1
|
Jeannerat A, Peneveyre C, Jaccoud S, Philippe V, Scaletta C, Hirt-Burri N, Abdel-Sayed P, Martin R, Applegate LA, Pioletti DP, Laurent A. Banked Primary Progenitor Cells for Allogeneic Intervertebral Disc (IVD) Therapy: Preclinical Qualification and Functional Optimization within a Cell Spheroid Formulation Process. Pharmaceutics 2024; 16:1274. [PMID: 39458605 PMCID: PMC11510186 DOI: 10.3390/pharmaceutics16101274] [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: 09/03/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Biological products are emerging as therapeutic management options for intervertebral disc (IVD) degenerative affections and lower back pain. Autologous and allogeneic cell therapy protocols have been clinically implemented for IVD repair. Therein, several manufacturing process design considerations were shown to significantly influence clinical outcomes. The primary objective of this study was to preclinically qualify (chondrogenic potential, safety, resistance to hypoxic and inflammatory stimuli) cryopreserved primary progenitor cells (clinical grade FE002-Disc cells) as a potential cell source in IVD repair/regeneration. The secondary objective of this study was to assess the cell source's delivery potential as cell spheroids (optimization of culture conditions, potential storage solutions). Methods/Results: Safety (soft agar transformation, β-galactosidase, telomerase activity) and functionality-related assays (hypoxic and inflammatory challenge) confirmed that the investigated cellular active substance was highly sustainable in defined cell banking workflows, despite possessing a finite in vitro lifespan. Functionality-related assays confirmed that the retained manufacturing process yielded strong collagen II and glycosaminoglycan (GAG) synthesis in the spheroids in 3-week chondrogenic induction. Then, the impacts of various process parameters (induction medium composition, hypoxic incubation, terminal spheroid lyophilization) were studied to gain insights on their criticality. Finally, an optimal set of technical specifications (use of 10 nM dexamethasone for chondrogenic induction, 2% O2 incubation of spheroids) was set forth, based on specific fine tuning of finished product critical functional attributes. Conclusions: Generally, this study qualified the considered FE002-Disc progenitor cell source for further preclinical investigation based on safety, quality, and functionality datasets. The novelty and significance of this study resided in the establishment of defined processes for preparing fresh, off-the-freezer, or off-the-shelf IVD spheroids using a preclinically qualified allogeneic human cell source. Overall, this study underscored the importance of using robust product components and optimal manufacturing process variants for maximization of finished cell-based formulation quality attributes.
Collapse
Affiliation(s)
- Annick Jeannerat
- Development Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| | - Cédric Peneveyre
- Development Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| | - Sandra Jaccoud
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
- Laboratory of Biomechanical Orthopedics, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Virginie Philippe
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
- Orthopedics and Traumatology Unit, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Corinne Scaletta
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
- STI School of Engineering, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Robin Martin
- Orthopedics and Traumatology Unit, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Dominique P. Pioletti
- Laboratory of Biomechanical Orthopedics, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Alexis Laurent
- Development Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (V.P.); (C.S.); (N.H.-B.); (P.A.-S.); (L.A.A.)
