1
|
Farrugia A. A Sociology of Plasma Proteins - A Technocrat's Perspective. INTERNATIONAL JOURNAL OF SOCIAL DETERMINANTS OF HEALTH AND HEALTH SERVICES 2025; 55:88-91. [PMID: 39140301 PMCID: PMC11583512 DOI: 10.1177/27551938241264764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 05/13/2024] [Indexed: 08/15/2024]
Abstract
This commentary addresses the article "Toward a Sociology of Plasma Products" by Holloway and Grundy in this issue of the International Journal of Social Determinants of Health and Health Services. The program of research proposed by the authors positioning the medico-industrial field of plasma products within a sociological context is supported, this being an endeavor which has not been attempted previously. I seek to augment Holloway and Grundy's proposed approach through some additional insights which are the result of over forty years of personal commitment in the field. Holloway and Grundy's proposed areas of engagement involving the products, the recipients, the donors and the governance of the systems binding these together is widened through an examination of additional technological factors that have shaped the field. These factors include the influence of the medical industry, the role of patient groups, the continuing controversy on the sourcing of plasma raw material, and the roles of different governance models. Converging these factors with Holloway and Grundy's proposed program should enhance its capacity to develop a framework for understanding the dynamics within this complex and unique sector. The concepts developed in both articles will assist stakeholders to develop a societal framework for the provision of these essential medicines.
Collapse
Affiliation(s)
- Albert Farrugia
- School of Surgery, University of Western Australia Medical School, The University of Western Australia (M509), Perth, Australia
| |
Collapse
|
2
|
Lundh S, Jung IY, Dimitri A, Vora A, Melenhorst JJ, Jadlowsky JK, Fraietta JA. Clinical practice: chimeric antigen receptor (CAR) T cells: a major breakthrough in the battle against cancer. Clin Exp Med 2020; 20:469-480. [PMID: 32333215 PMCID: PMC11413805 DOI: 10.1007/s10238-020-00628-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has come of age, offering a potentially curative option for patients who are refractory to standard anti-cancer treatments. The success of CAR T cell therapy in the setting of acute lymphoblastic leukemia and specific types of B cell lymphoma led to rapid regulatory approvals of CD19-directed CAR T cells, first in the United States and subsequently across the globe. Despite these major milestones in the field of immuno-oncology, growing experience with CAR T cells has also highlighted the major limitations of this strategy, namely challenges associated with manufacturing a bespoke patient-specific product, intrinsic immune cell defects leading to poor CAR T cell function as well as persistence, and/or tumor cell resistance resulting from loss or modulation of the targeted antigen. In addition, both on- and off-tumor immunotoxicities and the financial burden inherent in conventional cellular biomanufacturing often hamper the success of CAR T cell-based treatment approaches. Herein, we provide an overview of the opportunities and challenges related to the first form of gene transfer therapy to gain commercial approval in the United States. Ongoing advances in the areas of genetic engineering, precision genome editing, toxicity mitigation methods and cell manufacturing will improve the efficacy and safety of CAR T cells for hematologic malignancies and expand the use of this novel class of therapeutics to reach solid tumors.
Collapse
Affiliation(s)
- Stefan Lundh
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA
| | - In-Young Jung
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander Dimitri
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anish Vora
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA
| | - J Joseph Melenhorst
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie K Jadlowsky
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, University of Pennsylvania, South Pavilion Expansion, Room 9-104, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA.
