1
|
Francius G, Audoux K, Gaye I, Guedon E, Chebil L. Impact of Substrate Surface Chemistry on Wharton's Jelly Mesenchymal Stem Cell Morphological Characteristics, Adherence, and Detachment. ACS APPLIED BIO MATERIALS 2025; 8:4033-4045. [PMID: 40278837 DOI: 10.1021/acsabm.5c00160] [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] [Indexed: 04/26/2025]
Abstract
Using a multidisciplinary approach combining cell biology assays and physicochemical analyses, the effect of surface chemistry, usually involved in 2D and 3D cell cultures, on the adherence and detachment of Wharton's jelly mesenchymal stem cells (WJ-MSC) was clearly characterized. Values calculated from AFM measurements showed that the Young's modulus of the cells depends on the coating substrate: diethylaminoethyl-dextran (DEAE-D) and carboxypolystyrene (cPS). The adhesion of WJ-MSC was influenced by the composition, charge, and Young's modulus of the surface. High adhesion was observed on DEAE-D. Increasing the wall shear stress decreased the proportion of attached cells on DEAE-D down to ∼20%. For the cPS-coated glass substrate, the fraction of detached cells was higher than that for the DEAE-D surfaces, and at the end of the experiment (maximum wall shear stress of 1 Pa), almost complete detachment was achieved. The experimental strategy used in this work provides information in the field of surface chemistry and may help in the selection and design of efficient microcarriers for cell attachment and detachment during bioreactor cultivation.
Collapse
Affiliation(s)
| | - Kévin Audoux
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| | - Ibrahima Gaye
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| | | | - Latifa Chebil
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| |
Collapse
|
2
|
Bodiou V, Cristini N, De Cristofaro L, Pareek T, Rajagopal V, Verrougstraete L, Heinrich JM, Post MJ, Moutsatsou P. Process intensification of cultivated meat production through microcarrier addition strategy optimisation. Sci Rep 2025; 15:14080. [PMID: 40269015 PMCID: PMC12019398 DOI: 10.1038/s41598-025-97813-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
The use of microcarriers (MCs) is currently the most promising method for scaling up bovine satellite cell (bSC) cultures for cultivated meat production. Thanks to the inherent ability of the cells to migrate from one MC to another, also known as bead-to-bead transfer, the need for cell detachment is limited to a minimum, leading to a seamless seeding train. With this study, we aim to intensify the bioprocessing of bSCs in serum-free medium, by exploring the parameters influencing bead-to-bead transfer and cell growth and by optimising the seeding conditions and the MC addition strategy. Keeping production scale bioprocessing requirements into consideration, such as maximisation of fold increase within the same system, we have grown bSCs in up to 80 cm2/ml MC concentrations, using seeding cell densities of 1,000 to 4,750 cells/cm2. We also demonstrated optimisation of the MC addition strategy by determining an optimal confluence range (15,000 to 25,000 cells/cm2) for MC additions and by maximising the MC expansion ratio to 10, without impairing growth. Finally, to ensure scalability of these findings, we successfully applied them at a 3 L bioreactor scale.
Collapse
Affiliation(s)
- Vincent Bodiou
- Mosa Meat BV, Maastricht, Netherlands
- CARIM (Cardiovascular Research Institute Maastricht), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | | | | | | | | | | | | | - Mark J Post
- Mosa Meat BV, Maastricht, Netherlands
- CARIM (Cardiovascular Research Institute Maastricht), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | | |
Collapse
|
3
|
Maldonado VV, Jensen H, Barnes CL, Samsonraj RM. Phenotypic changes associated with continuous long term in vitro expansion of bone marrow-derived mesenchymal stem cells. Biochimie 2025; 234:62-75. [PMID: 40209891 DOI: 10.1016/j.biochi.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
In vitro expansion of mesenchymal stem cells is necessary to obtain a higher cell number for clinical applications. However, long-term expansion can produce significant phenotypic changes on these cells, decreasing their therapeutic utility. Therefore, understanding the phenotypic changes that long-term expansion triggers in mesenchymal stem cells will allow for better and more consistent cell therapy results. Here, we evaluate the phenotypic changes caused by continuous passaging through colony forming unit-fibroblast assay, senescence beta-galactosidase staining, morphology examination, secretome analysis, surface marker expression, protein quantification, osteogenic and adipogenic differentiation, and CD4+ T lymphocyte immunosuppressive potential. Long-term in vitro culture decreases mesenchymal stem cell osteogenic potential and self-renewal, increases cell size, and senescence, but does not consistently affect adipogenic differentiation. Surface marker expression remains similar for positive and negative markers, while secretory phenotype shifts with decreased p14ARF, MMP-3, p21 Waf1/Cip1,ENA-78, GCP-2, GROα, IL-3, IL-7, IL-8, RANTES, TNFβ, and VEGF-A expression, and increased p53, p16 INK4a, MCP-1, and SDF-1 expression. Immunomodulatory potential remains unchanged. These findings can help better understand the phenotypic changes that mesenchymal stem cells undergo while expanded in vitro.
Collapse
Affiliation(s)
- Vitali V Maldonado
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Hanna Jensen
- Department of Surgery, University of Arkansas for Medical Sciences, Northwest Regional Campus, Fayetteville, AR, 72701, USA
| | - C Lowry Barnes
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Rebekah M Samsonraj
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA; Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
4
|
Barekzai J, Refflinghaus L, Okpara M, Tasto L, Tertel T, Giebel B, Czermak P, Salzig D. Process development for the production of mesenchymal stromal cell-derived extracellular vesicles in conventional 2D systems. Cytotherapy 2024; 26:999-1012. [PMID: 38819363 DOI: 10.1016/j.jcyt.2024.04.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND In recent years, the importance of extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) has increased significantly. For their widespread use, a standardized EV manufacturing is needed which often includes conventional, static 2D systems. For these system critical process parameters need to be determined. METHODS We studied the impact of process parameters on MSC proliferation, MSC-derived particle production including EVs, EV- and MSC-specific marker expression, and particle functionality in a HaCaT cell migration assay. RESULTS We found that cell culture growth surface and media affected MSCs and their secretory behavior. Interestingly, the materials that promoted MSC proliferation did not necessarily result in the most functional MSC-derived particles. In addition, we found that MSCs seeded at 4 × 103 cells cm-2 produced particles with improved functional properties compared to higher seeding densities. MSCs in a highly proliferative state did not produce the most particles, although these particles were significantly more effective in promoting HaCaT cell migration. The same correlation was found when investigating the cultivation temperature. A physiological temperature of 37°C was not optimal for particle yield, although it resulted in the most functional particles. We observed a proliferation-associated particle production and found potential correlations between particle production and glucose consumption, enabling the estimation of final particle yields. CONCLUSIONS Our findings suggest that parameters, which must be defined prior to each individual cultivation and do not require complex and expensive equipment, can significantly increase MSC-derived particle production including EVs. Integrating these parameters into a standardized EV process development paves the way for robust and efficient EV manufacturing for early clinical phases.
Collapse
Affiliation(s)
- Jan Barekzai
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Laura Refflinghaus
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Maduwuike Okpara
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Lars Tasto
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany; Faculty of Biology and Chemistry, Justus-Liebig-University of Giessen, Giessen Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany; Faculty of Biology and Chemistry, Justus-Liebig-University of Giessen, Giessen Germany.
| |
Collapse
|
5
|
Thorbow J, Strauch A, Pfening V, Klee JP, Brücher P, Boshof B, Petry F, Czermak P, Herrera Sanchez MB, Salzig D. Large-Scale Expansion of Human Liver Stem Cells Using Two Different Bioreactor Systems. Bioengineering (Basel) 2024; 11:692. [PMID: 39061774 PMCID: PMC11274206 DOI: 10.3390/bioengineering11070692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
The assessment of human liver stem cells (HLSCs) as cell therapeutics requires scalable, controlled expansion processes. We first focused on defining appropriate process parameters for HLSC expansion such as seeding density, use of antibiotics, optimal cell age and critical metabolite concentrations in conventional 2D culture systems. For scale-up, we transferred HLSC expansion to multi-plate and stirred-tank bioreactor systems to determine their limitations. A seeding density of 4000 cells cm-2 was needed for efficient expansion. Although growth was not significantly affected by antibiotics, the concentrations of lactate and ammonia were important. A maximum expansion capacity of at least 20 cumulative population doublings (cPDs) was observed, confirming HLSC growth, identity and functionality. For the expansion of HLSCs in the multi-plate bioreactor system Xpansion (XPN), the oxygen supply strategy was optimized due to a low kLa of 0.076 h-1. The XPN bioreactor yielded a final mean cell density of 94 ± 8 × 103 cells cm-2, more than double that of the standard process in T-flasks. However, in the larger XPN50 device, HLSC density reached only 28 ± 0.9 × 103 cells cm-2, while the glucose consumption rate increased 8-fold. In a fully-controlled 2 L stirred-tank bioreactor (STR), HLSCs expanded at a comparable rate to the T-flask and XPN50 processes in a homogeneous microenvironment using advanced process analytical technology. Ultimately, the scale-up of HLSCs was successful using two different bioreactor systems, resulting in sufficient numbers of viable, functional and undifferentiated HLSCs for therapeutic applications.
Collapse
Affiliation(s)
- Jan Thorbow
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Andrea Strauch
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Viktoria Pfening
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Jan-Philip Klee
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Patricia Brücher
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Björn Boshof
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Florian Petry
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
- Faculty of Biology and Chemistry, University of Giessen, 35392 Giessen, Germany
| | - Maria Beatriz Herrera Sanchez
- 2i3T, Società per la Gestione Dell’Incubatore di Imprese e per il Trasferimento Tecnologico, University of Turin, 10126 Turin, Italy
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany; (J.T.)
| |
Collapse
|
6
|
Kuchemüller KB, Pörtner R, Möller J. Implementation of mDoE-methods to a microcarrier-based expansion processes for mesenchymal stem cells. Biotechnol Prog 2024; 40:e3429. [PMID: 38334218 DOI: 10.1002/btpr.3429] [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: 09/04/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 02/10/2024]
Abstract
The need for advanced therapy medicinal products (ATMPs) has gained increased attention in recent years. In this respect, a well-designed cell expansion process is needed to efficiently manufacture the required number of cells with the desired product quality. This step is challenging due to the biological complexity of the respective primary cell (e.g., mesenchymal stem cells (MSC)) and the usage of microcarrier-based expansion systems. One accelerating approach for process design is model-assisted Design of Experiments (mDoE) combining mathematical process models and statistical tools. In this study, the mDoE workflow was used for the development of an expansion processes with human immortalized mesenchymal stem cells (hMSC-TERT) and the aim of maximizing cell yield assuming only a limited amount of prior knowledge at a very early stage of development. First, suitable microcarriers for expansion in shake flasks were screened and the differentiation of the cells was proven. Second, initial experiments were performed to generate prior knowledge, which was then used to set up the mathematical model and to estimate the model parameters. Finally, the mDoE was used to determine and evaluate the design space to be performed experimentally. Overall, a cell expansion process using microcarriers in a shake flask culture was successfully implemented and a significant increase in cell yield (up to 6,2-fold) was achieved compared to literature.