| |
Collapse
|
2
|
Ponta S, Bonato A, Neidenbach P, Bruhin VF, Laurent A, Applegate LA, Zenobi-Wong M, Barreto G. Streamlined, single-step non-viral CRISPR-Cas9 knockout strategy enhances gene editing efficiency in primary human chondrocyte populations. Arthritis Res Ther 2024; 26:66. [PMID: 38468277 PMCID: PMC10926593 DOI: 10.1186/s13075-024-03294-w] [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: 01/05/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND CRISPR-Cas9-based genome engineering represents a powerful therapeutic tool for cartilage tissue engineering and for understanding molecular pathways driving cartilage diseases. However, primary chondrocytes are difficult to transfect and rapidly dedifferentiate during monolayer (2D) cell culture, making the lengthy expansion of a single-cell-derived edited clonal population not feasible. For this reason, functional genetics studies focused on cartilage and rheumatic diseases have long been carried out in cellular models that poorly recapitulate the native molecular properties of human cartilaginous tissue (e.g., cell lines, induced pluripotent stem cells). Here, we set out to develop a non-viral CRISPR-Cas9, bulk-gene editing method suitable for chondrocyte populations from different cartilaginous sources. METHODS We screened electroporation and lipid nanoparticles for ribonucleoprotein (RNP) delivery in primary polydactyly chondrocytes, and optimized RNP reagents assembly. We knocked out RELA (also known as p65), a subunit of the nuclear factor kappa B (NF-κB), in polydactyly chondrocytes and further characterized knockout (KO) cells with RT-qPCR and Western Blot. We tested RELA KO in chondrocytes from diverse cartilaginous sources and characterized their phenotype with RT-qPCR. We examined the chondrogenic potential of wild-type (WT) and KO cell pellets in presence and absence of interleukin-1β (IL-1β). RESULTS We established electroporation as the optimal transfection technique for chondrocytes enhancing transfection and editing efficiency, while preserving high cell viability. We knocked out RELA with an unprecedented efficiency of ~90%, confirming lower inflammatory pathways activation upon IL-1β stimulation compared to unedited cells. Our protocol could be easily transferred to primary human chondrocytes harvested from osteoarthritis (OA) patients, human FE002 chondroprogenitor cells, bovine chondrocytes, and a human chondrocyte cell line, achieving comparable mean RELA KO editing levels using the same protocol. All KO pellets from primary human chondrocytes retained chondrogenic ability equivalent to WT cells, and additionally displayed enhanced matrix retention under inflamed conditions. CONCLUSIONS We showcased the applicability of our bulk gene editing method to develop effective autologous and allogeneic off-the-shelf gene therapies strategies and to enable functional genetics studies in human chondrocytes to unravel molecular mechanisms of cartilage diseases.
Collapse
Affiliation(s)
- Simone Ponta
- Department of Health Sciences and Technology, ETH Zürich, Zurich, 8093, Switzerland
| | - Angela Bonato
- Department of Health Sciences and Technology, ETH Zürich, Zurich, 8093, Switzerland
| | - Philipp Neidenbach
- Schulthess Clinic, Department of Lower Extremity Orthopaedics, Musculoskeletal Centre, Zurich, 8008, Switzerland
| | - Valentino F Bruhin
- Schulthess Clinic, Department of Lower Extremity Orthopaedics, Musculoskeletal Centre, Zurich, 8008, Switzerland
| | - Alexis Laurent
- Regenerative Therapy Unit, Plastic, Reconstructive & Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Epalinges, 1066, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive & Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Epalinges, 1066, Switzerland
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, ETH Zürich, Zurich, 8093, Switzerland
| | - Goncalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00014, Finland.
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, 02150, Finland.
- Orton Orthopedic Hospital, Tenholantie 10, Helsinki, 00280, Finland.
| |
Collapse
|
3
|
Philippe V, Jeannerat A, Peneveyre C, Jaccoud S, Scaletta C, Hirt-Burri N, Abdel-Sayed P, Raffoul W, Darwiche S, Applegate LA, Martin R, Laurent A. Autologous and Allogeneic Cytotherapies for Large Knee (Osteo)Chondral Defects: Manufacturing Process Benchmarking and Parallel Functional Qualification. Pharmaceutics 2023; 15:2333. [PMID: 37765301 PMCID: PMC10536774 DOI: 10.3390/pharmaceutics15092333] [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: 08/16/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Cytotherapies are often necessary for the management of symptomatic large knee (osteo)-chondral defects. While autologous chondrocyte implantation (ACI) has been clinically used for 30 years, allogeneic cells (clinical-grade FE002 primary chondroprogenitors) have been investigated in translational settings (Swiss progenitor cell transplantation program). The aim of this study was to comparatively assess autologous and allogeneic approaches (quality, safety, functional attributes) to cell-based knee chondrotherapies developed for clinical use. Protocol benchmarking from a manufacturing process and control viewpoint enabled us to highlight the respective advantages and risks. Safety data (telomerase and soft agarose colony formation assays, high passage cell senescence) and risk analyses were reported for the allogeneic FE002 cellular active substance in preparation for an autologous to allogeneic clinical protocol transposition. Validation results on autologous bioengineered grafts (autologous chondrocyte-bearing Chondro-Gide scaffolds) confirmed significant chondrogenic induction (COL2 and ACAN upregulation, extracellular matrix synthesis) after 2 weeks of co-culture. Allogeneic grafts (bearing FE002 primary chondroprogenitors) displayed comparable endpoint quality and functionality attributes. Parameters of translational relevance (transport medium, finished product suturability) were validated for the allogeneic protocol. Notably, the process-based benchmarking of both approaches highlighted the key advantages of allogeneic FE002 cell-bearing grafts (reduced cellular variability, enhanced process standardization, rationalized logistical and clinical pathways). Overall, this study built on our robust knowledge and local experience with ACI (long-term safety and efficacy), setting an appropriate standard for further clinical investigations into allogeneic progenitor cell-based orthopedic protocols.
