1
|
Dhoundiyal S, Alam MA. Advancements in Biotechnology and Stem Cell Therapies for Breast Cancer Patients. Curr Stem Cell Res Ther 2024; 19:1072-1083. [PMID: 37815191 DOI: 10.2174/011574888x268109230924233850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
This comprehensive review article examines the integration of biotechnology and stem cell therapy in breast cancer diagnosis and treatment. It discusses the use of biotechnological tools such as liquid biopsies, genomic profiling, and imaging technologies for accurate diagnosis and monitoring of treatment response. Stem cell-based approaches, their role in modeling breast cancer progression, and their potential for breast reconstruction post-mastectomy are explored. The review highlights the importance of personalized treatment strategies that combine biotechnological tools and stem cell therapies. Ethical considerations, challenges in clinical translation, and regulatory frameworks are also addressed. The article concludes by emphasizing the potential of integrating biotechnology and stem cell therapy to improve breast cancer outcomes, highlighting the need for continued research and collaboration in this field.
Collapse
Affiliation(s)
- Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar
Pradesh, India
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar
Pradesh, India
| |
Collapse
|
2
|
Meiser I, Alstrup M, Khalesi E, Stephan B, Speicher AM, Majer J, Kwok CK, Neubauer JC, Hansson M, Zimmermann H. Application-Oriented Bulk Cryopreservation of Human iPSCs in Cryo Bags Followed by Direct Inoculation in Scalable Suspension Bioreactors for Expansion and Neural Differentiation. Cells 2023; 12:1914. [PMID: 37508576 PMCID: PMC10378238 DOI: 10.3390/cells12141914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cell-based therapies are promising tools for regenerative medicine and require bulk numbers of high-quality cells. Currently, cells are produced on demand and have a limited shelf-life as conventional cryopreservation is primarily designed for stock keeping. We present a study on bulk cryopreservation of the human iPSC lines UKKi011-A and BIONi010-C-41. By increasing cell concentration and volume, compared to conventional cryopreservation routines in cryo vials, one billion cells were frozen in 50 mL cryo bags. Upon thawing, the cells were immediately seeded in scalable suspension-based bioreactors for expansion to assess the stemness maintenance and for neural differentiation to assess their differentiation potential on the gene and protein levels. Both the conventional and bulk cryo approach show comparative results regarding viability and aggregation upon thawing and bioreactor inoculation. Reduced performance compared to the non-frozen control was compensated within 3 days regarding biomass yield. Stemness was maintained upon thawing in expansion. In neural differentiation, a delay of the neural marker expression on day 4 was compensated at day 9. We conclude that cryopreservation in cryo bags, using high cell concentrations and volumes, does not alter the cells' fate and is a suitable technology to avoid pre-cultivation and enable time- and cost-efficient therapeutic approaches with bulk cell numbers.
Collapse
Affiliation(s)
- Ina Meiser
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Monica Alstrup
- Cell Therapy R&D, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Elham Khalesi
- Cell Therapy R&D, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Bianca Stephan
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Anna M Speicher
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Julia Majer
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Chee Keong Kwok
- Cell Therapy R&D, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Julia C Neubauer
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Mattias Hansson
- Cell Therapy R&D, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
- Department of Molecular and Cellular Biotechnology, Saarland University, 66123 Saarbruecken, Germany
- Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo 1781421, Chile
| |
Collapse
|
3
|
Jankovic MG, Stojkovic M, Bojic S, Jovicic N, Kovacevic MM, Ivosevic Z, Juskovic A, Kovacevic V, Ljujic B. Scaling up human mesenchymal stem cell manufacturing using bioreactors for clinical uses. Curr Res Transl Med 2023; 71:103393. [PMID: 37163885 DOI: 10.1016/j.retram.2023.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are multipotent cells and an attractive therapeutic agent in regenerative medicine and intensive clinical research. Despite the great potential, the limitation that needs to be overcome is the necessity of ex vivo expansion because of insufficient number of hMSCs presented within adult organs and the high doses required for a transplantation. As a result, numerous research studies aim to provide novel expansion methods in order to achieve appropriate numbers of cells with preserved therapeutic quality. Bioreactor-based cell expansion provide high-level production of hMSCs in accordance with good manufacturing practice (GMP) and quality standards. This review summarizes current knowledge about the hMSCs manufacturing platforms with a main focus to the application of bioreactors for large-scale production of GMP-grade hMSCs.
Collapse
Affiliation(s)
- Marina Gazdic Jankovic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia.
| | | | - Sanja Bojic
- Newcastle University, School of Computing, Newcastle upon Tyne, UK
| | - Nemanja Jovicic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Histology and Embryology, Serbia
| | - Marina Miletic Kovacevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Histology and Embryology, Serbia
| | - Zeljko Ivosevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia
| | - Aleksandar Juskovic
- Department of Orthopaedic Surgery, Clinical Centre of Montenegro, 81110 Podgorica, Montenegro
| | - Vojin Kovacevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Surgery, Serbia
| | - Biljana Ljujic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia
| |
Collapse
|
4
|
Torizal FG, Utami T, Lau QY, Inamura K, Nishikawa M, Sakai Y. Dialysis based-culture medium conditioning improved the generation of human induced pluripotent stem cell derived-liver organoid in a high cell density. Sci Rep 2022; 12:20774. [PMID: 36456801 PMCID: PMC9715714 DOI: 10.1038/s41598-022-25325-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Human pluripotent stem cell-derived liver organoids (HLOs) have recently become a promising alternative for liver regenerative therapy. To realize this application, a large amount of human-induced pluripotent stem cells (hiPSCs) derived-liver cells are required for partial liver replacement during transplantation. This method requires stepwise induction using costly growth factors to direct the hiPSCs into the hepatic lineage. Therefore, we developed a simple dialysis-based medium conditioning that fully utilized growth factors accumulation to improve hepatic differentiation of hiPSCs at a high cell density. The results demonstrated that the dialysis culture system could accumulate the four essential growth factors required in each differentiation stage: activin A, bone morphogenetic protein 4 (BMP4), hepatocyte growth factor (HGF), and oncostatin M (OSM). As a result, this low lactate culture environment allowed high-density bipotential hepatic differentiation of up to 4.5 × 107 cells/mL of human liver organoids (HLOs), consisting of hiPSC derived-hepatocyte like cells (HLCs) and cholangiocyte like-cells (CLCs). The differentiated HLOs presented a better or comparable hepatic marker and hepatobiliary physiology to the one that differentiated in suspension culture with routine daily medium replacement at a lower cell density. This simple miniaturized dialysis culture system demonstrated the feasibility of cost-effective high-density hepatic differentiation with minimum growth factor usage.
