed to increase confidence in process development for these advanced products.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité-University Medicine, Campus Virchow, Berlin, Germany.
| | - Gerhard Bauer
- Institute for Regenerative Cures, University of California Davis, Sacramento, CA, USA
| | - Nicholas Medcalf
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Campus Virchow, Berlin, Germany; Institute of Medical Immunology, Charité University Medicine, Campus Virchow, Berlin, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité-University Medicine, Campus Virchow, Berlin, Germany
| |
Collapse
|
38
|
Heathman TR, Rafiq QA, Chan AK, Coopman K, Nienow AW, Kara B, Hewitt CJ. Characterization of human mesenchymal stem cells from multiple donors and the implications for large scale bioprocess development. Biochem Eng J 2016. [DOI: 10.1016/j.bej.2015.06.018] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
39
|
Hassan S, Huang H, Warren K, Mahdavi B, Smith D, Jong S, Farid SS. Process change evaluation framework for allogeneic cell therapies: impact on drug development and commercialization. Regen Med 2016; 11:287-305. [DOI: 10.2217/rme-2015-0034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Aims: Some allogeneic cell therapies requiring a high dose of cells for large indication groups demand a change in cell expansion technology, from planar units to microcarriers in single-use bioreactors for the market phase. The aim was to model the optimal timing for making this change. Materials & methods: A development lifecycle cash flow framework was created to examine the implications of process changes to microcarrier cultures at different stages of a cell therapy's lifecycle. Results: The analysis performed under assumptions used in the framework predicted that making this switch earlier in development is optimal from a total expected out-of-pocket cost perspective. From a risk-adjusted net present value view, switching at Phase I is economically competitive but a post-approval switch can offer the highest risk-adjusted net present value as the cost of switching is offset by initial market penetration with planar technologies. Conclusion: The framework can facilitate early decision-making during process development.
Collapse
Affiliation(s)
- Sally Hassan
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, Gordon Street, London, WC1H 0AH, UK
| | - Hsini Huang
- Graduate Institute of Public Affairs & Department of Political Science, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei, 10617 Taiwan
| | - Kim Warren
- Cell Processing Technologies, Lonza Walkersville, Inc., 8830 Biggs Ford Road, Walkersville, MD 21793-0127, USA
| | - Behzad Mahdavi
- Cell Processing Technologies, Lonza Walkersville, Inc., 8830 Biggs Ford Road, Walkersville, MD 21793-0127, USA
| | - David Smith
- Cell Processing Technologies, Lonza Walkersville, Inc., 8830 Biggs Ford Road, Walkersville, MD 21793-0127, USA
| | - Simcha Jong
- Department of Management Science & Innovation, University College London, Gower St, London, WC1E 6BT, UK
- Harvard TH Chan School of Public Health, Dept Global Health & Population, Boston, MA 02115, USA
| | - Suzanne S Farid
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, Gordon Street, London, WC1H 0AH, UK
| |
Collapse
|
40
|
Swioklo S, Constantinescu A, Connon CJ. Alginate-Encapsulation for the Improved Hypothermic Preservation of Human Adipose-Derived Stem Cells. Stem Cells Transl Med 2016; 5:339-49. [PMID: 26826163 DOI: 10.5966/sctm.2015-0131] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/16/2015] [Indexed: 12/16/2022] Open
Abstract
Despite considerable progress within the cell therapy industry, unmet bioprocessing and logistical challenges associated with the storage and distribution of cells between sites of manufacture and the clinic exist. We examined whether hypothermic (4°C-23°C) preservation of human adipose-derived stem cells could be improved through their encapsulation in 1.2% calcium alginate. Alginate encapsulation improved the recovery of viable cells after 72 hours of storage. Viable cell recovery was highly temperature-dependent, with an optimum temperature of 15°C. At this temperature, alginate encapsulation preserved the ability for recovered cells to attach to tissue culture plastic on rewarming, further increasing its effect on total cell recovery. On attachment, the cells were phenotypically normal, displayed normal growth kinetics, and maintained their capacity for trilineage differentiation. The number of cells encapsulated (up to 2 × 10(6) cells per milliliter) did not affect viable cell recovery nor did storage of encapsulated cells in a xeno-free, serum-free,current Good Manufacturing Practice-grade medium. We present a simple, low-cost system capable of enhancing the preservation of human adipose-derived stem cells stored at hypothermic temperatures, while maintaining their normal function. The storage of cells in this manner has great potential for extending the time windows for quality assurance and efficacy testing, distribution between the sites of manufacture and the clinic, and reducing the wastage associated with the limited shelf life of cells stored in their liquid state.