Collapse
Affiliation(s)
- Virginie Philippe
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Annick Jeannerat
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| | - Cédric Peneveyre
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| | - Sandra Jaccoud
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- Laboratory of Biomechanical Orthopedics, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- STI School of Engineering, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Wassim Raffoul
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Salim Darwiche
- Musculoskeletal Research Unit, Vetsuisse Faculty, University of Zurich, CH-8057 Zurich, Switzerland;
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Robin Martin
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Alexis Laurent
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| |
Collapse
|
4
|
Philippe V, Laurent A, Hirt-Burri N, Abdel-Sayed P, Scaletta C, Schneebeli V, Michetti M, Brunet JF, Applegate LA, Martin R. Retrospective Analysis of Autologous Chondrocyte-Based Cytotherapy Production for Clinical Use: GMP Process-Based Manufacturing Optimization in a Swiss University Hospital. Cells 2022; 11:1016. [PMID: 35326468 PMCID: PMC8947208 DOI: 10.3390/cells11061016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Cultured autologous human articular chondrocyte (HAC) implantation has been extensively investigated for safe and effective promotion of structural and functional restoration of knee cartilage lesions. HAC-based cytotherapeutic products for clinical use must be manufactured under an appropriate quality assurance system and follow good manufacturing practices (GMP). A prospective clinical trial is ongoing in the Lausanne University Hospital, where the HAC manufacturing processes have been implemented internally. Following laboratory development and in-house GMP transposition of HAC cell therapy manufacturing, a total of 47 patients have been treated to date. The main aim of the present study was to retrospectively analyze the available manufacturing records of the produced HAC-based cytotherapeutic products, outlining the inter-individual variability existing among the 47 patients regarding standardized transplant product preparation. These data were used to ameliorate and to ensure the continued high quality of cytotherapeutic care in view of further clinical investigations, based on the synthetic analyses of existing GMP records. Therefore, a renewed risk analysis-based process definition was performed, with specific focus set on process parameters, controls, targets, and acceptance criteria. Overall, high importance of the interdisciplinary collaboration and of the manufacturing process robustness was underlined, considering the high variability (i.e., quantitative, functional) existing between the treated patients and between the derived primary HAC cell types.
Collapse
Affiliation(s)
- Virginie Philippe
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (V.S.); (R.M.)
| | - Alexis Laurent
- Manufacturing Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland;
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
- DLL Bioengineering, Discovery Learning Program, STI School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Valentine Schneebeli
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (V.S.); (R.M.)
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Murielle Michetti
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Jean-François Brunet
- Cell Production Center, Service of Pharmacy, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland;
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Robin Martin
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (V.S.); (R.M.)
| |
Collapse
|
5
|
Nasrollahzadeh N, Karami P, Wang J, Bagheri L, Guo Y, Abdel-Sayed P, Laurent-Applegate L, Pioletti DP. Temperature evolution following joint loading promotes chondrogenesis by synergistic cues via calcium signaling. eLife 2022; 11:72068. [PMID: 35256051 PMCID: PMC8903839 DOI: 10.7554/elife.72068] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 02/12/2022] [Indexed: 12/29/2022] Open
Abstract
During loading of viscoelastic tissues, part of the mechanical energy is transformed into heat that can locally increase the tissue temperature, a phenomenon known as self-heating. In the framework of mechanobiology, it has been accepted that cells react and adapt to mechanical stimuli. However, the cellular effect of temperature increase as a by-product of loading has been widely neglected. In this work, we focused on cartilage self-heating to present a 'thermo-mechanobiological' paradigm, and demonstrate how the coupling of a biomimetic temperature evolution and mechanical loading could influence cell behavior. We thereby developed a customized in vitro system allowing to recapitulate pertinent in vivo physical cues and determined the cells chondrogenic response to thermal and/or mechanical stimuli. Cellular mechanisms of action and potential signaling pathways of thermo-mechanotransduction process were also investigated. We found that co-existence of thermo-mechanical cues had a superior effect on chondrogenic gene expression compared to either signal alone. Specifically, the expression of Sox9 was significantly upregulated by application of the physiological thermo-mechanical stimulus. Multimodal transient receptor potential vanilloid 4 (TRPV4) channels were identified as key mediators of thermo-mechanotransduction process, which becomes ineffective without external calcium sources. We also observed that the isolated temperature evolution, as a by-product of loading, is a contributing factor to the cell response and this could be considered as important as the conventional mechanical loading. Providing an optimal thermo-mechanical environment by synergy of heat and loading portrays new opportunity for development of novel treatments for cartilage regeneration and can furthermore signal key elements for emerging cell-based therapies.