Collapse
Affiliation(s)
- Kim B Kuchemüller
- Institute of Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Ralf Pörtner
- Institute of Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Johannes Möller
- Institute of Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| |
Collapse
|
7
|
López-Fernández A, Codinach M, Coca MI, Prat-Vidal C, Castaño J, Torrents S, Aran G, Rodríguez L, Querol S, Vives J. Comparability exercise of critical quality attributes of clinical-grade human mesenchymal stromal cells from the Wharton's jelly: single-use stirred tank bioreactors versus planar culture systems. Cytotherapy 2024; 26:418-426. [PMID: 37715777 DOI: 10.1016/j.jcyt.2023.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AIMS The increasing demand of clinical-grade mesenchymal stromal cells (MSCs) for use in advanced therapy medicinal products (ATMPs) require a re-evaluation of manufacturing strategies, ensuring scalability from two-dimensional (2D) surfaces to volumetric (3D) productivities. Herein we describe the design and validation of a Good Manufacturing Practice-compliant 3D culture methodology using microcarriers and 3-L single-use stirred tank bioreactors (STRs) for the expansion of Wharton's jelly (WJ)-derived MSCs in accordance to current regulatory and quality requirements. METHODS MSC,WJ were successfully expanded in 3D and final product characterization was in conformity with Critical Quality Attributes and product specifications previously established for 2D expansion conditions. RESULTS After 6 days of culture, cell yields in the final product from the 3D cultures (mean 9.48 × 108 ± 1.07 × 107 cells) were slightly lower but comparable with those obtained from 2D surfaces (mean 9.73 × 108 ± 2.36 × 108 cells) after 8 days. In all analyzed batches, viability was >90%. Immunophenotype of MSC,WJ was highly positive for CD90 and CD73 markers and lacked of expression of CD31, CD45 and HLA-DR. Compared with 2D expansions, CD105 was detected at lower levels in 3D cultures due to the harvesting procedure from microcarriers involving trypsin at high concentration, and this had no impact on multipotency. Cells presented normal karyotype and strong immunomodulatory potential in vitro. Sterility, Mycoplasma, endotoxin and adventitious virus were negative in both batches produced. CONCLUSIONS In summary, we demonstrated the establishment of a feasible and reproducible 3D bioprocess using single-use STR for clinical-grade MSC,WJ production and provide evidence supporting comparability of 3D versus 2D production strategies. This comparability exercise evaluates the direct implementation of using single-use STR for the scale-up production of MSC,WJ and, by extension, other cell types intended for allogeneic therapies.
Collapse
Affiliation(s)
- Alba López-Fernández
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain; Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Margarita Codinach
- Laboratori Cel·lular, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Maria Isabel Coca
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Cristina Prat-Vidal
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Julio Castaño
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Sílvia Torrents
- Laboratori Cel·lular, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Gemma Aran
- Laboratori Cel·lular, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Luciano Rodríguez
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Sergi Querol
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain; Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
8
|
Ogawa M, Kermani AS, Huynh MJ, Baar K, Leach JK, Block DE. Edible mycelium as proliferation and differentiation support for anchorage-dependent animal cells in cultivated meat production. NPJ Sci Food 2024; 8:23. [PMID: 38693150 PMCID: PMC11063153 DOI: 10.1038/s41538-024-00263-0] [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: 08/25/2023] [Accepted: 03/26/2024] [Indexed: 05/03/2024] Open
Abstract
Cultivated meat production requires bioprocess optimization to achieve cell densities that are multiple orders of magnitude higher compared to conventional cell culture techniques. These processes must maximize resource efficiency and cost-effectiveness by attaining high cell growth productivity per unit of medium. Microcarriers, or carriers, are compatible with large-scale bioreactor use, and offer a large surface-area-to-volume ratio for the adhesion and proliferation of anchorage-dependent animal cells. An ongoing challenge persists in the efficient retrieval of cells from the carriers, with conflicting reports on the effectiveness of trypsinization and the need for additional optimization measures such as carrier sieving. To surmount this issue, edible carriers have been proposed, offering the advantage of integration into the final food product while providing opportunities for texture, flavor, and nutritional incorporation. Recently, a proof of concept (POC) utilizing inactivated mycelium biomass derived from edible filamentous fungus demonstrated its potential as a support structure for myoblasts. However, this POC relied on a model mammalian cell line combination with a single mycelium species, limiting realistic applicability to cultivated meat production. This study aims to advance the POC. We found that the species of fungi composing the carriers impacts C2C12 myoblast cell attachment-with carriers derived from Aspergillus oryzae promoting the best proliferation. C2C12 myoblasts effectively differentiated on mycelium carriers when induced in myogenic differentiation media. Mycelium carriers also supported proliferation and differentiation of bovine satellite cells. These findings demonstrate the potential of edible mycelium carrier technology to be readily adapted in product development within the cultivated meat industry.
Collapse
Affiliation(s)
- Minami Ogawa
- Department of Food Science and Technology, University of California, Davis, Davis, CA, 95616, USA
| | - Alex S Kermani
- Department of Materials Science and Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Mayrene J Huynh
- Department of Food Science and Technology, University of California, Davis, Davis, CA, 95616, USA
| | - Keith Baar
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA, 95616, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - David E Block
- Department of Chemical Engineering, University of California, Davis, Davis, CA, 95616, USA.
- Department of Viticulture and Enology, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
9
|
López-Fernández A, Garcia-Gragera V, Lecina M, Vives J. Identification of critical process parameters for expansion of clinical grade human Wharton's jelly-derived mesenchymal stromal cells in stirred-tank bioreactors. Biotechnol J 2024; 19:e2300381. [PMID: 38403461 DOI: 10.1002/biot.202300381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 02/27/2024]
Abstract
Cell therapies based on multipotent mesenchymal stromal cells (MSCs) are traditionally produced using 2D culture systems and platelet lysate- or serum-containing media (SCM). Although cost-effective for single-dose autologous treatments, this approach is not suitable for larger scale manufacturing (e.g., multiple-dose autologous or allogeneic therapies with banked MSCs); automated, scalable and Good Manufacturing Practices (GMP)-compliant platforms are urgently needed. The feasibility of transitioning was evaluated from an established Wharton's jelly MSCs (WJ-MSCs) 2D production strategy to a new one with stirred-tank bioreactors (STRs). Experimental conditions included four GMP-compliant xeno- and serum-free media (XSFM) screened in 2D conditions and two GMP-grade microcarriers assessed in 0.25 L-STRs using SCM. From the screening, a XSFM was selected and compared against SCM using the best-performing microcarrier. It was observed that SCM outperformed the 2D-selected medium in STRs, reinforcing the importance of 2D-to-3D transition studies before translation into clinical production settings. It was also found that attachment efficiency and microcarrier colonization were essential to attain higher fold expansions, and were therefore defined as critical process parameters. Nevertheless, WJ-MSCs were readily expanded in STRs with both media, preserving critical quality attributes in terms of identity, viability and differentiation potency, and yielding up to 1.47 × 109 cells in a real-scale 2.4-L batch.
Collapse
Affiliation(s)
- Alba López-Fernández
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
| | - Víctor Garcia-Gragera
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Engineering Materials Group (GEMAT), Bioprocessing Lab, IQS School of Engineering, Universitat Ramón Llull, Barcelona, Spain
| | - Martí Lecina
- Engineering Materials Group (GEMAT), Bioprocessing Lab, IQS School of Engineering, Universitat Ramón Llull, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel·lular i Avançada, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
Kaneko M, Sato A, Ayano S, Fujita A, Kobayashi G, Ito A. Expansion of human mesenchymal stem cells on poly(vinyl alcohol) microcarriers. J Biosci Bioeng 2023; 136:407-414. [PMID: 37657971 DOI: 10.1016/j.jbiosc.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023]
Abstract
Microcarriers provide a high surface-area-to-volume ratio that can realize high yields of cell products, including human mesenchymal stem cells (hMSCs). Here, we report a novel poly(vinyl alcohol) (PVA)-based microcarrier for hMSC expansion in suspension culture. PVA microcarriers were prepared as collagen-coated PVA hydrogels 181 μm in size and a high surface-area-to-weight ratio of 2945 cm2/g. The PVA microcarriers supported a 2.6-fold expansion of hMSCs in a 30-mL single-use stirred bioreactor after a 7 d culture period, comparable to that of commercially available microcarriers. Interestingly, we observed that hMSCs on PVA microcarriers adhered to adjacent microcarriers, resulting in the aggregation of hMSC-PVA microcarriers. Therefore, we conducted a long-term expansion culture using a bead-to-bead cell transfer method with PVA microcarriers. Fresh microcarriers were added to the cell-populated microcarriers in the bioreactor on days 7 and 14. hMSCs on PVA microcarriers continued to grow for 21 d using the bead-to-bead cell transfer method. Furthermore, magnetic PVA (PVA-mag) microcarriers were developed by loading magnetic nanoparticles into PVA microcarriers, and we demonstrated that these PVA-mag microcarriers enabled cell recovery by magnetic separation. These results suggest that these PVA microcarriers can contribute to the large-scale culture of hMSCs for regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Masahiro Kaneko
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Airi Sato
- College of Bioscience and Biotechnology, Chubu University, 1200 Matsumoto, Kasugai, Aichi 487-8501, Japan
| | - Satoru Ayano
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Akio Fujita
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Goro Kobayashi
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Akira Ito
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
| |
Collapse
|
11
|
Teale MA, Schneider S, Eibl D, van den Bos C, Neubauer P, Eibl R. Mesenchymal and induced pluripotent stem cell-based therapeutics: a comparison. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12583-4. [PMID: 37246986 DOI: 10.1007/s00253-023-12583-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/30/2023]
Abstract
Stem cell-based cell therapeutics and especially those based on human mesenchymal stem cells (hMSCs) and induced pluripotent stem cells (hiPSCs) are said to have enormous developmental potential in the coming years. Their applications range from the treatment of orthopedic disorders and cardiovascular diseases to autoimmune diseases and even cancer. However, while more than 27 hMSC-derived therapeutics are currently commercially available, hiPSC-based therapeutics have yet to complete the regulatory approval process. Based on a review of the current commercially available hMSC-derived therapeutic products and upcoming hiPSC-derived products in phase 2 and 3, this paper compares the cell therapy manufacturing process between these two cell types. Moreover, the similarities as well as differences are highlighted and the resulting impact on the production process discussed. Here, emphasis is placed on (i) hMSC and hiPSC characteristics, safety, and ethical aspects, (ii) their morphology and process requirements, as well as (iii) their 2- and 3-dimensional cultivations in dependence of the applied culture medium and process mode. In doing so, also downstream processing aspects are covered and the role of single-use technology is discussed. KEY POINTS: • Mesenchymal and induced pluripotent stem cells exhibit distinct behaviors during cultivation • Single-use stirred bioreactor systems are preferred for the cultivation of both cell types • Future research should adapt and modify downstream processes to available single-use devices.