Collapse
Affiliation(s)
- Fuad Gandhi Torizal
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tia Utami
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Qiao You Lau
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kousuke Inamura
- grid.26999.3d0000 0001 2151 536XDepartment of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masaki Nishikawa
- grid.26999.3d0000 0001 2151 536XDepartment of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yasuyuki Sakai
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
5
|
Chang M, Bogacheva MS, Lou YR. Challenges for the Applications of Human Pluripotent Stem Cell-Derived Liver Organoids. Front Cell Dev Biol 2021; 9:748576. [PMID: 34660606 PMCID: PMC8517247 DOI: 10.3389/fcell.2021.748576] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
The current organoid culture systems allow pluripotent and adult stem cells to self-organize to form three-dimensional (3D) structures that provide a faithful recapitulation of the architecture and function of in vivo organs. In particular, human pluripotent stem cell-derived liver organoids (PSC-LOs) can be used in regenerative medicine and preclinical applications, such as disease modeling and drug discovery. New bioengineering tools, such as microfluidics, biomaterial scaffolds, and 3D bioprinting, are combined with organoid technologies to increase the efficiency of hepatic differentiation and enhance the functional maturity of human PSC-LOs by precise control of cellular microenvironment. Long-term stabilization of hepatocellular functions of in vitro liver organoids requires the combination of hepatic endodermal, endothelial, and mesenchymal cells. To improve the biological function and scalability of human PSC-LOs, bioengineering methods have been used to identify diverse and zonal hepatocyte populations in liver organoids for capturing heterogeneous pathologies. Therefore, constructing engineered liver organoids generated from human PSCs will be an extremely versatile tool in in vitro disease models and regenerative medicine in future. In this review, we aim to discuss the recent advances in bioengineering technologies in liver organoid culture systems that provide a timely and necessary study to model disease pathology and support drug discovery in vitro and to generate cell therapy products for transplantation.
Collapse
Affiliation(s)
- Mingyang Chang
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Mariia S. Bogacheva
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Yan-Ru Lou
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Hernigou P. Bone marrow in orthopaedics (part II): a three hundred and seventy million-year saga from the Devonian to the coronavirus disease 2019 pandemic-osteonecrosis; transplantation; "human chimera"; stem cells, bioreactors, and coronavirus disease. INTERNATIONAL ORTHOPAEDICS 2020; 44:2787-2805. [PMID: 33037446 PMCID: PMC7546933 DOI: 10.1007/s00264-020-04843-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE Three hundred seventy million years ago, bone marrow appeared in skeleton of a fish. More than one hundred years ago, the concept of bone marrow transplantation was proposed to treat human diseases. During the last five decades, this concept became a reality first in hematology and later for orthopaedic diseases. MATERIAL AND METHODS These advances were possible due to the comprehension of the three major components of bone marrow: the fat part, the haematologic part, and the stroma part. Each part has a different history, but the three parts are linked in physiology as in history. RESULTS During many centuries, bone marrow was considered just as food; however, one hundred years ago, the concept of bone marrow transplantation to treat humans was proposed by the French physician Brown-Séquard. During the last five decades, this concept became a reality first in haematology and later for orthopaedic diseases. Transferring what was known from experimental animal models to humans was met with many challenges, the atomic bomb research, and many deaths. Yet through the recognition and subsequent understanding of fundamental processes, medical resiliency, and the determination of a few pioneers, local bone marrow transplantation in orthopaedic surgery became a therapeutic option first for a limited number of diseases and patients. Over the last two decades, mesenchymal stromal cells (MSCs) have been the focus of intense research by acadaemia and industry due to their unique features. MSCs can be easily isolated and expanded through in vitro culture by taking full advantage of their self-renewing capacity. In addition, MSCs exert immunomodulatory effects and can be differentiated into various lineages, which makes them highly attractive for clinical applications in cell-based therapies. CONCLUSION In this review, we attempted to provide a historical overview of bone marrow history, MSC discovery, characterization, and the first clinical studies conducted.
Collapse
Affiliation(s)
- Philippe Hernigou
- Orthopedic Department Henri Mondor Hospital, University Paris East, Paris, France.
| |
Collapse
|
7
|
Numerical Methods for the Design and Description of In Vitro Expansion Processes of Human Mesenchymal Stem Cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2020; 177:185-228. [PMID: 33090237 DOI: 10.1007/10_2020_147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are a valuable source of cells for clinical applications (e.g., treatment of acute myocardial infarction or inflammatory diseases), especially in the field of regenerative medicine. However, for autologous (patient-specific) and allogeneic (off-the-shelf) hMSC-based therapies, in vitro expansion is necessary prior to the clinical application in order to achieve the required cell numbers. Safe, reproducible, and economic in vitro expansion of hMSCs for autologous and allogeneic therapies can be problematic because the cell material is restricted and the cells are sensitive to environmental changes. It is beneficial to collect detailed information on the hydrodynamic conditions and cell growth behavior in a bioreactor system, in order to develop a so called "Digital Twin" of the cultivation system and expansion process. Numerical methods, such as Computational Fluid Dynamics (CFD) which has become widely used in the biotech industry for studying local characteristics within bioreactors or kinetic growth modelling, provide possible solutions for such tasks.In this review, we will present the current state-of-the-art for the in vitro expansion of hMSCs. Different numerical tools, including numerical fluid flow simulations and cell growth modelling approaches for hMSCs, will be presented. In addition, a case study demonstrating the applicability of CFD and kinetic growth modelling for the development of an microcarrier-based hMSC process will be shown.