Collapse
Affiliation(s)
- Naser Nasrollahzadeh
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Jian Wang
- Institut des Matériaux et Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Lausanne, Switzerland
| | - Lida Bagheri
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Lee Laurent-Applegate
- Regenerative Therapy Unit, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| |
Collapse
|
6
|
Laurent A, Porcello A, Fernandez PG, Jeannerat A, Peneveyre C, Abdel-Sayed P, Scaletta C, Hirt-Burri N, Michetti M, de Buys Roessingh A, Raffoul W, Allémann E, Jordan O, Applegate LA. Combination of Hyaluronan and Lyophilized Progenitor Cell Derivatives: Stabilization of Functional Hydrogel Products for Therapeutic Management of Tendinous Tissue Disorders. Pharmaceutics 2021; 13:2196. [PMID: 34959477 PMCID: PMC8706504 DOI: 10.3390/pharmaceutics13122196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 01/10/2023] Open
Abstract
Cultured progenitor cells and derivatives have been used in various homologous applications of cutaneous and musculoskeletal regenerative medicine. Active pharmaceutical ingredients (API) in the form of progenitor cell derivatives such as lysates and lyophilizates were shown to retain function in controlled cellular models of wound repair. On the other hand, hyaluronan-based hydrogels are widely used as functional vehicles in therapeutic products for tendon tissue disorders. The aim of this study was the experimental characterization of formulations containing progenitor tenocyte-derived APIs and hyaluronan, for the assessment of ingredient compatibility and stability in view of eventual therapeutic applications in tendinopathies. Lyophilized APIs were determined to contain relatively low quantities of proteins and growth factors, while being physicochemically stable and possessing significant intrinsic antioxidant properties. Physical and rheological quantifications of the combination formulas were performed after hydrogen peroxide challenge, outlining significantly improved evolutive viscoelasticity values in accelerated degradation settings. Thus, potent effects of physicochemical protection or stability enhancement of hyaluronan by the incorporated APIs were observed. Finally, combination formulas were found to be easily injectable into ex vivo tendon tissues, confirming their compatibility with further translational clinical approaches. Overall, this study provides the technical bases for the development of progenitor tenocyte derivative-based injectable therapeutic products or devices, to potentially be applied in tendinous tissue disorders.
Collapse
Affiliation(s)
- Alexis Laurent
- Applied Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland; (A.J.); (C.P.)
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Lausanne, Switzerland; (P.A.-S.); (C.S.); (N.H.-B.); (M.M.); (L.A.A.)
- Manufacturing Department, TEC-PHARMA SA, CH-1038 Bercher, Switzerland
| | - Alexandre Porcello
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland; (A.P.); (P.G.F.); (E.A.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Paula Gonzalez Fernandez
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland; (A.P.); (P.G.F.); (E.A.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Annick Jeannerat
- Applied Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland; (A.J.); (C.P.)
| | - Cédric Peneveyre
- Applied Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland; (A.J.); (C.P.)
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Lausanne, Switzerland; (P.A.-S.); (C.S.); (N.H.-B.); (M.M.); (L.A.A.)
- DLL Bioengineering, Discovery Learning Program, STI School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Lausanne, Switzerland; (P.A.-S.); (C.S.); (N.H.-B.); (M.M.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Lausanne, Switzerland; (P.A.-S.); (C.S.); (N.H.-B.); (M.M.); (L.A.A.)
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Lausanne, Switzerland; (P.A.-S.); (C.S.); (N.H.-B.); (M.M.); (L.A.A.)
| | - Anthony de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland; (A.P.); (P.G.F.); (E.A.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland; (A.P.); (P.G.F.); (E.A.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Lausanne, Switzerland; (P.A.-S.); (C.S.); (N.H.-B.); (M.M.); (L.A.A.)