Collapse
Affiliation(s)
- Misha A Teale
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland.
| | - Samuel Schneider
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| | - Dieter Eibl
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| | | | - Peter Neubauer
- Institute of Biotechnology, Chair of Bioprocess Engineering, Technical University of Berlin, ACK24, Ackerstraße 76, 13355, Berlin, Germany
| | - Regine Eibl
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| |
Collapse
|
12
|
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: 7] [Impact Index Per Article: 3.5] [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
|
13
|
Xiang Y, Yan J, Bao X, Gleadall A, Roach P, Sun T. Evaluation of Polymeric Particles for Modular Tissue Cultures in Developmental Engineering. Int J Mol Sci 2023; 24:ijms24065234. [PMID: 36982306 PMCID: PMC10049291 DOI: 10.3390/ijms24065234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Developmental engineering (DE) aims to culture mammalian cells on corresponding modular scaffolds (scale: micron to millimeter), then assemble these into functional tissues imitating natural developmental biology processes. This research intended to investigate the influences of polymeric particles on modular tissue cultures. When poly(methyl methacrylate) (PMMA), poly(lactic acid) (PLA) and polystyrene (PS) particles (diameter: 5-100 µm) were fabricated and submerged in culture medium in tissue culture plastics (TCPs) for modular tissue cultures, the majority of adjacent PMMA, some PLA but no PS particles aggregated. Human dermal fibroblasts (HDFs) could be directly seeded onto large (diameter: 30-100 µm) PMMA particles, but not small (diameter: 5-20 µm) PMMA, nor all the PLA and PS particles. During tissue cultures, HDFs migrated from the TCPs surfaces onto all the particles, while the clustered PMMA or PLA particles were colonized by HDFs into modular tissues with varying sizes. Further comparisons revealed that HDFs utilized the same cell bridging and stacking strategies to colonize single or clustered polymeric particles, and the finely controlled open pores, corners and gaps on 3D-printed PLA discs. These observed cell-scaffold interactions, which were then used to evaluate the adaptation of microcarrier-based cell expansion technologies for modular tissue manufacturing in DE.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Jiongyi Yan
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Xujin Bao
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Andrew Gleadall
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Paul Roach
- Department of Chemistry, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Tao Sun
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| |
Collapse
|
14
|
Sakisaka Y, Ishihata H, Maruyama K, Nemoto E, Chiba S, Nagamine M, Hasegawa H, Hatsuzawa T, Yamada S. Serial Cultivation of an MSC-Like Cell Line with Enzyme-Free Passaging Using a Microporous Titanium Scaffold. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1165. [PMID: 36770173 PMCID: PMC9919603 DOI: 10.3390/ma16031165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
In vitro studies on adherent cells require a process of passage to dissociate the cells from the culture substrate using enzymes or other chemical agents to maintain cellular activity. However, these proteolytic enzymes have a negative influence on the viability and phenotype of cells. The mesenchymal stem cell (MSC)-like cell line, C3H10T1/2, adhered, migrated, and proliferated to the same extent on newly designed microporous titanium (Ti) membrane and conventional culture dish, and spontaneous transfer to another substrate without enzymatic or chemical dissociation was achieved. The present study pierced a 10 μm-thick pure Ti sheet with 25 μm square holes at 75 μm intervals to create a dense porous structure with biomimetic topography. The pathway of machined holes allowed the cells to access both sides of the membrane frequently. In a culture with Ti membranes stacked above- and below-seeded cells, cell migration between the neighboring membranes was confirmed using the through-holes of the membrane and contact between the membranes as migration routes. Furthermore, the cells on each membrane migrated onto the conventional culture vessel. Therefore, a cell culture system with enzyme-free passaging was developed.
Collapse
Affiliation(s)
- Yukihiko Sakisaka
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, 4-1 Seiryou-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hiroshi Ishihata
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, 4-1 Seiryou-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kentaro Maruyama
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, 4-1 Seiryou-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Eiji Nemoto
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, 4-1 Seiryou-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Shigeki Chiba
- Nagamine Manufacturing Co., Ltd., 1725-26, Kishinoue, Manno-cho, Nakatado-gun, Kagawa 766-0026, Japan
| | - Masaru Nagamine
- Nagamine Manufacturing Co., Ltd., 1725-26, Kishinoue, Manno-cho, Nakatado-gun, Kagawa 766-0026, Japan
| | - Hiroshi Hasegawa
- Department of Oral Surgery and Dentistry, Fukushima Medical University, 1, Hikariga-oka, Fukushima 960-1295, Japan
| | - Takeshi Hatsuzawa
- Laboratory for Future Interdisciplinary Research of Science and Technology, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Satoru Yamada
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, 4-1 Seiryou-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
15
|
Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and Mesenchymal Stem Cells: Basic Concepts, Challenges and Strategies. BIOLOGY 2022; 11:1678. [PMID: 36421393 PMCID: PMC9687158 DOI: 10.3390/biology11111678] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 08/27/2023]
Abstract
Aging and frailty are complex processes implicating multifactorial mechanisms, such as replicative senescence, oxidative stress, mitochondrial dysfunction, or autophagy disorder. All of these mechanisms drive dramatic changes in the tissue environment, such as senescence-associated secretory phenotype factors and inflamm-aging. Thus, there is a demand for new therapeutic strategies against the devastating effects of the aging and associated diseases. Mesenchymal stem cells (MSC) participate in a "galaxy" of tissue signals (proliferative, anti-inflammatory, and antioxidative stress, and proangiogenic, antitumor, antifibrotic, and antimicrobial effects) contributing to tissue homeostasis. However, MSC are also not immune to aging. Three strategies based on MSC have been proposed: remove, rejuvenate, or replace the senescent MSC. These strategies include the use of senolytic drugs, antioxidant agents and genetic engineering, or transplantation of younger MSC. Nevertheless, these strategies may have the drawback of the adverse effects of prolonged use of the different drugs used or, where appropriate, those of cell therapy. In this review, we propose the new strategy of "Exogenous Restitution of Intercellular Signalling of Stem Cells" (ERISSC). This concept is based on the potential use of secretome from MSC, which are composed of molecules such as growth factors, cytokines, and extracellular vesicles and have the same biological effects as their parent cells. To face this cell-free regenerative therapy challenge, we have to clarify key strategy aspects, such as establishing tools that allow us a more precise diagnosis of aging frailty in order to identify the therapeutic requirements adapted to each case, identify the ideal type of MSC in the context of the functional heterogeneity of these cellular populations, to optimize the mass production and standardization of the primary materials (cells) and their secretome-derived products, to establish the appropriate methods to validate the anti-aging effects and to determine the most appropriate route of administration for each case.
Collapse
Affiliation(s)
- Maria Fraile
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Luis A. Costa
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Arancha Martín
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Emergency, Hospital Universitario de Cabueñes, Los Prados, 395, 33394 Gijon, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Surgery, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| |
Collapse
|
16
|
Wu Y, Zheng Y, Jin Z, Li S, Wu W, An C, Guo J, Zhu Z, Zhou T, Zhou Y, Cen L. Controllable manipulation of alginate-gelatin core-shell microcarriers for HUMSCs expansion. Int J Biol Macromol 2022; 216:1-13. [PMID: 35777503 DOI: 10.1016/j.ijbiomac.2022.06.173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/20/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
Human umbilical cord mesenchymal stem cells (HUMSCs) are one of the most attractive sources of stem cells, and it is meaningful to design and develop a type of microcarriers with suitable mechanical strength for HUMSCs proliferation in order to acquire enough cells for cell-based therapy. Alginate-gelatin core-shell (AG) soft microcarriers were thus fabricated via a microfluidic device with droplet shearing/gelation facilities and surface coating for in vitro expansion of HUMSCs. The attachment and proliferation of HUMSCs on AG microcarriers with different mechanical strengths modulated by gelatin coating was studied, and the harvested cells were characterized to verity their differentiation potential. The obtained core-shell microcarriers were all uniform in size with a high mono-dispersity (CV < 5 %). An increase in the gelatin surface coating concentration from 0.5 % to 1.5 % would lead to the reduction in both the particle size of the microcarriers and swelling ratio upon the contact of culture medium, but increased elastic modulus. Microcarriers of 245.12 μm with a gelatin coating elastic modulus of 27.5 kPa (AG10) were found to be the optimal substrate for HUMSCs with an initial attachment efficiency of 44.41 % and a 5-day expansion efficiency of 647 %. The cells harvested from AG10 still reserved their outstanding pluripotency. Fresh AG10 could smoothly transfer cells from a running microcarrier-cell system of confluence to serve as a convenient way of scaling-up the existing culture. The current study thus developed suitable microcarriers, AG10, for in vitro HUMSCs expansion with well reserve of cell multipotency, and also provided a manufacturing and surface manipulating strategy of precise production and fine regulation of microcarrier properties.
Collapse
Affiliation(s)
- Yanfei Wu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Yiling Zheng
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Ziyang Jin
- State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Shihao Li
- State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Weiqian Wu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Chenjing An
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Jiahao Guo
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Zhihua Zhu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China
| | - Tian Zhou
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China..