Collapse
Affiliation(s)
- Stephen Swioklo
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Andrei Constantinescu
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Che J Connon
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
41
|
Juhl M, Tratwal J, Follin B, Søndergaard RH, Kirchhoff M, Ekblond A, Kastrup J, Haack-Sørensen M. Comparison of clinical grade human platelet lysates for cultivation of mesenchymal stromal cells from bone marrow and adipose tissue. Scandinavian Journal of Clinical and Laboratory Investigation 2016; 76:93-104. [PMID: 26878874 DOI: 10.3109/00365513.2015.1099723] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The utility of mesenchymal stromal cells (MSCs) in therapeutic applications for regenerative medicine has gained much attention. Clinical translation of MSC-based approaches requires in vitro culture-expansion to achieve a sufficient number of cells. The ideal cell culture medium should be devoid of any animal derived components. We have evaluated whether human Platelet Lysate (hPL) could be an attractive alternative to animal supplements. METHODS MSCs from bone marrow (BMSCs) and adipose tissue-derived stromal cells (ASCs) obtained from three donors were culture expanded in three different commercially available hPL fulfilling good manufacturing practice criteria for clinical use. BMSCs and ASCs cultured in Minimum Essential Medium Eagle-alpha supplemented with 5% PLT-Max (Mill Creek), Stemulate™ PL-S and Stemulate™ PL-SP (COOK General Biotechnology) were compared to standard culture conditions with 10% fetal bovine serum (FBS). Cell morphology, proliferation, phenotype, genomic stability, and differentiation potential were analyzed. RESULTS Regardless of manufacturer, BMSCs and ASCs cultured in hPL media showed a significant increase in proliferation capacity compared to FBS medium. In general, the immunophenotype of both BMSCs and ASCs fulfilled International Society for Cellular Therapy (ISCT) criteria after hPL media expansion. Comparative genomic hybridization measurements demonstrated no unbalanced chromosomal rearrangements for BMSCs or ASCs cultured in hPL media or FBS medium. The BMSCs and ASCs could differentiate into osteogenic, adipogenic, or chondrogenic lineages in all four culture conditions. CONCLUSION All three clinically approved commercial human platelet lysates accelerated proliferation of BMSCs and ASCs and the cells meet the ISCT mesenchymal phenotypic requirements without exhibiting chromosomal aberrations.
Collapse
Affiliation(s)
- Morten Juhl
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| | - Josefine Tratwal
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| | - Bjarke Follin
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| | - Rebekka H Søndergaard
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| | - Maria Kirchhoff
- b Department of Clinical Genetics, Rigshospitalet , Copenhagen University Hospital , Copenhagen , Denmark
| | - Annette Ekblond
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| | - Jens Kastrup
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| | - Mandana Haack-Sørensen
- a Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet , Copenhagen University Hospital
| |
Collapse
|
42
|
Schalkwyk MCIV, Maher J. Chimeric antigen receptors: On the road to realising their full potential. World J Immunol 2015; 5:86-94. [DOI: 10.5411/wji.v5.i3.86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/01/2015] [Accepted: 10/13/2015] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptors (CARs) are fusion molecules that may be genetically delivered ex-vivo to T-cells and other immune cell populations, thereby conferring specificity for native target antigens found on the surface of tumour and other target cell types. Antigen recognition by CARs is neither restricted by nor dependent upon human leukocyte antigen antigen expression, favouring widespread use of this technology across transplantation barriers. Signalling is delivered by a designer endodomain that provides a tailored and target-dependent activation signal to polyclonal circulating T-cells. Recent clinical data emphasise the enormous promise of this emerging immunotherapeutic strategy for B-cell malignancy, notably acute lymphoblastic leukaemia. In that context, CARs are generally targeted against the ubiquitous B-cell antigen, CD19. However, CAR T-cell immunotherapy is limited by potential for severe on-target toxicity, notably due to cytokine release syndrome. Furthermore, efficacy in the context of solid tumours remains unproven, owing in part to lack of availability of safe tumour-specific targets, inadequate CAR T-cell homing and hostility of the tumour microenvironment to immune effector deployment. Manufacture and commercial development of this strategy also impose new challenges not encountered with more traditional drug products. Finally, there is increasing interest in the application of this technology to the treatment of non-malignant disease states, such as autoimmunity, chronic infection and in the suppression of allograft rejection. Here, we consider the background and direction of travel of this emerging and highly promising treatment for malignant and other disease types.