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| |
Collapse
|
7
|
Laurent A, Abdel-Sayed P, Scaletta C, Laurent P, Laurent E, Michetti M, de Buys Roessingh A, Raffoul W, Hirt-Burri N, Applegate LA. Back to the Cradle of Cytotherapy: Integrating a Century of Clinical Research and Biotechnology-Based Manufacturing for Modern Tissue-Specific Cellular Treatments in Switzerland. Bioengineering (Basel) 2021; 8:bioengineering8120221. [PMID: 34940374 PMCID: PMC8698568 DOI: 10.3390/bioengineering8120221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Empirically studied by Dr. Brown-Séquard in the late 1800s, cytotherapies were later democratized by Dr. Niehans during the twentieth century in Western Switzerland. Many local cultural landmarks around the Léman Riviera are reminiscent of the inception of such cell-based treatments. Despite the discreet extravagance of the remaining heirs of "living cell therapy" and specific enforcements by Swiss health authorities, current interest in modern and scientifically sound cell-based regenerative medicine has never been stronger. Respective progress made in bioengineering and in biotechnology have enabled the clinical implementation of modern cell-based therapeutic treatments within updated medical and regulatory frameworks. Notably, the Swiss progenitor cell transplantation program has enabled the gathering of two decades of clinical experience in Lausanne for the therapeutic management of cutaneous and musculoskeletal affections, using homologous allogeneic cell-based approaches. While striking conceptual similarities exist between the respective works of the fathers of cytotherapy and of modern highly specialized clinicians, major and important iterative updates have been implemented, centered on product quality and risk-analysis-based patient safety insurance. This perspective article highlights some historical similarities and major evolutive differences, particularly regarding product safety and quality issues, characterizing the use of cell-based therapies in Switzerland over the past century. We outline the vast therapeutic potential to be harnessed for the benefit of overall patient health and the importance of specific scientific methodological aspects.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, 1066 Epalinges, Switzerland; (A.L.); (P.A.-S.); (C.S.); (M.M.); (N.H.-B.)
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland;
- Applied Research Department, LAM Biotechnologies SA, 1066 Epalinges, Switzerland
- Manufacturing Department, TEC-PHARMA SA, 1038 Bercher, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, 1066 Epalinges, Switzerland; (A.L.); (P.A.-S.); (C.S.); (M.M.); (N.H.-B.)
- DLL Bioengineering, Discovery Learning Program, STI School of Engineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, 1066 Epalinges, Switzerland; (A.L.); (P.A.-S.); (C.S.); (M.M.); (N.H.-B.)
| | - Philippe Laurent
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland;
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
- Private Practice, Pharmacie du Gros-de-Vaud SA, 1038 Bercher, Switzerland;
| | - Elénie Laurent
- Private Practice, Pharmacie du Gros-de-Vaud SA, 1038 Bercher, Switzerland;
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, 1066 Epalinges, Switzerland; (A.L.); (P.A.-S.); (C.S.); (M.M.); (N.H.-B.)
| | - Anthony de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Wassim Raffoul
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland;
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, 1066 Epalinges, Switzerland; (A.L.); (P.A.-S.); (C.S.); (M.M.); (N.H.-B.)
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland;
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, 1066 Epalinges, Switzerland; (A.L.); (P.A.-S.); (C.S.); (M.M.); (N.H.-B.)
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland;
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, 8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
- Correspondence: ; Tel.: +41-21-314-35-10
| |
Collapse
|
8
|
Walzer SM, Toegel S, Chiari C, Farr S, Rinner B, Weinberg AM, Weinmann D, Fischer MB, Windhager R. A 3-Dimensional In Vitro Model of Zonally Organized Extracellular Matrix. Cartilage 2021; 13:336S-345S. [PMID: 31370667 PMCID: PMC8804753 DOI: 10.1177/1947603519865320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Functional cartilage repair requires the new formation of organized hyaline cartilaginous matrix to avoid the generation of fibrous repair tissue. The potential of mesenchymal progenitors was used to assemble a 3-dimensional structure in vitro, reflecting the zonation of collagen matrix in hyaline articular cartilage. DESIGN The 3-dimensional architecture of collagen alignment in pellet cultures of chondroprogenitors (CPs) was assessed with Picrosirius red staining analyzed under polarized light. In parallel assays, the trilineage capability was confirmed by calcium deposition during osteogenesis by alizarin S staining and alkaline phosphatase staining. Using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), mRNA levels of ALP, RUNX2, and BGLAP were assessed after 21 days of osteoinduction. Lipid droplets were stained with oil red O and adipogenic differentiation was confirmed by RT-qPCR analysis of PPARG and LPL gene expression. RESULTS Under conditions promoting the chondrogenic signature in self-assembling constructs, CPs formed an aligned extracellular matrix, positive for glycosaminoglycans and collagen type II, showing developing zonation of birefringent collagen fibers along the cross section of pellets, which reflect the distribution of collagen fibers in hyaline cartilage. Induced osteogenic and adipogenic differentiation confirmed the trilineage potential of CPs. CONCLUSION This model promotes the differentiation and self-organization of postnatal chondroprogenitors, resulting in the formation of zonally organized engineered hyaline cartilage comparable to the 3 zones of native cartilage.