Collapse
|
8
|
Kim MH, Kino-Oka M. Bioengineering Considerations for a Nurturing Way to Enhance Scalable Expansion of Human Pluripotent Stem Cells. Biotechnol J 2020; 15:e1900314. [PMID: 31904180 DOI: 10.1002/biot.201900314] [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: 07/22/2019] [Revised: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Understanding how defects in mechanotransduction affect cell-to-cell variability will add to the fundamental knowledge of human pluripotent stem cell (hPSC) culture, and may suggest new approaches for achieving a robust, reproducible, and scalable process that result in consistent product quality and yields. Here, the current state of the understanding of the fundamental mechanisms that govern the growth kinetics of hPSCs between static and dynamic cultures is reviewed, the factors causing fluctuations are identified, and culture strategies that might eliminate or minimize the occurrence of cell-to-cell variability arising from these fluctuations are discussed. The existing challenges in the development of hPSC expansion methods for enabling the transition from process development to large-scale production are addressed, a mandatory step for industrial and clinical applications of hPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
9
|
Schwedhelm I, Zdzieblo D, Appelt-Menzel A, Berger C, Schmitz T, Schuldt B, Franke A, Müller FJ, Pless O, Schwarz T, Wiedemann P, Walles H, Hansmann J. Automated real-time monitoring of human pluripotent stem cell aggregation in stirred tank reactors. Sci Rep 2019; 9:12297. [PMID: 31444389 PMCID: PMC6707254 DOI: 10.1038/s41598-019-48814-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 08/13/2019] [Indexed: 12/21/2022] Open
Abstract
The culture of human induced pluripotent stem cells (hiPSCs) at large scale becomes feasible with the aid of scalable suspension setups in continuously stirred tank reactors (CSTRs). Innovative monitoring options and emerging automated process control strategies allow for the necessary highly defined culture conditions. Next to standard process characteristics such as oxygen consumption, pH, and metabolite turnover, a reproducible and steady formation of hiPSC aggregates is vital for process scalability. In this regard, we developed a hiPSC-specific suspension culture unit consisting of a fully monitored CSTR system integrated into a custom-designed and fully automated incubator. As a step towards cost-effective hiPSC suspension culture and to pave the way for flexibility at a large scale, we constructed and utilized tailored miniature CSTRs that are largely made from three-dimensional (3D) printed polylactic acid (PLA) filament, which is a low-cost material used in fused deposition modelling. Further, the monitoring tool for hiPSC suspension cultures utilizes in situ microscopic imaging to visualize hiPSC aggregation in real-time to a statistically significant degree while omitting the need for time-intensive sampling. Suitability of our culture unit, especially concerning the developed hiPSC-specific CSTR system, was proven by demonstrating pluripotency of CSTR-cultured hiPSCs at RNA (including PluriTest) and protein level.
Collapse
Affiliation(s)
- Ivo Schwedhelm
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Daniela Zdzieblo
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Antje Appelt-Menzel
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany
| | - Constantin Berger
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Tobias Schmitz
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Bernhard Schuldt
- University Hospital Schleswig-Holstein, Department of Psychiatry and Psychotherapy, 24105, Kiel, Germany
| | - Andre Franke
- University Hospital Schleswig-Holstein, Institute of Clinical Molecular Biology, 24105, Kiel, Germany
| | - Franz-Josef Müller
- University Hospital Schleswig-Holstein, Department of Psychiatry and Psychotherapy, 24105, Kiel, Germany
| | - Ole Pless
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 22525, Hamburg, Germany
| | - Thomas Schwarz
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany
| | - Philipp Wiedemann
- Mannheim University of Applied Sciences, Institute of Molecular and Cell Biology, 68163, Mannheim, Germany
| | - Heike Walles
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany
| | - Jan Hansmann
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany.
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany.
| |
Collapse
|
10
|
Growth Behavior of Human Adipose Tissue-Derived Stromal/Stem Cells at Small Scale: Numerical and Experimental Investigations. Bioengineering (Basel) 2018; 5:bioengineering5040106. [PMID: 30518117 PMCID: PMC6315405 DOI: 10.3390/bioengineering5040106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 01/25/2023] Open
Abstract
Human adipose tissue-derived stromal/stem cells (hASCs) are a valuable source of cells for clinical applications, especially in the field of regenerative medicine. Therefore, it comes as no surprise that the interest in hASCs has greatly increased over the last decade. However, in order to use hASCs in clinically relevant numbers, in vitro expansion is required. Single-use stirred bioreactors in combination with microcarriers (MCs) have shown themselves to be suitable systems for this task. However, hASCs tend to be less robust, and thus, more shear sensitive than conventional production cell lines for therapeutic antibodies and vaccines (e.g., Chinese Hamster Ovary cells CHO, Baby Hamster Kidney cells BHK), for which these bioreactors were originally designed. Hence, the goal of this study was to investigate the influence of different shear stress levels on the growth of humane telomerase reversed transcriptase immortalized hASCs (hTERT-ASC) and aggregate formation in stirred single-use systems at the mL scale: the 125 mL (= SP100) and the 500 mL (= SP300) disposable Corning® spinner flask. Computational fluid dynamics (CFD) simulations based on an Euler⁻Euler and Euler⁻Lagrange approach were performed to predict the hydrodynamic stresses (0.06⁻0.87 Pa), the residence times (0.4⁻7.3 s), and the circulation times (1.6⁻16.6 s) of the MCs in different shear zones for different impeller speeds and the suspension criteria (Ns1u, Ns1). The numerical findings were linked to experimental data from cultivations studies to develop, for the first time, an unstructured, segregated mathematical growth model for hTERT-ASCs. While the 125 mL spinner flask with 100 mL working volume (SP100) provided up to 1.68.10⁵ hTERT-ASC/cm² (= 0.63 × 10⁶ living hTERT-ASCs/mL, EF 56) within eight days, the peak living cell density of the 500 mL spinner flask with 300 mL working volume (SP300) was 2.46 × 10⁵ hTERT-ASC/cm² (= 0.88 × 10⁶ hTERT-ASCs/mL, EF 81) and was achieved on day eight. Optimal cultivation conditions were found for Ns1u < N < Ns1, which corresponded to specific power inputs of 0.3⁻1.1 W/m³. The established growth model delivered reliable predictions for cell growth on the MCs with an accuracy of 76⁻96% for both investigated spinner flask types.
Collapse
|
11
|
Mesenchymal Stromal Cells: From Discovery to Manufacturing and Commercialization. Stem Cells Int 2018; 2018:4083921. [PMID: 30057622 PMCID: PMC6051015 DOI: 10.1155/2018/4083921] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/01/2018] [Accepted: 03/11/2018] [Indexed: 02/07/2023] Open
Abstract
Over the last decades, mesenchymal stromal cells (MSC) have been the focus of intense research by academia and industry due to their unique features. MSC can be easily isolated and expanded through in vitro culture by taking full advantage of their self-renewing capacity. In addition, MSC exert immunomodulatory effects and can be differentiated into various lineages, which makes them highly attractive for clinical applications in cell-based therapies. In this review, we attempt to provide a brief historical overview of MSC discovery, characterization, and the first clinical studies conducted. The current MSC manufacturing platforms are reviewed with special attention regarding the use of bioreactors for the production of GMP-compliant clinically relevant cell numbers. The first commercial MSC-based products are also addressed, as well as the remaining challenges to the widespread use of MSC-derived products.