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Khalil AS, Yu X, Dang PN, Alsberg E, Murphy WL. A microparticle approach for non-viral gene delivery within 3D human mesenchymal stromal cell aggregates. Acta Biomater 2019; 95:408-417. [PMID: 31004846 PMCID: PMC6888862 DOI: 10.1016/j.actbio.2019.04.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/03/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Abstract
Three-dimensional (3D) multicellular aggregates, in comparison to two-dimensional monolayer culture, can provide tissue culture models that better recapitulate the abundant cell-cell and cell-matrix interactions found in vivo. In addition, aggregates are potentially useful building blocks for tissue engineering. However, control over the interior aggregate microenvironment is challenging due to inherent barriers for diffusion of biological mediators (e.g. growth factors) throughout the multicellular aggregates. Previous studies have shown that incorporation of biomaterials into multicellular aggregates can support cell survival and control differentiation of stem cell aggregates by delivering morphogens from within the 3D construct. In this study, we developed a highly efficient microparticle-based gene delivery approach to uniformly transfect human mesenchymal stromal cells (hMSC) within multicellular aggregates and cell sheets. We hypothesized that release of plasmid DNA (pDNA) lipoplexes from mineral-coated microparticles (MCMs) within 3D hMSC constructs would improve transfection in comparison to standard free pDNA lipoplex delivery in the media. Our approach increased transfection efficiency 5-fold over delivery of free pDNA lipoplexes in the media and resulted in homogenous distribution of transfected cells throughout the 3D constructs. Additionally, we found that MCMs improved hMSC transfection by specifically increasing macropinocytosis-mediated uptake of pDNA. Finally, we showed up to a three-fold increase of bone morphogenetic protein-2 (BMP-2) expression and enhanced calcium deposition within 3D hMSC constructs following MCM-mediated delivery of a BMP-2 encoding plasmid and culture in osteogenic medium. The technology described here provides a valuable tool for achieving efficient and homogenous transfection of 3D cell constructs and is therefore of particular value in tissue engineering and regenerative medicine applications. STATEMENT OF SIGNIFICANCE: This original research describes a materials-based approach, whereby use of mineral-coated microparticles improves the efficiency of non-viral gene delivery in three-dimensional human mesenchymal stromal cell constructs. Specifically, it demonstrates the use of mineral-coated microparticles to enable highly efficient transfection of human mesenchymal stromal cells in large, 3D culture formats. The manuscript also identifies specific endocytosis pathways that interact with the mineral coating to afford the improved transfection efficiency, as well as demonstrates the utility of this approach toward improving differentiation of large cell constructs. We feel that this manuscript will impact the current understanding and near-term development of materials for non-viral gene delivery in broad tissue engineering and biofabrication applications, and therefore be of interest to a diverse biomaterials audience.
Collapse
Affiliation(s)
- Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Phuong N Dang
- Departments of Biomedical Engineering and Orthopedic Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eben Alsberg
- AO Foundation Collaborative Research Center, Davos, Switzerland; Departments of Biomedical Engineering and Orthopedic Surgery, Case Western Reserve University, Cleveland, OH 44106, USA; The National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; School of Dentistry, Kyung Hee University, Seoul, South Korea
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Materials Science Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA; AO Foundation Collaborative Research Center, Davos, Switzerland.
| |
Collapse
|
4
|
Pettitt D, Arshad Z, Davies B, Smith J, French A, Cole D, Bure K, Dopson S, DiGiusto D, Karp J, Reeve B, Barker R, Holländer G, Brindley D. An assessment of the factors affecting the commercialization of cell-based therapeutics: a systematic review protocol. Syst Rev 2017; 6:120. [PMID: 28651620 PMCID: PMC5485574 DOI: 10.1186/s13643-017-0517-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 06/09/2017] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Cellular-based therapies represent a platform technology within the rapidly expanding field of regenerative medicine and are distinct from conventional therapeutics-offering a unique approach to managing what were once considered untreatable diseases. Despite a significant increase in basic science activity within the cell therapy arena, alongside a growing portfolio of cell therapy trials and promising investment, the translation of cellular-based therapeutics from "bench to bedside" remains challenging, and the number of industry products available for widespread clinical use remains comparatively low. This systematic review identifies unique intrinsic and extrinsic barriers in the cell-based therapy domain. METHODS/DESIGN Eight electronic databases will be searched, specifically Medline, EMBASE (OvidSP), BIOSIS & Web of Science, Cochrane Library & HEED, EconLit (ProQuest), WHOLIS WHO Library Database, PAIS International (ProQuest), and Scopus. Addition to this gray literature was searched by manually reviewing relevant work. All identified articles will be subjected for review by two authors who will decide whether or not each article passes our inclusion/exclusion criteria. Eligible papers will subsequently be reviewed, and key data extracted into a pre-designed data extraction scorecard. An assessment of the perceived impact of broad commercial barriers to the adoption of cell-based therapies will be conducted. These broad categories will include manufacturing, regulation and intellectual property, reimbursement, clinical trials, clinical adoption, ethics, and business models. This will inform further discussion in the review. There is no PROSPERO registration number. DISCUSSION Through a systematic search and appraisal of available literature, this review will identify key challenges in the commercialization pathway of cellular-based therapeutics and highlights significant barriers impeding successful clinical adoption. This will aid in creating an adaptable, acceptable, and harmonized approach supported by apposite regulatory frameworks and pertinent expertise throughout the respective stages of the adoption cycle to facilitate the adoption of new products and technologies in the industry.