Collapse
Affiliation(s)
- Sonja M. Walzer
- Karl Chiari Lab for Orthopaedic Biology,
Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna,
Austria,Sonja M. Walzer, Karl Chiari Lab for
Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical
University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria.
| | - Stefan Toegel
- Karl Chiari Lab for Orthopaedic Biology,
Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna,
Austria
| | - Catharina Chiari
- Karl Chiari Lab for Orthopaedic Biology,
Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna,
Austria
| | | | - Beate Rinner
- Division of Biomedical Research, Medical
University of Graz, Graz, Steiermark, Austria
| | - Annelie-Martina Weinberg
- Department of Orthopaedic and Trauma
Surgery, Medical University of Graz, Graz, Steiermark, Austria
| | - Daniela Weinmann
- Karl Chiari Lab for Orthopaedic Biology,
Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna,
Austria
| | - Michael B. Fischer
- Center for Biomedical Technology, Danube
University Krems, Krems an der Donau, Austria,Clinic for Bloodgroup Serology and
Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopaedic Biology,
Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna,
Austria
| |
Collapse
|
9
|
Tosoratti E, Fisch P, Taylor S, Laurent‐Applegate LA, Zenobi‐Wong M. 3D-Printed Reinforcement Scaffolds with Targeted Biodegradation Properties for the Tissue Engineering of Articular Cartilage. Adv Healthc Mater 2021; 10:e2101094. [PMID: 34633151 PMCID: PMC11469315 DOI: 10.1002/adhm.202101094] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/20/2021] [Indexed: 01/03/2023]
Abstract
Achieving regeneration of articular cartilage is challenging due to the low healing capacity of the tissue. Appropriate selection of cell source, hydrogel, and scaffold materials are critical to obtain good integration and long-term stability of implants in native tissues. Specifically, biomechanical stability and in vivo integration can be improved if the rate of degradation of the scaffold material matches the stiffening of the sample by extracellular matrix secretion of the encapsulated cells. To this end, a novel 3D-printed lactide copolymer is presented as a reinforcement scaffold for an enzymatically crosslinked hyaluronic acid hydrogel. In this system, the biodegradable properties of the reinforced scaffold are matched to the matrix deposition of articular chondrocytes embedded in the hydrogel. The lactide reinforcement provides stability to the soft hydrogel in the early stages, allowing the composite to be directly implanted in vivo with no need for a preculture period. Compared to pure cellular hydrogels, maturation and matrix secretion remain unaffected by the reinforced scaffold. Furthermore, excellent biocompatibility and production of glycosaminoglycans and collagens are observed at all timepoints. Finally, in vivo subcutaneous implantation in nude mice shows cartilage-like tissue maturation, indicating the possibility for the use of these composite materials in one-step surgical procedures.