Collapse
|
12
|
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
|
13
|
Raju R, Chau D, Cho DS, Park Y, Verfaillie CM, Hu WS. Cell Expansion During Directed Differentiation of Stem Cells Toward the Hepatic Lineage. Stem Cells Dev 2016; 26:274-284. [PMID: 27806669 DOI: 10.1089/scd.2016.0119] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The differentiation of human pluripotent stem cells toward the hepatocyte lineage can potentially provide an unlimited source of functional hepatocytes for transplantation and extracorporeal bioartificial liver applications. It is anticipated that the quantities of cells needed for these applications will be in the order of 109-1010 cells, because of the size of the liver. An ideal differentiation protocol would be to enable directed differentiation to the hepatocyte lineage with simultaneous cell expansion. We introduced a cell expansion stage after the commitment of human embryonic stem cells to the endodermal lineage, to allow for at least an eightfold increase in cell number, with continuation of cell maturation toward the hepatocyte lineage. The progressive changes in the transcriptome were measured by expression array, and the expression dynamics of certain lineage markers was measured by mass cytometry during the differentiation and expansion process. The findings revealed that while cells were expanding they were also capable of progressing in their differentiation toward the hepatocyte lineage. In addition, our transcriptome, protein and functional studies, including albumin secretion, drug-induced CYP450 expression and urea production, all indicated that the hepatocyte-like cells obtained with or without cell expansion are very similar. This method of simultaneous cell expansion and hepatocyte differentiation should facilitate obtaining large quantities of cells for liver cell applications.
Collapse
Affiliation(s)
- Ravali Raju
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - David Chau
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota.,3 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | - Dong Seong Cho
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - Yonsil Park
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - Catherine M Verfaillie
- 4 Department of Development and Regeneration, Stem Cell Institute Leuven , KU Leuven, Leuven, Belgium
| | - Wei-Shou Hu
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
14
|
Weegman BP, Essawy A, Nash P, Carlson AL, Voltzke KJ, Geng Z, Jahani M, Becker BB, Papas KK, Firpo MT. Nutrient Regulation by Continuous Feeding for Large-scale Expansion of Mammalian Cells in Spheroids. J Vis Exp 2016:52224. [PMID: 27768027 PMCID: PMC5092061 DOI: 10.3791/52224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this demonstration, spheroids formed from the β-TC6 insulinoma cell line were cultured as a model of manufacturing a mammalian islet cell product to demonstrate how regulating nutrient levels can improve cell yields. In previous studies, bioreactors facilitated increased culture volumes over static cultures, but no increase in cell yields were observed. Limitations in key nutrients such as glucose, which were consumed between batch feedings, can lead to limitations in cell expansion. Large fluctuations in glucose levels were observed, despite the increase in glucose concentrations in the media. The use of continuous feeding systems eliminated fluctuations in glucose levels, and improved cell growth rates when compared with batch fed static and SSB culture methods. Additional increases in growth rates were observed by adjusting the feed rate based on calculated nutrient consumption, which allowed the maintenance of physiological glucose over three weeks in culture. This method can also be adapted for other cell types.
Collapse
|
15
|
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
|
16
|
Effects of short-term inflammatory and/or hypoxic pretreatments on periodontal ligament stem cells: in vitro and in vivo studies. Cell Tissue Res 2016; 366:311-328. [PMID: 27301447 DOI: 10.1007/s00441-016-2437-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/28/2016] [Indexed: 12/20/2022]
Abstract
In this study, we extensively screened the in vitro and in vivo effects of PDLSCs following short-term inflammatory and/or hypoxic pretreatments. We found that the 24-h hypoxic pretreatment of PDLSCs significantly enhanced cell migration and improved cell surface CXCR4 expression. In addition, hypoxia-pretreated PDLSCs exhibited improved cell colony formation and proliferation. Cells that were dually stimulated also formed more colonies compared to untreated cells but their proliferation did not increase. Importantly, the hypoxic pretreatment of PDLSCs enhanced cell differentiation as determined by elevated RUNX-2 and ALP protein expression. In this context, the inflammatory stimulus impaired cell OCN protein expression, while dual stimuli led to decreased RUNX-2 and OCN mRNA levels. Although preconditioning PDLSCs with inflammatory and/or hypoxic pretreatments resulted in no differences in the production of matrix proteins, hypoxic pretreatment led to the generation of thicker cell sheets; the inflammatory stimulus weakened the ability of cells to form sheets. All the resultant cell sheets exhibited clear bone regeneration following ectopic transplantation as well as in periodontal defect models; the amount of new bone formed by hypoxia-preconditioned cells was significantly greater than that formed by inflammatory stimulus- or dual-stimuli-treated cells or by nonpreconditioned cells. The regeneration of new cementum and periodontal ligaments was only identified in the hypoxia-stimulus and no-stimulus cell groups. Our findings suggest that PDLSCs that undergo short-term hypoxic pretreatment show improved cellular behavior in vitro and enhanced regenerative potential in vivo. The preconditioning of PDLSCs via combined treatments or an inflammatory stimulus requires further investigation.