Collapse
Affiliation(s)
- David Pettitt
- The Oxford - UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK.,Department of Paediatrics, University of Oxford, Oxford, UK
| | - Zeeshaan Arshad
- University of St. Andrews School of Medicine, University of St. Andrews, St. Andrews, UK. .,, Docherty Gardens, Glenrothes, KY7 5GA, UK.
| | - Benjamin Davies
- Orthopedic Surgery Departement, University of Cambridge, Cambridge, UK
| | - James Smith
- The Oxford - UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK.,Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anna French
- The Oxford - UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK
| | | | - Kim Bure
- Sartorius Stedim, Göttingen, Germany
| | - Sue Dopson
- Said Business School, University of Oxford, Oxford, UK
| | - David DiGiusto
- Division of Cell Transplantation and Regenerative Medicine, University of Stanford, Stanford, USA
| | - Jeff Karp
- Harvard Medical School, Harvard University, Boston, USA.,Brigham and Women's Hospital, Boston, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, USA
| | | | | | - Georg Holländer
- Department of Paediatrics, University of Oxford, Oxford, UK.,Department of Biomedicine, University of Basel and Basel University Children's Hospital, Basel, Switzerland
| | - David Brindley
- The Oxford - UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK.,Department of Paediatrics, University of Oxford, Oxford, UK.,Said Business School, University of Oxford, Oxford, UK.,Harvard Stem Cell Institute, Cambridge, USA.,Centre for Behavioral Medicine, UCL School of Pharmacy, University College London, London, UK.,USCF-Stanford Center of Excellence in Regulatory Science and Innovation (CERSI), Stanford, USA
| |
Collapse
|
5
|
Leijten J, Chai Y, Papantoniou I, Geris L, Schrooten J, Luyten F. Cell based advanced therapeutic medicinal products for bone repair: Keep it simple? Adv Drug Deliv Rev 2015; 84:30-44. [PMID: 25451134 DOI: 10.1016/j.addr.2014.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/18/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
Abstract
The development of cell based advanced therapeutic medicinal products (ATMPs) for bone repair has been expected to revolutionize the health care system for the clinical treatment of bone defects. Despite this great promise, the clinical outcomes of the few cell based ATMPs that have been translated into clinical treatments have been far from impressive. In part, the clinical outcomes have been hampered because of the simplicity of the first wave of products. In response the field has set-out and amassed a plethora of complexities to alleviate the simplicity induced limitations. Many of these potential second wave products have remained "stuck" in the development pipeline. This is due to a number of reasons including the lack of a regulatory framework that has been evolving in the last years and the shortage of enabling technologies for industrial manufacturing to deal with these novel complexities. In this review, we reflect on the current ATMPs and give special attention to novel approaches that are able to provide complexity to ATMPs in a straightforward manner. Moreover, we discuss the potential tools able to produce or predict 'goldilocks' ATMPs, which are neither too simple nor too complex.
Collapse
|
6
|
Roy DC, Alarco AM, Isasi R. CellCAN: A Unique Enabler of Regenerative Medicine and Cell Therapy in Canada. Stem Cells Dev 2014; 23 Suppl 1:24-8. [DOI: 10.1089/scd.2014.0354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Denis-Claude Roy
- Division of Hematology–Oncology/Stem Cell Transplantation, Hôpital Maisonneuve-Rosemont Research Center, and Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Anne-Marie Alarco
- CellCAN Regenerative Medicine and Cell Therapy Network, Montreal, Quebec, Canada
| | - Rosario Isasi
- Department of Human Genetics, Faculty of Medicine, Centre of Genomics and Policy, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Lambrechts T, Papantoniou I, Sonnaert M, Schrooten J, Aerts JM. Model-based cell number quantification using online single-oxygen sensor data for tissue engineering perfusion bioreactors. Biotechnol Bioeng 2014; 111:1982-92. [PMID: 24771348 DOI: 10.1002/bit.25274] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/09/2014] [Accepted: 04/15/2014] [Indexed: 01/31/2023]
Abstract
Online and non-invasive quantification of critical tissue engineering (TE) construct quality attributes in TE bioreactors is indispensable for the cost-effective up-scaling and automation of cellular construct manufacturing. However, appropriate monitoring techniques for cellular constructs in bioreactors are still lacking. This study presents a generic and robust approach to determine cell number and metabolic activity of cell-based TE constructs in perfusion bioreactors based on single oxygen sensor data in dynamic perfusion conditions. A data-based mechanistic modeling technique was used that is able to correlate the number of cells within the scaffold (R(2) = 0.80) and the metabolic activity of the cells (R(2) = 0.82) to the dynamics of the oxygen response to step changes in the perfusion rate. This generic non-destructive measurement technique is effective for a large range of cells, from as low as 1.0 × 10(5) cells to potentially multiple millions of cells, and can open-up new possibilities for effective bioprocess monitoring.