Collapse
Affiliation(s)
- Enrico Tosoratti
- Institute for BiomechanicsOtto‐Stern‐Weg 7, ETH ZürichZürichCH‐8093Switzerland
| | - Philipp Fisch
- Institute for BiomechanicsOtto‐Stern‐Weg 7, ETH ZürichZürichCH‐8093Switzerland
| | - Scott Taylor
- Poly‐Med Inc51 Technology DriveAndersonSC29625USA
| | - Lee Ann Laurent‐Applegate
- Regenerative Therapy UnitLausanne University HospitalUniversity of LausanneÉpalingesCH‐1066Switzerland
- Center for Applied Biotechnology and Molecular MedicineUniversity of ZürichZürichCH‐8057Switzerland
| | - Marcy Zenobi‐Wong
- Institute for BiomechanicsOtto‐Stern‐Weg 7, ETH ZürichZürichCH‐8093Switzerland
| |
Collapse
|
10
|
Nguyen TPT, Li F, Shrestha S, Tuan RS, Thissen H, Forsythe JS, Frith JE. Cell-laden injectable microgels: Current status and future prospects for cartilage regeneration. Biomaterials 2021; 279:121214. [PMID: 34736147 DOI: 10.1016/j.biomaterials.2021.121214] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/19/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022]
Abstract
Injectable hydrogels have been employed extensively as versatile materials for cartilage regeneration due to their excellent biocompatibility, tunable structure, and ability to accommodate bioactive factors, as well as their ability to be locally delivered via minimally invasive injection to fill irregular defects. More recently, in vitro and in vivo studies have revealed that processing these materials to produce cell-laden microgels can enhance cell-cell and cell-matrix interactions and boost nutrient and metabolite exchange. Moreover, these studies have demonstrated gene expression profiles and matrix regeneration that are superior compared to conventional injectable bulk hydrogels. As cell-laden microgels and their application in cartilage repair are moving closer to clinical translation, this review aims to present an overview of the recent developments in this field. Here we focus on the currently used biomaterials and crosslinking strategies, the innovative fabrication techniques being used for the production of microgels, the cell sources used, the signals used for induction of chondrogenic differentiation and the resultant biological responses, and the ability to create three-dimensional, functional cartilage tissues. In addition, this review also covers the current clinical approaches for repairing cartilage as well as specific challenges faced when attempting the regeneration of damaged cartilage tissue. New findings related to the macroporous nature of the structures formed by the assembled microgel building blocks and the novel use of microgels in 3D printing for cartilage tissue engineering are also highlighted. Finally, we outline the challenges and future opportunities for employing cell-laden microgels in clinical applications.
Collapse
Affiliation(s)
- Thuy P T Nguyen
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Surakshya Shrestha
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| |
Collapse
|
11
|
Laurent A, Abdel-Sayed P, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh A, Raffoul W, Applegate LA. Evolution of Diploid Progenitor Lung Cell Applications: From Optimized Biotechnological Substrates to Potential Active Pharmaceutical Ingredients in Respiratory Tract Regenerative Medicine. Cells 2021; 10:2526. [PMID: 34685505 PMCID: PMC8533713 DOI: 10.3390/cells10102526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
The objective of this review is to describe the evolution of lung tissue-derived diploid progenitor cell applications, ranging from historical biotechnological substrate functions for vaccine production and testing to current investigations around potential therapeutic use in respiratory tract regenerative medicine. Such cell types (e.g., MRC-5 or WI-38 sources) were extensively studied since the 1960s and have been continuously used over five decades as safe and sustainable industrial vaccine substrates. Recent research and development efforts around diploid progenitor lung cells (e.g., FE002-Lu or Walvax-2 sources) consist in qualification for potential use as optimal and renewed vaccine production substrates and, alternatively, for potential therapeutic applications in respiratory tract regenerative medicine. Potentially effective, safe, and sustainable cell therapy approaches for the management of inflammatory lung diseases or affections and related symptoms (e.g., COVID-19 patients and burn patient severe inhalation syndrome) using local homologous allogeneic cell-based or cell-derived product administrations are considered. Overall, lung tissue-derived progenitor cells isolated and produced under good manufacturing practices (GMP) may be used with high versatility. They can either act as key industrial platforms optimally conforming to specific pharmacopoeial requirements or as active pharmaceutical ingredients (API) for potentially effective promotion of lung tissue repair or regeneration.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
- TEC-PHARMA SA, Manufacturing Department, CH-1038 Bercher, Switzerland
- LAM Biotechnologies SA, Manufacturing Department, CH-1066 Épalinges, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Anthony de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| |
Collapse
|
12
|
Laurent A, Abdel-Sayed P, Ducrot A, Hirt-Burri N, Scaletta C, Jaccoud S, Nuss K, de Buys Roessingh AS, Raffoul W, Pioletti D, von Rechenberg B, Applegate LA, Darwiche S. Development of Standardized Fetal Progenitor Cell Therapy for Cartilage Regenerative Medicine: Industrial Transposition and Preliminary Safety in Xenogeneic Transplantation. Biomolecules 2021; 11:250. [PMID: 33572428 PMCID: PMC7916236 DOI: 10.3390/biom11020250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/04/2021] [Accepted: 02/04/2021] [Indexed: 12/27/2022] Open