Collapse
|
17
|
Lambrechts T, Papantoniou I, Viazzi S, Bovy T, Schrooten J, Luyten F, Aerts JM. Evaluation of a monitored multiplate bioreactor for large-scale expansion of human periosteum derived stem cells for bone tissue engineering applications. Biochem Eng J 2016. [DOI: 10.1016/j.bej.2015.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Quantitation of residual trypsin in cell-based therapeutics using immobilized α-1-antitrypsin or SBTI in an ELISA format. J Immunol Methods 2015; 417:131-133. [DOI: 10.1016/j.jim.2014.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/08/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022]
|
19
|
Sullivan DC, Repper JP, Frock AW, McFetridge PS, Petersen BE. Current Translational Challenges for Tissue Engineering: 3D Culture, Nanotechnology, and Decellularized Matrices. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0066-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Liu Y, Ma T. Metabolic regulation of mesenchymal stem cell in expansion and therapeutic application. Biotechnol Prog 2014; 31:468-81. [PMID: 25504836 DOI: 10.1002/btpr.2034] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/28/2014] [Indexed: 12/13/2022]
Abstract
Human mesenchymal or stromal cells (hMSCs) isolated from various adult tissues are primary candidates in cell therapy and tissue regeneration. Despite promising results in preclinical studies, robust therapeutic responses to MSC treatment have not been reproducibly demonstrated in clinical trials. In the translation of MSC-based therapy to clinical application, studies of MSC metabolism have significant implication in optimizing bioprocessing conditions to obtain therapeutically competent hMSC population for clinical application. In addition, understanding the contribution of metabolic cues in directing hMSC fate also provides avenues to potentiate their therapeutic effects by modulating their metabolic properties. This review focuses on MSC metabolism and discusses their unique metabolic features in the context of common metabolic properties shared by stem cells. Recent advances in the fundamental understanding of MSC metabolic characteristics in relation to their in vivo origin and metabolic regulation during proliferation, lineage-specific differentiation, and exposure to in vivo ischemic conditions are summarized. Metabolic strategies in directing MSC fate to enhance their therapeutic potential in tissue engineering and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Yijun Liu
- Dept. of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, 32310
| | | |
Collapse
|
21
|
Sanal MG. A highly efficient method for generation of therapeutic quality human pluripotent stem cells by using naive induced pluripotent stem cells nucleus for nuclear transfer. SAGE Open Med 2014; 2:2050312114550375. [PMID: 26770740 PMCID: PMC4607203 DOI: 10.1177/2050312114550375] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/06/2014] [Indexed: 02/06/2023] Open
Abstract
Even after several years since the discovery of human embryonic stem cells and induced pluripotent stem cells (iPSC), we are still unable to make any significant therapeutic benefits out of them such as cell therapy or generation of organs for transplantation. Recent success in somatic cell nuclear transfer (SCNT) made it possible to generate diploid embryonic stem cells, which opens up the way to make high-quality pluripotent stem cells. However, the process is highly inefficient and hence expensive compared to the generation of iPSC. Even with the latest SCNT technology, we are not sure whether one can make therapeutic quality pluripotent stem cell from any patient's somatic cells or by using oocytes from any donor. Combining iPSC technology with SCNT, that is, by using the nucleus of the candidate somatic cell which got reprogrammed to pluripotent state instead that of the unmodified nucleus of the candidate somatic cell, would boost the efficiency of the technique, and we would be able to generate therapeutic quality pluripotent stem cells. Induced pluripotent stem cell nuclear transfer (iPSCNT) combines the efficiency of iPSC generation with the speed and natural reprogramming environment of SCNT. The new technique may be called iPSCNT. This technique could prove to have very revolutionary benefits for humankind. This could be useful in generating organs for transplantation for patients and for reproductive cloning, especially for childless men and women who cannot have children by any other techniques. When combined with advanced gene editing techniques (such as CRISPR-Cas system) this technique might also prove useful to those who want to have healthy children but suffer from inherited diseases. The current code of ethics may be against reproductive cloning. However, this will change with time as it happened with most of the revolutionary scientific breakthroughs. After all, it is the right of every human to have healthy offspring and it is the question of reproductive freedom and existence.
Collapse
|
22
|
Abstract
Stem cells have emerged as promising tools for the treatment of incurable neural and heart diseases and tissue damage. However, the survival of transplanted stem cells is reported to be low, reducing their therapeutic effects. The major causes of poor survival of stem cells in vivo are linked to anoikis, potential immune rejection, and oxidative damage mediating apoptosis. This review investigates novel methods and potential molecular mechanisms for stem cell preconditioning in vitro to increase their retention after transplantation in damaged tissues. Microenvironmental preconditioning (e.g., hypoxia, heat shock, and exposure to oxidative stress), aggregate formation, and hydrogel encapsulation have been revealed as promising strategies to reduce cell apoptosis in vivo while maintaining biological functions of the cells. Moreover, this review seeks to identify methods of optimizing cell dose preparation to enhance stem cell survival and therapeutic function after transplantation.
Collapse
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory , CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|
23
|
Mohamed MS, Chen Y, Yao CL. A serum-free medium developed for in vitro expansion of murine intestinal stem cells. Biotechnol J 2014; 9:962-70. [PMID: 24802949 DOI: 10.1002/biot.201400016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/25/2014] [Accepted: 05/05/2014] [Indexed: 12/15/2022]
Abstract
Intestinal stem cells (ISCs) are located at the base of the intestinal crypts and have the ability to self-renew as well as to differentiate into mature epithelial cells. Recently, ISCs have received much attention for the treatment of many intestinal diseases. However, many challenges face those studying ISCs because insufficient ISCs are available. Therefore, the development of a culture medium for ISC expansion is an important necessity for basic research and clinical application. In this study, we described the technique used to develop a serum-free medium for expanding ISCs in vitro. Furthermore, five serum substitutes were selected and optimized in order to maintain the long-term proliferation and enteroid-forming ability of ISCs: (i) ethanolamine; (ii) ascorbic acid phosphate; (iii) transferrin; (iv) glutathione; and (v) sodium selenite. Analysis of gene expression of Lgr5, Bmi1, Msi1 and PTEN demonstrated that our serum-free medium sustained the expression of genes involved in ISC-related functions in the expanded ISCs. Additionally, the expression intensity of surface markers, including Lgr5, CD24 and CD44, on serum-free expanded cells in crypts was greatly increased. Taken together, our results demonstrate that the number of ISCs can be expanded and their functionality maintained in our serum-free medium, indicating the suitability of this serum-free expansion medium for increasing the numbers of ICSs available for basic research and clinical applications in the future.