Collapse
Affiliation(s)
- T Lambrechts
- Division M3-BIORES: Measure, Model & Manage Bioresponses, KU Leuven, Heverlee, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
8
|
Elman JS, Murray RM, Wang F, Shen K, Gao S, Conway KE, Yarmush ML, Tannous BA, Weissleder R, Parekkadan B. Pharmacokinetics of natural and engineered secreted factors delivered by mesenchymal stromal cells. PLoS One 2014; 9:e89882. [PMID: 24587097 PMCID: PMC3931832 DOI: 10.1371/journal.pone.0089882] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/28/2014] [Indexed: 01/14/2023] Open
Abstract
Transient cell therapy is an emerging drug class that requires new approaches for pharmacological monitoring during use. Human mesenchymal stem cells (MSCs) are a clinically-tested transient cell therapeutic that naturally secrete anti-inflammatory factors to attenuate immune-mediated diseases. MSCs were used as a proof-of-concept with the hypothesis that measuring the release of secreted factors after cell transplantation, rather than the biodistribution of the cells alone, would be an alternative monitoring tool to understand the exposure of a subject to MSCs. By comparing cellular engraftment and the associated serum concentration of secreted factors released from the graft, we observed clear differences between the pharmacokinetics of MSCs and their secreted factors. Exploration of the effects of natural or engineered secreted proteins, active cellular secretion pathways, and clearance mechanisms revealed novel aspects that affect the systemic exposure of the host to secreted factors from a cellular therapeutic. We assert that a combined consideration of cell delivery strategies and molecular pharmacokinetics can provide a more predictive model for outcomes of MSC transplantation and potentially other transient cell therapeutics.
Collapse
Affiliation(s)
- Jessica S. Elman
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Ryan M. Murray
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Fangjing Wang
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Keyue Shen
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Shan Gao
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Kevin E. Conway
- Department of Neurology, Experimental Therapeutics and Molecular Imaging Laboratory, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Martin L. Yarmush
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, United States of America
| | - Bakhos A. Tannous
- Department of Neurology, Experimental Therapeutics and Molecular Imaging Laboratory, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Biju Parekkadan
- Department of Surgery, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospital for Children, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Boston, Massachusetts, United States of America
| |
Collapse
|
9
|
The global cell therapy industry continues to rise during the second and third quarters of 2012. Cell Stem Cell 2013; 11:735-9. [PMID: 23217418 DOI: 10.1016/j.stem.2012.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During Q2-Q3 2012, the cell therapy industry benefited from a number of positive external influences including advantageous changes to future FDA regulation, but stock market activity was highly mixed. The FDA approved two more products and an appreciable number of public-company-sponsored clinical trials are progressing through phases 1-3.
Collapse
|
10
|
Abstract
INTRODUCTION Mesenchymal stem cells (MSC) and MSC-like cells hold great promise and offer many advantages for developing effective cellular therapeutics. Current trends indicate that the clinical application of MSC will continue to increase markedly. For clinical applications, large numbers of MSC are usually required, ideally in an off-the-shelf format, thus requiring extensive MSC expansion ex vivo and subsequent cryopreservation and banking. AREAS COVERED To exploit the full potential of MSC for cell-based therapies requires overcoming significant cell-manufacturing, banking and regulatory challenges. The current review will focus on the identification of optimal cell source for MSC, the techniques for production scale-up, cryopreservation and banking and the regulatory challenges involved. EXPERT OPINION There has been considerable success manufacturing and cryopreserving MSC at laboratory scale. Surprisingly little attention, however, has been given to translate these technologies to an industrial scale. The development of cost-effective advanced technologies for producing and cryopreserving commercial-scale MSC is important for successful clinical cell therapy.