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China..
| | - Lian Cen
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai 200237, China.
| |
Collapse
|
17
|
Timsina H, McTyer J, Rao RR, Almodovar J. A comparative evaluation of layer-by-layer assembly techniques for surface modification of microcarriers used in human mesenchymal stromal cell manufacturing. Biotechnol J 2022; 17:e2100605. [PMID: 35377534 DOI: 10.1002/biot.202100605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/16/2022] [Accepted: 04/01/2022] [Indexed: 11/06/2022]
Abstract
The demand for large quantities of highly potent human mesenchymal stromal cells (hMSCs) is growing given their therapeutic potential. To meet high production needs, suspension-based cell cultures using microcarriers are commonly used. Microcarriers are commonly made of or coated with extracellular matrix proteins or charged compounds to promote cell adhesion and proliferation. In this work, we demonstrate a simple method (draining filter) to perform layer by layer (LbL) assembly on microcarriers to create multilayers of heparin and collagen and further demonstrate that these multilayers have a positive effect on hMSC viability after 48 hours of culture. The draining filter method is evaluated against two other methods found in literature - centrifugation and fluidized bed, showing that the draining filter method can perform the surface modification with greater efficiency and with less materials and steps needed in the coating process. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hemanta Timsina
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Jasmine McTyer
- Department of Chemical and Biomolecular Engineering, Clemson University, 127 Earle Hall, Clemson, SC, 29634, USA
| | - Raj R Rao
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Jorge Almodovar
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| |
Collapse
|
18
|
Pajčin I, Knežić T, Savic Azoulay I, Vlajkov V, Djisalov M, Janjušević L, Grahovac J, Gadjanski I. Bioengineering Outlook on Cultivated Meat Production. MICROMACHINES 2022; 13:402. [PMID: 35334693 PMCID: PMC8950996 DOI: 10.3390/mi13030402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023]
Abstract
Cultured meat (also referred to as cultivated meat or cell-based meat)-CM-is fabricated through the process of cellular agriculture (CA), which entails application of bioengineering, i.e., tissue engineering (TE) principles to the production of food. The main TE principles include usage of cells, grown in a controlled environment provided by bioreactors and cultivation media supplemented with growth factors and other needed nutrients and signaling molecules, and seeded onto the immobilization elements-microcarriers and scaffolds that provide the adhesion surfaces necessary for anchor-dependent cells and offer 3D organization for multiple cell types. Theoretically, many solutions from regenerative medicine and biomedical engineering can be applied in CM-TE, i.e., CA. However, in practice, there are a number of specificities regarding fabrication of a CM product that needs to fulfill not only the majority of functional criteria of muscle and fat TE, but also has to possess the sensory and nutritional qualities of a traditional food component, i.e., the meat it aims to replace. This is the reason that bioengineering aimed at CM production needs to be regarded as a specific scientific discipline of a multidisciplinary nature, integrating principles from biomedical engineering as well as from food manufacturing, design and development, i.e., food engineering. An important requirement is also the need to use as little as possible of animal-derived components in the whole CM bioprocess. In this review, we aim to present the current knowledge on different bioengineering aspects, pertinent to different current scientific disciplines but all relevant for CM engineering, relevant for muscle TE, including different cell sources, bioreactor types, media requirements, bioprocess monitoring and kinetics and their modifications for use in CA, all in view of their potential for efficient CM bioprocess scale-up. We believe such a review will offer a good overview of different bioengineering strategies for CM production and will be useful to a range of interested stakeholders, from students just entering the CA field to experienced researchers looking for the latest innovations in the field.
Collapse
Affiliation(s)
- Ivana Pajčin
- Department of Biotechnology and Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia; (I.P.); (V.V.); (J.G.)
| | - Teodora Knežić
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| | - Ivana Savic Azoulay
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Vanja Vlajkov
- Department of Biotechnology and Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia; (I.P.); (V.V.); (J.G.)
| | - Mila Djisalov
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| | - Ljiljana Janjušević
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| | - Jovana Grahovac
- Department of Biotechnology and Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia; (I.P.); (V.V.); (J.G.)
| | - Ivana Gadjanski
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| |
Collapse
|
19
|
Peng L, Gautrot JE. Long term expansion profile of mesenchymal stromal cells at protein nanosheet-stabilised bioemulsions for next generation cell culture microcarriers. Mater Today Bio 2021; 12:100159. [PMID: 34841241 PMCID: PMC8605361 DOI: 10.1016/j.mtbio.2021.100159] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 12/27/2022] Open
Abstract
Tremendous progress in the identification, isolation and expansion of stem cells has allowed their application in regenerative medicine and tissue engineering, and their use as advanced in vitro models. As a result, stem cell manufacturing increasingly requires scale up, parallelisation and automation. However, solid substrates currently used for the culture of adherent cells are poorly adapted for such applications, owing to their difficult processing from cell products, relatively high costs and their typical reliance on difficult to recycle plastics and microplastics. In this work, we show that bioemulsions formed of microdroplets stabilised by protein nanosheets displaying strong interfacial mechanics are well-suited for the scale up of adherent stem cells such as mesenchymal stromal cells (MSCs). We demonstrate that, over multiple passages (up to passage 10), MSCs retain comparable phenotypes when cultured on such bioemulsions, solid microcarriers (Synthemax II) and classic 2D tissue culture polystyrene. Phenotyping (cell proliferation, morphometry, flow cytometry and differentiation assays) of MSCs cultured for multiple passages on these systems indicate that, although stemness is lost at late passages when cultured on these different substrates, stem cell phenotypes remained comparable between different culture conditions, at any given passage. Hence our study validates the use of bioemulsions for the long term expansion of adherent stem cells and paves the way to the design of novel 3D bioreactors based on microdroplet microcarriers.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Julien E Gautrot
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| |
Collapse
|
20
|
Teale M, Jossen V, Eibl D, Eibl R. Chemically Defined, Xeno-Free Expansion of Human Mesenchymal Stem Cells (hMSCs) on Benchtop-Scale Using a Stirred Single-Use Bioreactor. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2436:83-111. [PMID: 34611815 DOI: 10.1007/7651_2021_426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In recent years, the use of hMSCs, which may be isolated from adipose tissue among others, for the treatment of diseases has increased significantly. The cell quantities required for such therapeutic approaches, between 1012 and 1013, have thus far been predominantly produced using commercially available multi-tray systems, such as the Cell Factory (Thermo Fisher Scientific) or HYPERStack (Corning), which can be purchased with up to 40 layers. However, the handling of these planar multilayer systems is difficult, and process monitoring opportunities remain limited. Here, automated stirred single-use bioreactors provide a viable alternative to the time-consuming multiplication of cells using such planar systems, while still managing to achieve the desired clinically relevant quantities. In these stirred single-use systems, adherent cells are predominantly cultivated in suspension up to pilot scale using carrier materials, also referred to as microcarriers (MCs).This chapter describes the steps which need to be realized to guarantee successful hMSC expansion within a stirred single-use bioreactor (Eppendorf's BioBLU® 0.3c) operated using MCs under serum- and xeno-free conditions at benchtop scale. The cultivations were performed using an immortalized human adipose-derived mesenchymal stem cell (hASC) line, hence referred to as hASC52telo, and a new chemically defined, xeno-free medium, hence referred to as the UrSuppe formulation. Spinner flask cultivations were performed under comparable process conditions.
Collapse
Affiliation(s)
- Misha Teale
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland.
| | - Valentin Jossen
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Dieter Eibl
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Regine Eibl
- Centre for Biochemical Engineering and Cell Cultivation Techniques, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| |
Collapse
|
21
|
Rogers RE, Haskell A, White BP, Dalal S, Lopez M, Tahan D, Pan S, Kaur G, Kim H, Barreda H, Woodard SL, Benavides OR, Dai J, Zhao Q, Maitland KC, Han A, Nikolov ZL, Liu F, Lee RH, Gregory CA, Kaunas R. A scalable system for generation of mesenchymal stem cells derived from induced pluripotent cells employing bioreactors and degradable microcarriers. Stem Cells Transl Med 2021; 10:1650-1665. [PMID: 34505405 PMCID: PMC8641084 DOI: 10.1002/sctm.21-0151] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are effective in treating disorders resulting from an inflammatory or heightened immune response. The hMSCs derived from induced pluripotent stem cells (ihMSCs) share the characteristics of tissue derived hMSCs but lack challenges associated with limited tissue sources and donor variation. To meet the expected future demand for ihMSCs, there is a need to develop scalable methods for their production at clinical yields while retaining immunomodulatory efficacy. Herein, we describe a platform for the scalable expansion and rapid harvest of ihMSCs with robust immunomodulatory activity using degradable gelatin methacryloyl (GelMA) microcarriers. GelMA microcarriers were rapidly and reproducibly fabricated using a custom microfluidic step emulsification device at relatively low cost. Using vertical wheel bioreactors, 8.8 to 16.3‐fold expansion of ihMSCs was achieved over 8 days. Complete recovery by 5‐minute digestion of the microcarriers with standard cell dissociation reagents resulted in >95% viability. The ihMSCs matched or exceeded immunomodulatory potential in vitro when compared with ihMSCs expanded on monolayers. This is the first description of a robust, scalable, and cost‐effective method for generation of immunomodulatory ihMSCs, representing a significant contribution to their translational potential.
Collapse
Affiliation(s)
- Robert E Rogers
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Andrew Haskell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Berkley P White
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| | - Sujata Dalal
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Megan Lopez
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Daniel Tahan
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Simin Pan
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Gagandeep Kaur
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Hyemee Kim
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Heather Barreda
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Susan L Woodard
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA
| | - Oscar R Benavides
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| | - Jing Dai
- Department of Electrical and Computer Engineering, Texas A&M University, Wisenbaker Engineering Building, College Station, Texas, USA
| | - Qingguo Zhao
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Kristen C Maitland
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| | - Arum Han
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA.,Department of Electrical and Computer Engineering, Texas A&M University, Wisenbaker Engineering Building, College Station, Texas, USA
| | - Zivko L Nikolov
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA.,Biological and Agricultural Engineering, Texas A&M University, Scoates Hall, College Station, Texas, USA
| | - Fei Liu
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Ryang Hwa Lee
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Roland Kaunas
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| |
Collapse
|
22
|
Abdul Halim NSS, Yahaya BH, Lian J. Therapeutic Potential of Adipose-Derived Stem Cells in the Treatment of Pulmonary Diseases. Curr Stem Cell Res Ther 2021; 17:103-112. [PMID: 34387168 DOI: 10.2174/1574888x16666210812145202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/07/2021] [Accepted: 05/25/2021] [Indexed: 11/22/2022]
Abstract
Stem cells derived from adipose tissues (ADSCs) have emerged as an ideal candidate for various models of respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome. ADSCs have qualities that may make them better suited for treating inflammatory lung diseases than other MSCs. ADSCs show a lower senescence ratio, higher proliferative capacity and stability in terms of their genetic and morphology during long-term culture over bone marrow-derived mesenchymal stem cells (BMMSCs). With advanced research techniques, the advantageous effects of ADSCs seem limited to their ability to engraft, differentiate, and be related to their secretion of trophic factors. These trophic factors regulate the therapeutic and regenerative outcomes in various lung inflammatory diseases. Taken together, these particular qualities of ADSCs make them significantly relevant for clinical applications. This article discusses a recent advance of ADSCs biology and their translational application emphasizing their anti-inflammatory, immunomodulatory and regenerative properties particularly on lung inflammatory diseases. Besides, the relevant advancements made in the field, the regulatory aspects, and other challenges and obstacles will be highlighted.