Collapse
Affiliation(s)
- Mahmoud S Mohamed
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan, Taiwan
| | | | | |
Collapse
|
24
|
Liu N, Li Y, Yang ST. Expansion of embryonic stem cells in suspension and fibrous bed bioreactors. J Biotechnol 2014; 178:54-64. [DOI: 10.1016/j.jbiotec.2014.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 02/14/2014] [Accepted: 03/06/2014] [Indexed: 12/23/2022]
|
25
|
Schirmaier C, Jossen V, Kaiser SC, Jüngerkes F, Brill S, Safavi-Nab A, Siehoff A, van den Bos C, Eibl D, Eibl R. Scale-up of adipose tissue-derived mesenchymal stem cell production in stirred single-use bioreactors under low-serum conditions. Eng Life Sci 2014. [DOI: 10.1002/elsc.201300134] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Carmen Schirmaier
- Institute of Biotechnology; Biochemical Engineering and Cell Cultivation Techniques; Zurich University of Applied Sciences; Grüental Wädenswil Switzerland
| | - Valentin Jossen
- Institute of Biotechnology; Biochemical Engineering and Cell Cultivation Techniques; Zurich University of Applied Sciences; Grüental Wädenswil Switzerland
| | - Stephan C. Kaiser
- Institute of Biotechnology; Biochemical Engineering and Cell Cultivation Techniques; Zurich University of Applied Sciences; Grüental Wädenswil Switzerland
| | | | | | | | | | | | - Dieter Eibl
- Institute of Biotechnology; Biochemical Engineering and Cell Cultivation Techniques; Zurich University of Applied Sciences; Grüental Wädenswil Switzerland
| | - Regine Eibl
- Institute of Biotechnology; Biochemical Engineering and Cell Cultivation Techniques; Zurich University of Applied Sciences; Grüental Wädenswil Switzerland
| |
Collapse
|
26
|
Caruso SR, Orellana MD, Mizukami A, Fernandes TR, Fontes AM, Suazo CAT, Oliveira VC, Covas DT, Swiech K. Growth and functional harvesting of human mesenchymal stromal cells cultured on a microcarrier-based system. Biotechnol Prog 2014; 30:889-95. [DOI: 10.1002/btpr.1886] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/27/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Sâmia R. Caruso
- Hemotherapy Center of Ribeirão Preto, School of Medicine of Ribeirão Preto; University of São Paulo; Tenente Catão Roxo Street, 2501 CEP 14051-140 Ribeirão Preto-SP Brazil
| | - Maristela D. Orellana
- Hemotherapy Center of Ribeirão Preto, School of Medicine of Ribeirão Preto; University of São Paulo; Tenente Catão Roxo Street, 2501 CEP 14051-140 Ribeirão Preto-SP Brazil
| | - Amanda Mizukami
- Hemotherapy Center of Ribeirão Preto, School of Medicine of Ribeirão Preto; University of São Paulo; Tenente Catão Roxo Street, 2501 CEP 14051-140 Ribeirão Preto-SP Brazil
| | - Taisa R. Fernandes
- Hemotherapy Center of Ribeirão Preto, School of Medicine of Ribeirão Preto; University of São Paulo; Tenente Catão Roxo Street, 2501 CEP 14051-140 Ribeirão Preto-SP Brazil
| | - Aparecida M. Fontes
- Hemotherapy Center of Ribeirão Preto, School of Medicine of Ribeirão Preto; University of São Paulo; Tenente Catão Roxo Street, 2501 CEP 14051-140 Ribeirão Preto-SP Brazil
- Dept. of Genetics; School of Medicine of Ribeirão Preto; Café av w/n CEP 14040-903 Ribeirão Preto-SP Brazil
| | - Claudio A. T. Suazo
- Dept. of Chemistry Engineering; Federal University of São Carlos; Washington Luís road, km 235 São Carlos Brazil
| | - Viviane C. Oliveira
- Dept. of Dental Materials and Prosthodontics, Faculty of Odontology of Ribeirão Preto; University of São Paulo; Café av w/n CEP 14040-903 Ribeirão Preto-SP Brazil
| | - Dimas T. Covas
- Hemotherapy Center of Ribeirão Preto, School of Medicine of Ribeirão Preto; University of São Paulo; Tenente Catão Roxo Street, 2501 CEP 14051-140 Ribeirão Preto-SP Brazil
- Dept. of Medical Clinic; Faculty of Medicine of Ribeirão Preto; Café av w/n CEP 14040-903 Ribeirão Preto-SP Brazil
| | - Kamilla Swiech
- Dept. of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo; Café av w/n CEP 14040-903 Ribeirão Preto-SP Brazil
| |
Collapse
|
27
|
Sart S, Schneider YJ, Li Y, Agathos SN. Stem cell bioprocess engineering towards cGMP production and clinical applications. Cytotechnology 2014; 66:709-22. [PMID: 24500393 DOI: 10.1007/s10616-013-9687-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 12/31/2013] [Indexed: 12/17/2022] Open
Abstract
Stem cells, including mesenchymal stem cells and pluripotent stem cells, are becoming an indispensable tool for various biomedical applications including drug discovery, disease modeling, and tissue engineering. Bioprocess engineering, targeting large scale production, provides a platform to generate a controlled microenvironment that could potentially recreate the stem cell niche to promote stem cell proliferation or lineage-specific differentiation. This survey aims at defining the characteristics of stem cell populations currently in use and the present-day limits in their applications for therapeutic purposes. Furthermore, a bioprocess engineering strategy based on bioreactors and 3-D cultures is discussed in order to achieve the improved stem cell yield, function, and safety required for production under current good manufacturing practices.
Collapse
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, 2525 Pottsdamer St, Tallahassee, FL, 32310, USA
| | | | | | | |
Collapse
|
28
|
Li Y, Liu M, Yang ST. Dendritic cells derived from pluripotent stem cells: Potential of large scale production. World J Stem Cells 2014; 6:1-10. [PMID: 24567783 PMCID: PMC3927009 DOI: 10.4252/wjsc.v6.i1.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/23/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, are promising sources for hematopoietic cells due to their unlimited growth capacity and the pluripotency. Dendritic cells (DCs), the unique immune cells in the hematopoietic system, can be loaded with tumor specific antigen and used as vaccine for cancer immunotherapy. While autologous DCs from peripheral blood are limited in cell number, hPSC-derived DCs provide a novel alternative cell source which has the potential for large scale production. This review summarizes recent advances in differentiating hPSCs to DCs through the intermediate stage of hematopoietic stem cells. Step-wise growth factor induction has been used to derive DCs from hPSCs either in suspension culture of embryoid bodies (EBs) or in co-culture with stromal cells. To fulfill the clinical potential of the DCs derived from hPSCs, the bioprocess needs to be scaled up to produce a large number of cells economically under tight quality control. This requires the development of novel bioreactor systems combining guided EB-based differentiation with engineered culture environment. Hence, recent progress in using bioreactors for hPSC lineage-specific differentiation is reviewed. In particular, the potential scale up strategies for the multistage DC differentiation and the effect of shear stress on hPSC differentiation in bioreactors are discussed in detail.