Collapse
|
43
|
Gardner J, Faulkner A, Mahalatchimy A, Webster A. Are there specific translational challenges in regenerative medicine? Lessons from other fields. Regen Med 2015; 10:885-95. [PMID: 26541074 DOI: 10.2217/rme.15.50] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
There is concern that translation 'from bench to bedside' within regenerative medicine (RM) will fail to materialize, or will be dismally slow, due to various challenges arising from the highly novel and disruptive nature of RM. In this article, we provide a summary of these challenges, and we critically engage with the notion that such challenges are specific to RM. It is important, we argue, not to overstate the exceptional nature of RM, as valuable lessons can be learned from elsewhere in medicine. Using several examples of technology adoption, we suggest that emerging RM products and procedures will have to work hard to find or create an adoption space if translation into the clinic is to be successful.
Collapse
Affiliation(s)
- John Gardner
- Science & Technology Studies Unit, Department of Sociology, University of York, Wentworth College, York, Y010 5DD, UK
| | - Alex Faulkner
- Sociology of Biomedicine & Healthcare Policy, Centre for Global Health Policy, University of Sussex, Brighton, BN1 9RH, UK
| | | | - Andrew Webster
- Science & Technology Studies Unit, Department of Sociology, University of York, Wentworth College, York, Y010 5DD, UK
| |
Collapse
|
44
|
Alrifai D, Sarker D, Maher J. Prospects for adoptive immunotherapy of pancreatic cancer using chimeric antigen receptor-engineered T-cells. Immunopharmacol Immunotoxicol 2015; 38:50-60. [DOI: 10.3109/08923973.2015.1100204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
45
|
Hourd P, Chandra A, Alvey D, Ginty P, McCall M, Ratcliffe E, Rayment E, Williams DJ. Qualification of academic facilities for small-scale automated manufacture of autologous cell-based products. Regen Med 2015; 9:799-815. [PMID: 25431916 DOI: 10.2217/rme.14.47] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Academic centers, hospitals and small companies, as typical development settings for UK regenerative medicine assets, are significant contributors to the development of autologous cell-based therapies. Often lacking the appropriate funding, quality assurance heritage or specialist regulatory expertise, qualifying aseptic cell processing facilities for GMP compliance is a significant challenge. The qualification of a new Cell Therapy Manufacturing Facility with automated processing capability, the first of its kind in a UK academic setting, provides a unique demonstrator for the qualification of small-scale, automated facilities for GMP-compliant manufacture of autologous cell-based products in these settings. This paper shares our experiences in qualifying the Cell Therapy Manufacturing Facility, focusing on our approach to streamlining the qualification effort, the challenges, project delays and inefficiencies we encountered, and the subsequent lessons learned.
Collapse
Affiliation(s)
- Paul Hourd
- EPSRC Center for Innovative Manufacturing in Regenerative Medicine, Center for Biological Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kellathur SN. Regulation of Cell- and Tissue-Based Therapeutic Products in Singapore. Tissue Eng Part A 2015; 21:2802-5. [PMID: 26096855 DOI: 10.1089/ten.tea.2014.0712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The regulatory environment for cell- and tissue-based therapeutic (CTT) products is rapidly evolving and drug regulatory agencies are working toward establishing a risk-based system in their regulatory approach. In Singapore, CTT products such as cell therapy products, stem cell products, and tissue-engineered products in regenerative medicine are regulated as medicinal products. CTT products are defined as articles containing or consisting of autologous or allogeneic human or xenogeneic cells or tissues that are used for or administered to, or intended to be used for or administered to human beings for the diagnosis, treatment, or prevention of human diseases or conditions. Currently, we have applied a risk-based tiered approach whereby high-risk CTT products (substantially manipulated products, products intended for nonhomologous use or combined/used in conjunction with a drug, biologic, or device) are regulated under the Medicines Act. A new standalone regulation for CTT products is being proposed under the Health Products Act where we propose to regulate the entire spectrum (high and low risk) of CTT products.
Collapse
Affiliation(s)
- Srinivasan Nadathur Kellathur
- Advanced Therapy Products Unit, Premarketing, Health Products Regulation Group, Health Sciences Authority , Singapore, Singapore
| |
Collapse
|