Collapse
Affiliation(s)
- Nur Shuhaidatul Sarmiza Abdul Halim
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200 Kepala Batas, Penang . Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200 Kepala Batas, Penang . Malaysia
| | - Jie Lian
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200 Kepala Batas, Penang . Malaysia
| |
Collapse
|
23
|
Petry F, Salzig D. Impact of Bioreactor Geometry on Mesenchymal Stem Cell Production in Stirred‐Tank Bioreactors. CHEM-ING-TECH 2021. [DOI: 10.1002/cite.202100041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Florian Petry
- University of Applied Sciences Mittelhessen Institute of Bioprocess Engineering and Pharmaceutical Technology Wiesenstraße 14 35390 Giessen Germany
| | - Denise Salzig
- University of Applied Sciences Mittelhessen Institute of Bioprocess Engineering and Pharmaceutical Technology Wiesenstraße 14 35390 Giessen Germany
| |
Collapse
|
24
|
Tsai AC, Pacak CA. Bioprocessing of Human Mesenchymal Stem Cells: From Planar Culture to Microcarrier-Based Bioreactors. Bioengineering (Basel) 2021; 8:bioengineering8070096. [PMID: 34356203 PMCID: PMC8301102 DOI: 10.3390/bioengineering8070096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 01/14/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) have demonstrated great potential to be used as therapies for many types of diseases. Due to their immunoprivileged status, allogeneic hMSCs therapies are particularly attractive options and methodologies to improve their scaling and manufacturing are needed. Microcarrier-based bioreactor systems provide higher volumetric hMSC production in automated closed systems than conventional planar cultures. However, more sophisticated bioprocesses are necessary to successfully convert from planar culture to microcarriers. This article summarizes key steps involved in the planar culture to microcarrier hMSC manufacturing scheme, from seed train, inoculation, expansion and harvest. Important bioreactor parameters, such as temperature, pH, dissolved oxygen (DO), mixing, feeding strategies and cell counting techniques, are also discussed.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Pediatrics, University of Florida, Gainesville, FL 32603, USA
- Correspondence: (A.-C.T.); (C.A.P.)
| | - Christina A. Pacak
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: (A.-C.T.); (C.A.P.)
| |
Collapse
|
25
|
Kim JY, Rhim WK, Seo HJ, Lee JY, Park CG, Han DK. Comparative Analysis of MSC-Derived Exosomes Depending on Cell Culture Media for Regenerative Bioactivity. Tissue Eng Regen Med 2021; 18:355-367. [PMID: 34047999 DOI: 10.1007/s13770-021-00352-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In order to produce and isolate the exosome derived from the cell of interests, a serum free environment (starvation) has been essential for excluding the unknown effect from serum-derived exosomes. Recently, serum-free culture media have been developed as a substitute for serum supplemented media so that MSC proliferates with maintaining the original characteristics of the cells in a serum free condition. Due to the different properties of the exosomes representing the states and characteristics of the origin cells, a study is needed to compare the properties of the cell-derived exosomes according to the cell culture media. METHODS To compare the cell culture condition on exosomes, human umbilical cord mesenchymal stem cells (UCMSCs) were cultured with two different media, serum containing media, 10% FBS supplemented DMEM (NM) and serum-free chemically defined media, CellCor™ CD MSC (CDM). To remove FBS-derived exosomes from UCMSC cultured with NM, the medium was replaced with FBS-free DMEM for starvation during exosome isolation. The production yield and expression levels of angiogenic and pro-inflammatory factors were compared. And, the subpopulations of exosome were classified depending on the surface properties and loaded cytokines. Finally, the wound healing and angiogenic effects have been evaluated using in vitro assays. RESULTS The UCMSC-derived exosomes under two different cell culture media could be classified into subpopulations according to the surface composition and loaded cytokines. Especially, exosome derived from UCMSC cultured with CDM showed higher expression levels of cytokines related to regenerative bioactivities which resulted in enhanced wound healing and angiogenesis. CONCLUSION CDM has the advantages to maintain cell proliferation even during the period of exosome isolations and eliminate unknown side effects caused by serum-derived exosomes. Additionally, exosomes derived from UCMSC cultured with CDM show better wound healing and angiogenic effects due to a lot of regeneration-related cytokines and less pro-inflammatory cytokines compared to with NM.
Collapse
Affiliation(s)
- Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.,Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.,ntelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyo Jeong Seo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Joo Youn Lee
- Xcell Therapeutics, Hanhwa Biz metro Building, 242 Digital-ro, Guro-gu, Seoul, 08394, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.,ntelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
26
|
A Paradigm Shift in Tissue Engineering: From a Top–Down to a Bottom–Up Strategy. Processes (Basel) 2021. [DOI: 10.3390/pr9060935] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Tissue engineering (TE) was initially designed to tackle clinical organ shortage problems. Although some engineered tissues have been successfully used for non-clinical applications, very few (e.g., reconstructed human skin) have been used for clinical purposes. As the current TE approach has not achieved much success regarding more broad and general clinical applications, organ shortage still remains a challenging issue. This very limited clinical application of TE can be attributed to the constraints in manufacturing fully functional tissues via the traditional top–down approach, where very limited cell types are seeded and cultured in scaffolds with equivalent sizes and morphologies as the target tissues. The newly proposed developmental engineering (DE) strategy towards the manufacture of fully functional tissues utilises a bottom–up approach to mimic developmental biology processes by implementing gradual tissue assembly alongside the growth of multiple cell types in modular scaffolds. This approach may overcome the constraints of the traditional top–down strategy as it can imitate in vivo-like tissue development processes. However, several essential issues must be considered, and more mechanistic insights of the fundamental, underpinning biological processes, such as cell–cell and cell–material interactions, are necessary. The aim of this review is to firstly introduce and compare the number of cell types, the size and morphology of the scaffolds, and the generic tissue reconstruction procedures utilised in the top–down and the bottom–up strategies; then, it will analyse their advantages, disadvantages, and challenges; and finally, it will briefly discuss the possible technologies that may overcome some of the inherent limitations of the bottom–up strategy.
Collapse
|
27
|
Muoio F, Panella S, Jossen V, Lindner M, Harder Y, Müller M, Eibl R, Tallone T. Human Adipose Stem Cells (hASCs) Grown on Biodegradable Microcarriers in Serum- and Xeno-Free Medium Preserve Their Undifferentiated Status. J Funct Biomater 2021; 12:jfb12020025. [PMID: 33923488 PMCID: PMC8167760 DOI: 10.3390/jfb12020025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Human adipose stem cells (hASCs) are promising candidates for cell-based therapies, but they need to be efficiently expanded in vitro as they cannot be harvested in sufficient quantities. Recently, dynamic bioreactor systems operated with microcarriers achieved considerable high cell densities. Thus, they are a viable alternative to static planar cultivation systems to obtain high numbers of clinical-grade hASCs. Nevertheless, the production of considerable biomass in a short time must not be achieved to the detriment of the cells' quality. To facilitate the scalable expansion of hASC, we have developed a new serum- and xeno-free medium (UrSuppe) and a biodegradable microcarrier (BR44). In this study, we investigated whether the culture of hASCs in defined serum-free conditions on microcarriers (3D) or on planar (2D) cell culture vessels may influence the expression of some marker genes linked with the immature degree or the differentiated status of the cells. Furthermore, we investigated whether the biomaterials, which form our biodegradable MCs, may affect cell behavior and differentiation. The results confirmed that the quality and the undifferentiated status of the hASCs are very well preserved when they grow on BR44 MCs in defined serum-free conditions. Indeed, the ASCs showed a gene expression profile more compatible with an undifferentiated status than the same cells grown under standard planar conditions.
Collapse
Affiliation(s)
- Francesco Muoio
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
| | - Stefano Panella
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
| | - Valentin Jossen
- Institute of Chemistry & Biotechnology, Competence Center of Biochemical Engineering & Cell Cultivation Technique Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland; (V.J.); (R.E.)
| | | | - Yves Harder
- Department of Plastic, Reconstructive and Aesthetic Surgery, EOC, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | | | - Regine Eibl
- Institute of Chemistry & Biotechnology, Competence Center of Biochemical Engineering & Cell Cultivation Technique Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland; (V.J.); (R.E.)
| | - Tiziano Tallone
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
- Correspondence: ; Tel.: +41-91-805-38-85
| |
Collapse
|
28
|
Fernández-Francos S, Eiro N, Costa LA, Escudero-Cernuda S, Fernández-Sánchez ML, Vizoso FJ. Mesenchymal Stem Cells as a Cornerstone in a Galaxy of Intercellular Signals: Basis for a New Era of Medicine. Int J Mol Sci 2021; 22:ijms22073576. [PMID: 33808241 PMCID: PMC8036553 DOI: 10.3390/ijms22073576] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Around 40% of the population will suffer at some point in their life a disease involving tissue loss or an inflammatory or autoimmune process that cannot be satisfactorily controlled with current therapies. An alternative for these processes is represented by stem cells and, especially, mesenchymal stem cells (MSC). Numerous preclinical studies have shown MSC to have therapeutic effects in different clinical conditions, probably due to their mesodermal origin. Thereby, MSC appear to play a central role in the control of a galaxy of intercellular signals of anti-inflammatory, regenerative, angiogenic, anti-fibrotic, anti-oxidative stress effects of anti-apoptotic, anti-tumor, or anti-microbial type. This concept forces us to return to the origin of natural physiological processes as a starting point to understand the evolution of MSC therapy in the field of regenerative medicine. These biological effects, demonstrated in countless preclinical studies, justify their first clinical applications, and draw a horizon of new therapeutic strategies. However, several limitations of MSC as cell therapy are recognized, such as safety issues, handling difficulties for therapeutic purposes, and high economic cost. For these reasons, there is an ongoing tendency to consider the use of MSC-derived secretome products as a therapeutic tool, since they reproduce the effects of their parent cells. However, it will be necessary to resolve key aspects, such as the choice of the ideal type of MSC according to their origin for each therapeutic indication and the implementation of new standardized production strategies. Therefore, stem cell science based on an intelligently designed production of MSC and or their derivative products will be able to advance towards an innovative and more personalized medical biotechnology.