Collapse
|
29
|
Expansion of human amniotic fluid stem cells in 3-dimensional fibrous scaffolds in a stirred bioreactor. Biochem Eng J 2014. [DOI: 10.1016/j.bej.2013.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
30
|
Fan Y, Hsiung M, Cheng C, Tzanakakis ES. Facile engineering of xeno-free microcarriers for the scalable cultivation of human pluripotent stem cells in stirred suspension. Tissue Eng Part A 2013; 20:588-99. [PMID: 24098972 DOI: 10.1089/ten.tea.2013.0219] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A prerequisite for the realization of human pluripotent stem cell (hPSC) therapies is the development of bioprocesses for generating clinically relevant quantities of undifferentiated hPSCs and their derivatives under xeno-free conditions. Microcarrier stirred-suspension bioreactors are an appealing modality for the scalable expansion and directed differentiation of hPSCs. Comparative analyses of commercially available microcarriers clearly show the need for developing synthetic substrates supporting the adhesion and growth of hPSCs in three-dimensional cultures under agitation-induced shear. Moreover, the low seeding efficiencies during microcarrier loading with hPSC clusters poses a significant process bottleneck. To that end, a novel protocol was developed increasing hPSC seeding efficiency from 30% to over 80% and substantially shortening the duration of microcarrier loading. Importantly, this method was combined with the engineering of polystyrene microcarriers by surface conjugation of a vitronectin-derived peptide, which was previously shown to support the growth of human embryonic stem cells. Cells proliferated on peptide-conjugated beads in static culture but widespread detachment was observed after exposure to stirring. This prompted additional treatment of the microcarriers with a synthetic polymer commonly used to enhance cell adhesion. hPSCs were successfully cultivated on these microcarriers in stirred suspension vessels for multiple consecutive passages with attachment efficiencies close to 40%. Cultured cells exhibited on average a 24-fold increase in concentration per 6-day passage, over 85% viability, and maintained a normal karyotype and the expression of pluripotency markers such as Nanog, Oct4, and SSEA4. When subjected to spontaneous differentiation in embryoid body cultures or directed differentiation to the three embryonic germ layers, the cells adopted respective fates displaying relevant markers. Lastly, engineered microcarriers were successfully utilized for the expansion and differentiation of hPSCs to mesoderm progeny in stirred suspension vessels. Hence, we demonstrate a strategy for the facile engineering of xeno-free microcarriers for stirred-suspension cultivation of hPSCs. Our findings support the use of microcarrier bioreactors for the scalable, xeno-free propagation and differentiation of human stem cells intended for therapies.
Collapse
Affiliation(s)
- Yongjia Fan
- 1 Department of Chemical and Biological Engineering, State University of New York at Buffalo , Buffalo, New York
| | | | | | | |
Collapse
|
31
|
Weegman BP, Nash P, Carlson AL, Voltzke KJ, Geng Z, Jahani M, Becker BB, Papas KK, Firpo MT. Nutrient regulation by continuous feeding removes limitations on cell yield in the large-scale expansion of Mammalian cell spheroids. PLoS One 2013; 8:e76611. [PMID: 24204645 PMCID: PMC3799778 DOI: 10.1371/journal.pone.0076611] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/25/2013] [Indexed: 02/06/2023] Open
Abstract
Cellular therapies are emerging as a standard approach for the treatment of several diseases. However, realizing the promise of cellular therapies across the full range of treatable disorders will require large-scale, controlled, reproducible culture methods. Bioreactor systems offer the scale-up and monitoring needed, but standard stirred bioreactor cultures do not allow for the real-time regulation of key nutrients in the medium. In this study, β-TC6 insulinoma cells were aggregated and cultured for 3 weeks as a model of manufacturing a mammalian cell product. Cell expansion rates and medium nutrient levels were compared in static, stirred suspension bioreactors (SSB), and continuously fed (CF) SSB. While SSB cultures facilitated increased culture volumes, no increase in cell yields were observed, partly due to limitations in key nutrients, which were consumed by the cultures between feedings, such as glucose. Even when glucose levels were increased to prevent depletion between feedings, dramatic fluctuations in glucose levels were observed. Continuous feeding eliminated fluctuations and improved cell expansion when compared with both static and SSB culture methods. Further improvements in growth rates were observed after adjusting the feed rate based on calculated nutrient depletion, which maintained physiological glucose levels for the duration of the expansion. Adjusting the feed rate in a continuous medium replacement system can maintain the consistent nutrient levels required for the large-scale application of many cell products. Continuously fed bioreactor systems combined with nutrient regulation can be used to improve the yield and reproducibility of mammalian cells for biological products and cellular therapies and will facilitate the translation of cell culture from the research lab to clinical applications.
Collapse
Affiliation(s)
- Bradley P. Weegman
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Peter Nash
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Alexandra L. Carlson
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kristin J. Voltzke
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zhaohui Geng
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Marjan Jahani
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Benjamin B. Becker
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Klearchos K. Papas
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Institute for Cellular Transplantation, Department of Surgery, University of Arizona, Tucson, Arizona, United States of America
| | - Meri T. Firpo
- Stem Cell Institute, Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
32
|
The road to regenerative liver therapies: the triumphs, trials and tribulations. Biotechnol Adv 2013; 31:1085-93. [PMID: 24055818 DOI: 10.1016/j.biotechadv.2013.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 08/07/2013] [Accepted: 08/11/2013] [Indexed: 12/13/2022]
Abstract
The liver is one of the few organs that possess a high capacity to regenerate after liver failure or liver damage. The parenchymal cells of the liver, hepatocytes, contribute to the majority of the regeneration process. Thus, hepatocyte transplantation presents an alternative method to treating liver damage. However, shortage of hepatocytes and difficulties in maintaining primary hepatocytes still remain key obstacles that researchers must overcome before hepatocyte transplantation can be used in clinical practice. The unique properties of pluripotent stem cells (PSCs) and induced pluripotent stem cells (iPSCs) have provided an alternative approach to generating enough functional hepatocytes for cellular therapy. In this review, we will present a brief overview on the current state of hepatocyte differentiation from PSCs and iPSCs. Studies of liver regenerative processes using different cell sources (adult liver stem cells, hepatoblasts, hepatic progenitor cells, etc.) will be described in detail as well as how this knowledge can be applied towards optimizing culture conditions for the maintenance and differentiation of these cells towards hepatocytes. As the outlook of stem cell-derived therapy begins to look more plausible, researchers will need to address the challenges we must overcome in order to translate stem cell research to clinical applications.