Collapse
|
11
|
dos Santos FF, Andrade PZ, da Silva CL, Cabral JMS. Bioreactor design for clinical-grade expansion of stem cells. Biotechnol J 2013; 8:644-54. [PMID: 23625834 DOI: 10.1002/biot.201200373] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 03/25/2013] [Accepted: 04/02/2013] [Indexed: 01/24/2023]
Abstract
The many clinical trials currently in progress will likely lead to the widespread use of stem cell-based therapies for an extensive variety of diseases, either in autologous or allogeneic settings. With the current pace of progress, in a few years' time, the field of stem cell-based therapy should be able to respond to the market demand for safe, robust and clinically efficient stem cell-based therapeutics. Due to the limited number of stem cells that can be obtained from a single donor, one of the major challenges on the roadmap for regulatory approval of such medicinal products is the expansion of stem cells using Good Manufacturing Practices (GMP)-compliant culture systems. In fact, manufacturing costs, which include production and quality control procedures, may be the main hurdle for developing cost-effective stem cell therapies. Bioreactors provide a viable alternative to the traditional static culture systems in that bioreactors provide the required scalability, incorporate monitoring and control tools, and possess the operational flexibility to be adapted to the differing requirements imposed by various clinical applications. Bioreactor systems face a number of issues when incorporated into stem cell expansion protocols, both during development at the research level and when bioreactors are used in on-going clinical trials. This review provides an overview of the issues that must be confronted during the development of GMP-compliant bioreactors systems used to support the various clinical applications employing stem cells.
Collapse
Affiliation(s)
- Francisco F dos Santos
- Department of Bioengineering and IBB - Institute for Biotechnology and Bioengineering - Instituto Superior Técnico IST, Technical University of Lisbon, Lisboa, Portugal
| | | | | | | |
Collapse
|
12
|
Bertram TA, Tentoff E, Johnson PC, Tawil B, Van Dyke M, Hellman KB. Hurdles in tissue engineering/regenerative medicine product commercialization: a pilot survey of governmental funding agencies and the financial industry. Tissue Eng Part A 2012; 18:2187-94. [PMID: 22838399 DOI: 10.1089/ten.tea.2012.0186] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The Tissue Engineering and Regenerative Medicine International Society of the Americas (TERMIS-AM) Industry Committee conducted a semiquantitative opinion survey in 2010 to delineate potential hurdles to commercialization perceived by the TERMIS constituency groups that participate in the stream of technology commercialization (academia, start-up companies, development-stage companies, and established companies). A significant hurdle identified consistently by each group was access to capital for advancing potential technologies into development pathways leading to commercialization. A follow-on survey was developed by the TERMIS-AM Industry Committee to evaluate the financial industry's perspectives on investing in regenerative medical technologies. The survey, composed of 15 questions, was developed and provided to 37 investment organizations in one of three sectors (governmental, private, and public investors). The survey was anonymous and confidential with sector designation the only identifying feature of each respondent's organization. Approximately 80% of the survey was composed of respondents from the public (n=14) and private (n=15) sectors. Each respondent represents one investment organization with the potential of multiple participants participating to form the organization's response. The remaining organizations represented governmental agencies (n=8). Results from this survey indicate that a high percentage (<60%) of respondents (governmental, private, and public) were willing to invest >$2MM into regenerative medical companies at the different stages of a company's life cycle. Investors recognized major hurdles to this emerging industry, including regulatory pathway, clinical translation, and reimbursement of these new products. Investments in regenerative technologies have been cyclical over the past 10-15 years, but investors recognized a 1-5-year investment period before the exit via Merger and Acquisition (M&A). Investors considered musculoskeletal products and their top technology choice with companies in the clinical stage of development being the most preferred investment targets. All sectors indicated a limited interest in early-stage start-up companies potentially explaining why start-up companies have struggled to access to capital and investors based their investment on the stage of a company's life cycle, reflecting each sector's risk tolerance, exit strategy, time of holding an investment, and investment strategy priorities. Investors highlighted the limited number of regenerative medical companies that have achieved commercial status as a basis for why public investors have been approached by so few companies. Based on respondents to this survey, regenerative medical sponsors seeking capital from the financial industry must keep the explanation of their technology simple, since all sectors considered regenerative medical technology as difficult to evaluate. This survey's results indicate that under the current financial environment, many regenerative medical companies must consider codevelopment or even M&A as nondilutive means of raising capital. The overall summary for this survey highlights the highly varied goals and motivations for the various sectors of the government and financial industries.
Collapse
Affiliation(s)
- Timothy A Bertram
- Research and Development, Tengion, Inc., Winston-Salem, North Carolina 27104, USA.
| | | | | | | | | | | |
Collapse
|