Collapse
Affiliation(s)
| | - Noemi Eiro
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.F.-F.); (L.A.C.)
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-985320050 (ext. 84216)
| | - Luis A. Costa
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.F.-F.); (L.A.C.)
| | - Sara Escudero-Cernuda
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, 33006 Oviedo, Spain; (S.E.-C.); (M.L.F.-S.)
| | - María Luisa Fernández-Sánchez
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, 33006 Oviedo, Spain; (S.E.-C.); (M.L.F.-S.)
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.F.-F.); (L.A.C.)
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-985320050 (ext. 84216)
| |
Collapse
|
29
|
Eibl R, Senn Y, Gubser G, Jossen V, van den Bos C, Eibl D. Cellular Agriculture: Opportunities and Challenges. Annu Rev Food Sci Technol 2021; 12:51-73. [PMID: 33770467 DOI: 10.1146/annurev-food-063020-123940] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cellular agriculture is the controlled and sustainable manufacture of agricultural products with cells and tissues without plant or animal involvement. Today, microorganisms cultivated in bioreactors already produce egg and milk proteins, sweeteners, and flavors for human nutrition as well as leather and fibers for shoes, bags, and textiles. Furthermore, plant cell and tissue cultures provide ingredients that stimulate the immune system and improve skin texture, with another precommercial cellular agriculture product, in vitro meat, currently receiving a great deal of attention. All these approaches could assist traditional agriculture in continuing to provide for the dietary requirements of a growing world population while freeing up important resources such as arable land. Despite early successes, challenges remain and are discussed in this review, with a focus on production processes involving plant and animal cell and tissue cultures.
Collapse
Affiliation(s)
- Regine Eibl
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | - Yannick Senn
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | - Géraldine Gubser
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | - Valentin Jossen
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| | | | - Dieter Eibl
- Institute of Chemistry and Biotechnology, Department of Life Sciences and Facility Management, Zurich University of Applied Sciences, Wädenswil 8820, Switzerland;
| |
Collapse
|
30
|
Development of a Biodegradable Microcarrier for the Cultivation of Human Adipose Stem Cells (hASCs) with a Defined Xeno- and Serum-Free Medium. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11030925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stirred single-use bioreactors in combination with microcarriers (MCs) have established themselves as a technology that has the potential to meet the demands of current and future cell therapeutic markets. However, most of the published processes have been performed using fetal bovine serum (FBS) containing cell culture medium and non-biocompatible MCs. This approach has two significant drawbacks: firstly, the inevitable potential risks associated with the use of FBS for clinical applications; secondly, non-biocompatible MCs have to be removed from the cell suspension before implantation, requiring a step that causes loss of viable cells and adds further costs and complications. This study aimed to develop a new platform based on a chemically defined xeno- and serum-free cell culture medium and biodegradable MC that can support the growth of human adipose stem cells (hASCs) while still preserving their undifferentiated status. A specific combination of components and manufacturing parameters resulted in a MC prototype, called “BR44”, which delivered the desired functionality. MC BR44 allows the hASCs to stick to its surface and grow in a chemically defined xeno- and serum-free medium (UrSuppe). Although the cells’ expansion rate was not as high as with a commercial non-biodegradable standard MC, those cultured on BR44 maintained a better undifferentiated status in both static and dynamic conditions than those cultured on traditional 2D surfaces.
Collapse
|
31
|
Silva Couto P, Rotondi M, Bersenev A, Hewitt C, Nienow A, Verter F, Rafiq Q. Expansion of human mesenchymal stem/stromal cells (hMSCs) in bioreactors using microcarriers: lessons learnt and what the future holds. Biotechnol Adv 2020; 45:107636. [DOI: 10.1016/j.biotechadv.2020.107636] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/01/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
|
32
|
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: 0.6] [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
|
33
|
Jossen V, Muoio F, Panella S, Harder Y, Tallone T, Eibl R. An Approach towards a GMP Compliant In-Vitro Expansion of Human Adipose Stem Cells for Autologous Therapies. Bioengineering (Basel) 2020; 7:bioengineering7030077. [PMID: 32698363 PMCID: PMC7552624 DOI: 10.3390/bioengineering7030077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Human Adipose Tissue Stem Cells (hASCs) are a valuable source of cells for clinical applications (e.g., treatment of acute myocardial infarction and inflammatory diseases), especially in the field of regenerative medicine. However, for autologous (patient-specific) and allogeneic (off-the-shelf) hASC-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 hASCs for autologous therapies is more problematic because the cell material changes for each treatment. Moreover, cell material is normally isolated from non-healthy or older patients, which further complicates successful in-vitro expansion. Hence, the goal of this study was to perform cell expansion studies with hASCs isolated from two different patients/donors (i.e., different ages and health statuses) under xeno- and serum-free conditions in static, planar (2D) and dynamically mixed (3D) cultivation systems. Our primary aim was I) to compare donor variability under in-vitro conditions and II) to develop and establish an unstructured, segregated growth model as a proof-of-concept study. Maximum cell densities of between 0.49 and 0.65 × 105 hASCs/cm2 were achieved for both donors in 2D and 3D cultivation systems. Cell growth under static and dynamically mixed conditions was comparable, which demonstrated that hydrodynamic stresses (P/V = 0.63 W/m3, τnt = 4.96 × 10−3 Pa) acting at Ns1u (49 rpm for 10 g/L) did not negatively affect cell growth, even under serum-free conditions. However, donor-dependent differences in the cell size were found, which resulted in significantly different maximum cell densities for each of the two donors. In both cases, stemness was well maintained under static 2D and dynamic 3D conditions, as long as the cells were not hyperconfluent. The optimal point for cell harvesting was identified as between cell densities of 0.41 and 0.56 × 105 hASCs/cm2 (end of exponential growth phase). The growth model delivered reliable predictions for cell growth, substrate consumption and metabolite production in both types of cultivation systems. Therefore, the model can be used as a basis for future investigations in order to develop a robust MC-based hASC production process for autologous therapies.
Collapse
Affiliation(s)
- Valentin Jossen
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
- Correspondence: or ; Tel.: +41-58-934-5334
| | - Francesco Muoio
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.); (T.T.)
| | - Stefano Panella
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.); (T.T.)
| | - Yves Harder
- Department of Plastic, Reconstructive and Aesthetic Surgery, Ente Ospedaliero Cantonale (EOC), 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Tiziano Tallone
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.); (T.T.)
| | - Regine Eibl
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
| |
Collapse
|
34
|
Lembong J, Kirian R, Takacs JD, Olsen TR, Lock LT, Rowley JA, Ahsan T. Bioreactor Parameters for Microcarrier-Based Human MSC Expansion under Xeno-Free Conditions in a Vertical-Wheel System. Bioengineering (Basel) 2020; 7:E73. [PMID: 32650422 PMCID: PMC7552727 DOI: 10.3390/bioengineering7030073] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 12/27/2022] Open
Abstract
Human mesenchymal stem/stromal cells (hMSCs) have been investigated and proven to be a well-tolerated, safe therapy for a variety of indications, as shown by over 900 registered hMSC-based clinical trials. To meet the commercial demand for clinical manufacturing of hMSCs, production requires a scale that can achieve a lot size of ~100B cells, which requires innovative manufacturing technologies such as 3D bioreactors. A robust suspension bioreactor process that can be scaled-up to the relevant scale is therefore crucial. In this study, we developed a fed-batch, microcarrier-based bioreactor process, which enhances media productivity and drives a cost-effective and less labor-intensive hMSC expansion process. We determined parameter settings for various stages of the culture: inoculation, bioreactor culture, and harvest. Addition of a bioreactor feed, using a fed-batch approach, was necessary to replenish the mitogenic factors that were depleted from the media within the first 3 days of culture. Our study resulted in an optimized hMSC culture protocol that consistently achieved hMSC densities between 2 × 105-6 × 105 cells/mL within 5 days with no media exchange, maintaining the final cell population doubling level (PDL) at 16-20. Using multiple hMSC donors, we showed that this process was robust and yielded hMSCs that maintained expansion, phenotypic characteristic, and functional properties. The developed process in a vertical-wheel suspension bioreactor can be scaled to the levels needed to meet commercial demand of hMSCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tabassum Ahsan
- RoosterBio, Inc., 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA; (J.L.); (R.K.); (J.D.T.); (T.R.O.); (L.T.L.); (J.A.R.)
| |
Collapse
|
35
|
García-Fernández C, López-Fernández A, Borrós S, Lecina M, Vives J. Strategies for large-scale expansion of clinical-grade human multipotent mesenchymal stromal cells. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107601] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Wyrobnik TA, Ducci A, Micheletti M. Advances in human mesenchymal stromal cell-based therapies - Towards an integrated biological and engineering approach. Stem Cell Res 2020; 47:101888. [PMID: 32688331 DOI: 10.1016/j.scr.2020.101888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Recent advances of stem cell-based therapies in clinical trials have raised the need for large-scale manufacturing platforms that can supply clinically relevant doses to meet an increasing demand. Promising results have been reported using stirred-tank bioreactors, where human Mesenchymal Stromal Cells (hMSCs) were cultured in suspension on microcarriers (MCs), although the formation of microcarrier-cell-aggregates might still limit mass transfer and determine a heterogeneous distribution of hMSCs. A variety of MCs, bioreactor-impeller configurations, and agitation conditions have been established in an attempt to overcome the trade-off of ensuring good suspension while keeping the stresses to a minimum. While understanding and controlling the fluid flow environment of bioreactors has been initially under-appreciated, it has recently gained in popularity in the mission of providing ideal culture environments across different scales. This review article aims to provide a comprehensive overview of how rigorous engineering characterisation studies improved the outcome of biological process development and scale-up efforts. Reconciling these two disciplines is crucial to propose tailored bioprocessing solutions that can provide improved growth environments across a range of scales for the allogeneic cell therapies of the future.