Collapse
|
33
|
Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol Adv 2013; 31:1600-23. [PMID: 23962714 DOI: 10.1016/j.biotechadv.2013.08.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/20/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022]
Abstract
Recent technological advances in the generation, characterization, and bioprocessing of human pluripotent stem cells (hPSCs) have created new hope for their use as a source for production of cell-based therapeutic products. To date, a few clinical trials that have used therapeutic cells derived from hESCs have been approved by the Food and Drug Administration (FDA), but numerous new hPSC-based cell therapy products are under various stages of development in cell therapy-specialized companies and their future market is estimated to be very promising. However, the multitude of critical challenges regarding different aspects of hPSC-based therapeutic product manufacturing and their therapies have made progress for the introduction of new products and clinical applications very slow. These challenges include scientific, technological, clinical, policy, and financial aspects. The technological aspects of manufacturing hPSC-based therapeutic products for allogeneic and autologous cell therapies according to good manufacturing practice (cGMP) quality requirements is one of the most important challenging and emerging topics in the development of new hPSCs for clinical use. In this review, we describe main critical challenges and highlight a series of technological advances in all aspects of hPSC-based therapeutic product manufacturing including clinical grade cell line development, large-scale banking, upstream processing, downstream processing, and quality assessment of final cell therapeutic products that have brought hPSCs closer to clinical application and commercial cGMP manufacturing.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | |
Collapse
|
34
|
Liu N, Zang R, Yang ST, Li Y. Stem cell engineering in bioreactors for large-scale bioprocessing. Eng Life Sci 2013. [DOI: 10.1002/elsc.201300013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Ning Liu
- William G. Lowrie Department of Chemical and Biomolecular Engineering; Ohio State University; Columbus OH USA
| | - Ru Zang
- William G. Lowrie Department of Chemical and Biomolecular Engineering; Ohio State University; Columbus OH USA
| | - Shang-Tian Yang
- William G. Lowrie Department of Chemical and Biomolecular Engineering; Ohio State University; Columbus OH USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering; FAMU-FSU College of Engineering; Florida State University; Tallahassee FL USA
| |
Collapse
|
35
|
Liu N, Li Y, Yang ST. Microfibrous carriers for integrated expansion and neural differentiation of embryonic stem cells in suspension bioreactor. Biochem Eng J 2013. [DOI: 10.1016/j.bej.2013.03.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
|
37
|
Thirumala S, Goebel WS, Woods EJ. Manufacturing and banking of mesenchymal stem cells. Expert Opin Biol Ther 2013; 13:673-91. [PMID: 23339745 DOI: 10.1517/14712598.2013.763925] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
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
|
38
|
Wong VW, Sorkin M, Gurtner GC. Enabling stem cell therapies for tissue repair: current and future challenges. Biotechnol Adv 2012. [PMID: 23178704 DOI: 10.1016/j.biotechadv.2012.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Stem cells embody the tremendous potential of the human body to develop, grow, and repair throughout life. Understanding the biologic mechanisms that underlie stem cell-mediated tissue regeneration is key to harnessing this potential. Recent advances in molecular biology, genetic engineering, and material science have broadened our understanding of stem cells and helped bring them closer to widespread clinical application. Specifically, innovative approaches to optimize how stem cells are identified, isolated, grown, and utilized will help translate these advances into effective clinical therapies. Although there is growing interest in stem cells worldwide, this enthusiasm must be tempered by the fact that these treatments remain for the most part clinically unproven. Future challenges include refining the therapeutic manipulation of stem cells, validating these technologies in randomized clinical trials, and regulating the global expansion of regenerative stem cell therapies.
Collapse
Affiliation(s)
- Victor W Wong
- Department of Surgery, Stanford University School of Medicine, 257 Campus Drive, Stanford, CA 94305, USA
| | | | | |
Collapse
|
39
|
Ker DFE, Weiss LE, Junkers SN, Chen M, Yin Z, Sandbothe MF, Huh SI, Eom S, Bise R, Osuna-Highley E, Kanade T, Campbell PG. An engineered approach to stem cell culture: automating the decision process for real-time adaptive subculture of stem cells. PLoS One 2011; 6:e27672. [PMID: 22110715 PMCID: PMC3218005 DOI: 10.1371/journal.pone.0027672] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/21/2011] [Indexed: 11/18/2022] Open
Abstract
Current cell culture practices are dependent upon human operators and remain laborious and highly subjective, resulting in large variations and inconsistent outcomes, especially when using visual assessments of cell confluency to determine the appropriate time to subculture cells. Although efforts to automate cell culture with robotic systems are underway, the majority of such systems still require human intervention to determine when to subculture. Thus, it is necessary to accurately and objectively determine the appropriate time for cell passaging. Optimal stem cell culturing that maintains cell pluripotency while maximizing cell yields will be especially important for efficient, cost-effective stem cell-based therapies. Toward this goal we developed a real-time computer vision-based system that monitors the degree of cell confluency with a precision of 0.791±0.031 and recall of 0.559±0.043. The system consists of an automated phase-contrast time-lapse microscope and a server. Multiple dishes are sequentially imaged and the data is uploaded to the server that performs computer vision processing, predicts when cells will exceed a pre-defined threshold for optimal cell confluency, and provides a Web-based interface for remote cell culture monitoring. Human operators are also notified via text messaging and e-mail 4 hours prior to reaching this threshold and immediately upon reaching this threshold. This system was successfully used to direct the expansion of a paradigm stem cell population, C2C12 cells. Computer-directed and human-directed control subcultures required 3 serial cultures to achieve the theoretical target cell yield of 50 million C2C12 cells and showed no difference for myogenic and osteogenic differentiation. This automated vision-based system has potential as a tool toward adaptive real-time control of subculturing, cell culture optimization and quality assurance/quality control, and it could be integrated with current and developing robotic cell cultures systems to achieve complete automation.
Collapse
Affiliation(s)
- Dai Fei Elmer Ker
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhong JJ, Lee SY. Editorial: Biotechnology Journal shines the spotlight on ACB-2011. Biotechnol J 2011; 6:1298-9. [DOI: 10.1002/biot.201100445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|