Collapse
Affiliation(s)
- Tom A Wyrobnik
- Department of Biochemical Engineering, UCL, Gower Street, London WC1E 6BT, UK
| | - Andrea Ducci
- Department of Mechanical Engineering, UCL, Torrington Place, London WC1E 7JE, UK
| | - Martina Micheletti
- Department of Biochemical Engineering, UCL, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
37
|
Tsai AC, Jeske R, Chen X, Yuan X, Li Y. Influence of Microenvironment on Mesenchymal Stem Cell Therapeutic Potency: From Planar Culture to Microcarriers. Front Bioeng Biotechnol 2020; 8:640. [PMID: 32671039 PMCID: PMC7327111 DOI: 10.3389/fbioe.2020.00640] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are a promising candidate in cell therapy as they exhibit multilineage differentiation, homing to the site of injury, and secretion of trophic factors that facilitate tissue healing and/or modulate immune response. As a result, hMSC-derived products have attracted growing interests in preclinical and clinical studies. The development of hMSC culture platforms for large-scale biomanufacturing is necessary to meet the requirements for late-phase clinical trials and future commercialization. Microcarriers in stirred-tank bioreactors have been widely utilized in large-scale expansion of hMSCs for translational applications because of a high surface-to-volume ratio compared to conventional 2D planar culture. However, recent studies have demonstrated that microcarrier-expanded hMSCs differ from dish- or flask-expanded cells in size, morphology, proliferation, viability, surface markers, gene expression, differentiation potential, and secretome profile which may lead to altered therapeutic potency. Therefore, understanding the bioprocessing parameters that influence hMSC therapeutic efficacy is essential for the optimization of microcarrier-based bioreactor system to maximize hMSC quantity without sacrificing quality. In this review, biomanufacturing parameters encountered in planar culture and microcarrier-based bioreactor culture of hMSCs are compared and discussed with specific focus on cell-adhesion surface (e.g., discontinuous surface, underlying curvature, microcarrier stiffness, porosity, surface roughness, coating, and charge) and the dynamic microenvironment in bioreactor culture (e.g., oxygen and nutrients, shear stress, particle collision, and aggregation). The influence of dynamic culture in bioreactors on hMSC properties is also reviewed in order to establish connection between bioprocessing and stem cell function. This review addresses fundamental principles and concepts for future design of biomanufacturing systems for hMSC-based therapy.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
38
|
Abstract
Mesenchymal stromal cells (MSCs) are among of the most studied cell type for cellular therapy thanks to the ease of isolation, cultivation, and the high
ex vivo expansion potential. In 2018, the European Medicines Agency finally granted the first marketing authorization for an MSC product. Despite the numerous promising results in preclinical studies, translation into routine practice still lags behind: therapeutic benefits of MSCs are not as satisfactory in clinical trial settings as they appear to be in preclinical models. The bench-to-bedside-and-back approach and careful evaluation of discrepancies between preclinical and clinical results have provided valuable insights into critical components of MSC manufacturing, their mechanisms of action, and how to evaluate and quality-control them. We sum up these past developments in the introductory section (“Mesenchymal stromal cells: name follows function”). From the huge amount of information, we then selected a few examples to illustrate challenges and opportunities to improve MSCs for clinical purposes. These include tissue origin of MSCs, MSC culture conditions, immune compatibility, and route of application and dosing. Finally, we add some information on MSC mechanisms of action and translation into potency assays and give an outlook on future perspectives raising the question of whether the future clinical product may be cell-based or cell-derived.
Collapse
Affiliation(s)
- Erika Rendra
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Eleonora Scaccia
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,FlowCore Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,German Red Cross Blood Donor Service Baden-Württemberg - Hessen, Mannheim, 68167, Germany
| |
Collapse
|
39
|
Bodiou V, Moutsatsou P, Post MJ. Microcarriers for Upscaling Cultured Meat Production. Front Nutr 2020; 7:10. [PMID: 32154261 PMCID: PMC7045063 DOI: 10.3389/fnut.2020.00010] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/28/2020] [Indexed: 12/19/2022] Open
Abstract
Due to the considerable environmental impact and the controversial animal welfare associated with industrial meat production, combined with the ever-increasing global population and demand for meat products, sustainable production alternatives are indispensable. In 2013, the world's first laboratory grown hamburger made from cultured muscle cells was developed. However, coming at a price of $300.000, and being produced manually, substantial effort is still required to reach sustainable large-scale production. One of the main challenges is scalability. Microcarriers (MCs), offering a large surface/volume ratio, are the most promising candidates for upscaling muscle cell culture. However, although many MCs have been developed for cell lines and stem cells typically used in the medical field, none have been specifically developed for muscle stem cells and meat production. This paper aims to discuss the MCs' design criteria for skeletal muscle cell proliferation and subsequently for meat production based on three scenarios: (1) MCs are serving only as a temporary substrate for cell attachment and proliferation and therefore they need to be separated from the cells at some stage of the bioprocess, (2) MCs serve as a temporary substrate for cell proliferation but are degraded or dissolved during the bioprocess, and (3) MCs are embedded in the final product and therefore need to be edible. The particularities of each of these three bioprocesses will be discussed from the perspective of MCs as well as the feasibility of a one-step bioprocess. Each scenario presents advantages and drawbacks, which are discussed in detail, nevertheless the third scenario appears to be the most promising one for a production process. Indeed, using an edible material can limit or completely eliminate dissociation/degradation/separation steps and even promote organoleptic qualities when embedded in the final product. Edible microcarriers could also be used as a temporary substrate similarly to scenarios 1 and 2, which would limit the risk of non-edible residues.
Collapse
Affiliation(s)
- Vincent Bodiou
- Department of Physiology, Faculty of Health, Medicine and Life Sciences, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Mosa Meat BV, Maastricht, Netherlands
- CARIM, Faculty of Health, Medicine and Life Sciences, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Panagiota Moutsatsou
- Department of Physiology, Faculty of Health, Medicine and Life Sciences, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Mosa Meat BV, Maastricht, Netherlands
| | - Mark J. Post
- Department of Physiology, Faculty of Health, Medicine and Life Sciences, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Mosa Meat BV, Maastricht, Netherlands
- CARIM, Faculty of Health, Medicine and Life Sciences, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
40
|
Piñeiro-Ramil M, Sanjurjo-Rodríguez C, Castro-Viñuelas R, Rodríguez-Fernández S, Fuentes-Boquete I, Blanco F, Díaz-Prado S. Usefulness of Mesenchymal Cell Lines for Bone and Cartilage Regeneration Research. Int J Mol Sci 2019; 20:E6286. [PMID: 31847077 PMCID: PMC6940884 DOI: 10.3390/ijms20246286] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/18/2022] Open
Abstract
The unavailability of sufficient numbers of human primary cells is a major roadblock for in vitro repair of bone and/or cartilage, and for performing disease modelling experiments. Immortalized mesenchymal stromal cells (iMSCs) may be employed as a research tool for avoiding these problems. The purpose of this review was to revise the available literature on the characteristics of the iMSC lines, paying special attention to the maintenance of the phenotype of the primary cells from which they were derived, and whether they are effectively useful for in vitro disease modeling and cell therapy purposes. This review was performed by searching on Web of Science, Scopus, and PubMed databases from 1 January 2015 to 30 September 2019. The keywords used were ALL = (mesenchymal AND ("cell line" OR immortal*) AND (cartilage OR chondrogenesis OR bone OR osteogenesis) AND human). Only original research studies in which a human iMSC line was employed for osteogenesis or chondrogenesis experiments were included. After describing the success of the immortalization protocol, we focused on the iMSCs maintenance of the parental phenotype and multipotency. According to the literature revised, it seems that the maintenance of these characteristics is not guaranteed by immortalization, and that careful selection and validation of clones with particular characteristics is necessary for taking advantage of the full potential of iMSC to be employed in bone and cartilage-related research.
Collapse
Affiliation(s)
- M. Piñeiro-Ramil
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
| | - C. Sanjurjo-Rodríguez
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - R. Castro-Viñuelas
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
| | - S. Rodríguez-Fernández
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
| | - I.M. Fuentes-Boquete
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - F.J. Blanco
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - S.M. Díaz-Prado
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| |
Collapse
|
41
|
Gupta P, Hall GN, Geris L, Luyten FP, Papantoniou I. Human Platelet Lysate Improves Bone Forming Potential of Human Progenitor Cells Expanded in Microcarrier-Based Dynamic Culture. Stem Cells Transl Med 2019; 8:810-821. [PMID: 31038850 PMCID: PMC6646698 DOI: 10.1002/sctm.18-0216] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/19/2019] [Indexed: 12/22/2022] Open
Abstract
Xenogeneic‐free media are required for translating advanced therapeutic medicinal products to the clinics. In addition, process efficiency is crucial for ensuring cost efficiency, especially when considering large‐scale production of mesenchymal stem cells (MSCs). Human platelet lysate (HPL) has been increasingly adopted as an alternative for fetal bovine serum (FBS) for MSCs. However, its therapeutic and regenerative potential in vivo is largely unexplored. Herein, we compare the effects of FBS and HPL supplementation for a scalable, microcarrier‐based dynamic expansion of human periosteum‐derived cells (hPDCs) while assessing their bone forming capacity by subcutaneous implantation in small animal model. We observed that HPL resulted in faster cell proliferation with a total fold increase of 5.2 ± 0.61 in comparison to 2.7 ± 02.22‐fold in FBS. Cell viability and trilineage differentiation capability were maintained by HPL, although a suppression of adipogenic differentiation potential was observed. Differences in mRNA expression profiles were also observed between the two on several markers. When implanted, we observed a significant difference between the bone forming capacity of cells expanded in FBS and HPL, with HPL supplementation resulting in almost three times more mineralized tissue within calcium phosphate scaffolds. FBS‐expanded cells resulted in a fibrous tissue structure, whereas HPL resulted in mineralized tissue formation, which can be classified as newly formed bone, verified by μCT and histological analysis. We also observed the presence of blood vessels in our explants. In conclusion, we suggest that replacing FBS with HPL in bioreactor‐based expansion of hPDCs is an optimal solution that increases expansion efficiency along with promoting bone forming capacity of these cells. stem cells translational medicine2019;8:810&821
Collapse
Affiliation(s)
- Priyanka Gupta
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Gabriella Nilsson Hall
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA-R In Silico Medicine, Université de Liege, Liège, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Robb KP, Fitzgerald JC, Barry F, Viswanathan S. Mesenchymal stromal cell therapy: progress in manufacturing and assessments of potency. Cytotherapy 2018; 21:289-306. [PMID: 30528726 DOI: 10.1016/j.jcyt.2018.10.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cell (MSC) therapies have been pursued for a broad spectrum of indications but mixed reports on clinical efficacy have given rise to some degree of skepticism regarding the effectiveness of this approach. However, recent reports of successful clinical outcomes and regulatory approvals for graft-versus-host disease, Crohn's disease and critical limb ischemia have prompted a shift in this perspective. With hundreds of clinical trials involving MSCs currently underway and an increasing demand for large-scale manufacturing protocols, there is a critical need to develop standards that can be applied to processing methods and to establish consensus assays for both MSC processing control and MSC product release. Reference materials and validated, uniformly applied tests for quality control of MSC products are needed. Here, we review recent developments in MSC manufacturing technologies, release testing and potency assays. We conclude that, although MSCs hold considerable promise clinically, economies of scale have yet to be achieved although numerous bioreactor technologies for scalable production of MSCs exist. Additionally, rigorous disease-specific product testing and comprehensive understanding of mechanisms of action, which are linked to relevant process and product release potency assays, will be required to ensure that these therapies continue to be successful.
Collapse
Affiliation(s)
- Kevin P Robb
- The Arthritis Program, University Health Network, Toronto, Canada;; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Joan C Fitzgerald
- Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Frank Barry
- The Arthritis Program, University Health Network, Toronto, Canada;; Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Sowmya Viswanathan
- The Arthritis Program, University Health Network, Toronto, Canada;; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Cell Therapy Program, University Health Network, Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
43
|
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: 1.4] [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
|
44
|
McNeill EP, Reese RW, Tondon A, Clough BH, Pan S, Froese J, Palmer D, Krause U, Loeb DM, Kaunas R, Gregory CA. Three-dimensional in vitro modeling of malignant bone disease recapitulates experimentally accessible mechanisms of osteoinhibition. Cell Death Dis 2018; 9:1161. [PMID: 30478297 PMCID: PMC6255770 DOI: 10.1038/s41419-018-1203-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022]
Abstract
Malignant bone disease (MBD) occurs when tumors establish in bone, causing catastrophic tissue damage as a result of accelerated bone destruction and inhibition of repair. The resultant so-called osteolytic lesions (OL) take the form of tumor-filled cavities in bone that cause pain, fractures, and associated morbidity. Furthermore, the OL microenvironment can support survival of tumor cells and resistance to chemotherapy. Therefore, a deeper understanding of OL formation and MBD progression is imperative for the development of future therapeutic strategies. Herein, we describe a novel in vitro platform to study bone-tumor interactions based on three-dimensional co-culture of osteogenically enhanced human mesenchymal stem cells (OEhMSCs) in a rotating wall vessel bioreactor (RWV) while attached to micro-carrier beads coated with extracellular matrix (ECM) composed of factors found in anabolic bone tissue. Osteoinhibition was recapitulated in this model by co-culturing the OEhMSCs with a bone-tumor cell line (MOSJ-Dkk1) that secretes the canonical Wnt (cWnt) inhibitor Dkk-1, a tumor-borne osteoinhibitory factor widely associated with several forms of MBD, or intact tumor fragments from Dkk-1 positive patient-derived xenografts (PDX). Using the model, we observed that depending on the conditions of growth, tumor cells can biochemically inhibit osteogenesis by disrupting cWnt activity in OEhMSCs, while simultaneously co-engrafting with OEhMSCs, displacing them from the niche, perturbing their activity, and promoting cell death. In the absence of detectable co-engraftment with OEhMSCs, Dkk-1 positive PDX fragments had the capacity to enhance OEhMSC proliferation while inhibiting their osteogenic differentiation. The model described has the capacity to provide new and quantifiable insights into the multiple pathological mechanisms of MBD that are not readily measured using monolayer culture or animal models.
Collapse
Affiliation(s)
- Eoin P McNeill
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, TX, 77845, USA
| | - Robert W Reese
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Abishek Tondon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Bret H Clough
- Department of Medical Physiology, Texas A&M Health Science Center, Temple, TX, 76501, USA
| | - Simin Pan
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, TX, 77845, USA
| | - Jeremiah Froese
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, TX, 77845, USA
| | - Daniel Palmer
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, TX, 77845, USA
| | - Ulf Krause
- Institute for Transfusion Medicine and Transplant Immunology, University Hospital Muenster, Muenster, Germany
| | - David M Loeb
- Departments of Pediatrics and Developmental and Molecular Biology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, 3411 Wayne Avenue, Bronx, NY, 10467, USA
| | - Roland Kaunas
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA.
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, TX, 77845, USA.
| |
Collapse
|
45
|
Lerman MJ, Lembong J, Muramoto S, Gillen G, Fisher JP. The Evolution of Polystyrene as a Cell Culture Material. TISSUE ENGINEERING. PART B, REVIEWS 2018; 24:359-372. [PMID: 29631491 PMCID: PMC6199621 DOI: 10.1089/ten.teb.2018.0056] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/21/2018] [Indexed: 01/19/2023]
Abstract
Polystyrene (PS) has brought in vitro cell culture from its humble beginnings to the modern era, propelling dozens of research fields along the way. This review discusses the development of the material, fabrication, and treatment approaches to create the culture material. However, native PS surfaces poorly facilitate cell adhesion and growth in vitro. To overcome this, liquid surface deposition, energetic plasma activation, and emerging functionalization methods transform the surface chemistry. This review seeks to highlight the many potential applications of the first widely accepted polymer growth surface. Although the majority of in vitro research occurs on two-dimensional surfaces, the importance of three-dimensional (3D) culture models cannot be overlooked. The methods to transition PS to specialized 3D culture surfaces are also reviewed. Specifically, casting, electrospinning, 3D printing, and microcarrier approaches to shift PS to a 3D culture surface are highlighted. The breadth of applications of the material makes it impossible to highlight every use, but the aim remains to demonstrate the versatility and potential as both a general and custom cell culture surface. The review concludes with emerging scaffolding approaches and, based on the findings, presents our insights on the future steps for PS as a tissue culture platform.
Collapse
Affiliation(s)
- Max J. Lerman
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
- NIH/NIBIB Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Josephine Lembong
- NIH/NIBIB Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Shin Muramoto
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Greg Gillen
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - John P. Fisher
- NIH/NIBIB Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
46
|
Rafiq QA, Ruck S, Hanga MP, Heathman TR, Coopman K, Nienow AW, Williams DJ, Hewitt CJ. Qualitative and quantitative demonstration of bead-to-bead transfer with bone marrow-derived human mesenchymal stem cells on microcarriers: Utilising the phenomenon to improve culture performance. Biochem Eng J 2018. [DOI: 10.1016/j.bej.2017.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
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: 89] [Impact Index Per Article: 12.7] [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
|
48
|
Jossen V, van den Bos C, Eibl R, Eibl D. Manufacturing human mesenchymal stem cells at clinical scale: process and regulatory challenges. Appl Microbiol Biotechnol 2018; 102:3981-3994. [PMID: 29564526 PMCID: PMC5895685 DOI: 10.1007/s00253-018-8912-x] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/28/2018] [Accepted: 03/02/2018] [Indexed: 01/10/2023]
Abstract
Human mesenchymal stem cell (hMSC)-based therapies are of increasing interest in the field of regenerative medicine. As economic considerations have shown, allogeneic therapy seems to be the most cost-effective method. Standardized procedures based on instrumented single-use bioreactors have been shown to provide billion of cells with consistent product quality and to be superior to traditional expansions in planar cultivation systems. Furthermore, under consideration of the complex nature and requirements of allogeneic hMSC-therapeutics, a new equipment for downstream processing (DSP) was successfully evaluated. This mini-review summarizes both the current state of the hMSC production process and the challenges which have to be taken into account when efficiently producing hMSCs for the clinical scale. Special emphasis is placed on the upstream processing (USP) and DSP operations which cover expansion, harvesting, detachment, separation, washing and concentration steps, and the regulatory demands.
Collapse
Affiliation(s)
- Valentin Jossen
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820, Wädenswil, Switzerland.
| | | | - Regine Eibl
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820, Wädenswil, Switzerland
| | - Dieter Eibl
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820, Wädenswil, Switzerland
| |
Collapse
|
49
|
Petry F, Weidner T, Czermak P, Salzig D. Three-Dimensional Bioreactor Technologies for the Cocultivation of Human Mesenchymal Stem/Stromal Cells and Beta Cells. Stem Cells Int 2018; 2018:2547098. [PMID: 29731775 PMCID: PMC5872596 DOI: 10.1155/2018/2547098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a prominent health problem caused by the failure of pancreatic beta cells. One therapeutic approach is the transplantation of functional beta cells, but it is difficult to generate sufficient beta cells in vitro and to ensure these cells remain viable at the transplantation site. Beta cells suffer from hypoxia, undergo apoptosis, or are attacked by the host immune system. Human mesenchymal stem/stromal cells (hMSCs) can improve the functionality and survival of beta cells in vivo and in vitro due to direct cell contact or the secretion of trophic factors. Current cocultivation concepts with beta cells are simple and cannot exploit the favorable properties of hMSCs. Beta cells need a three-dimensional (3D) environment to function correctly, and the cocultivation setup is therefore more complex. This review discusses 3D cultivation forms (aggregates, capsules, and carriers) for hMSCs and beta cells and strategies for large-scale cultivation. We have determined process parameters that must be balanced and considered for the cocultivation of hMSCs and beta cells, and we present several bioreactor setups that are suitable for such an innovative cocultivation approach. Bioprocess engineering of the cocultivation processes is necessary to achieve successful beta cell therapy.
Collapse
Affiliation(s)
- Florian Petry
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| | - Tobias Weidner
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
- Department of Chemical Engineering, Kansas State University, Manhattan, KS, USA
- Project Group Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Winchesterstr. 3, 35394 Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| |
Collapse
|
50
|
Foreword Advances in “Stem Cell Bioengineering”. Process Biochem 2017. [DOI: 10.1016/j.procbio.2017.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|