Abstract
Diverse cell therapy approaches constitute prime developmental prospects for managing acute or degenerative cartilaginous tissue affections, synergistically complementing specific surgical solutions. Bone marrow stimulation (i.e., microfracture) remains a standard technique for cartilage repair promotion, despite incurring the adverse generation of fibrocartilagenous scar tissue, while matrix-induced autologous chondrocyte implantation (MACI) and alternative autologous cell-based approaches may partly circumvent this effect. Autologous chondrocytes remain standard cell sources, yet arrays of alternative therapeutic biologicals present great potential for regenerative medicine. Cultured human epiphyseal chondro-progenitors (hECP) were proposed as sustainable, safe, and stable candidates for chaperoning cartilage repair or regeneration. This study describes the development and industrial transposition of hECP multi-tiered cell banking following a single organ donation, as well as preliminary preclinical hECP safety. Optimized cell banking workflows were proposed, potentially generating millions of safe and sustainable therapeutic products. Furthermore, clinical hECP doses were characterized as non-toxic in a standardized chorioallantoic membrane model. Lastly, a MACI-like protocol, including hECPs, was applied in a three-month GLP pilot safety evaluation in a caprine model of full-thickness articular cartilage defect. The safety of hECP transplantation was highlighted in xenogeneic settings, along with confirmed needs for optimal cell delivery vehicles and implantation techniques favoring effective cartilage repair or regeneration.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Aurélie Ducrot
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Sandra Jaccoud
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, CH-2002 Neuchâtel, Switzerland;
| | - Katja Nuss
- Musculoskeletal Research Unit, Zurich Tierspital, University of Zurich, CH-8952 Schlieren, Switzerland; (K.N.); (B.v.R.)
| | - Anthony S. de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Dominique Pioletti
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, CH-2002 Neuchâtel, Switzerland;
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit, Zurich Tierspital, University of Zurich, CH-8952 Schlieren, Switzerland; (K.N.); (B.v.R.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, Jiangsu, China
| | - Salim Darwiche
- Musculoskeletal Research Unit, Zurich Tierspital, University of Zurich, CH-8952 Schlieren, Switzerland; (K.N.); (B.v.R.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
13
|
Li F, Levinson C, Truong VX, Laurent-Applegate LA, Maniura-Weber K, Thissen H, Forsythe JS, Zenobi-Wong M, Frith JE. Microencapsulation improves chondrogenesis in vitro and cartilaginous matrix stability in vivo compared to bulk encapsulation. Biomater Sci 2020; 8:1711-1725. [PMID: 31994552 DOI: 10.1039/c9bm01524h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The encapsulation of cells into microgels is attractive for applications in tissue regeneration. While cells are protected against shear stress during injection, the assembly of microgels after injection into a tissue defect also forms a macroporous scaffold that allows effective nutrient transport throughout the construct. However, in most of current strategies that form microgel-based macroporous scaffold or higher-order structures, cells are seeded during or post the assembly process and not microencapsulated in situ. The objective of this study is to investigate the chondrogenic phenotype of microencapsulated fetal chondrocytes in a biocompatible, assembled microgel system vs. bulk gels and to test the stability of the constructs in vivo. Here, we demonstrate that cell microencapsulation leads to increased expression of cartilage-specific genes in a TGF-β1-dependent manner. This correlates, as shown by histological staining, with the ability of microencapsulated cells to deposit cartilaginous matrix after migrating to the surface of the microgels, while keeping a macroscopic granular morphology. Implantation of precultured scaffolds in a subcutaneous mouse model results in vessel infiltration in bulk gels but not in assembled microgels, suggesting a higher stability of the matrix produced by the cells in the assembled microgel constructs. The cells are able to remodel the microgels as demonstrated by the gradual disappearance of the granular structure in vivo. The biocompatible microencapsulation and microgel assembly system presented in this article therefore hold great promise as an injectable system for cartilage repair.
Collapse
Affiliation(s)
- Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, VIC 3800, Australia. and CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - Clara Levinson
- Tissue Engineering + Biofabrication, Department of Health Sciences and Technology, ETH Zürich, Switzerland.
| | - Vinh X Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, VIC 3800, Australia.
| | | | - Katharina Maniura-Weber
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biointerfaces, St. Gallen, Switzerland
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, VIC 3800, Australia.
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication, Department of Health Sciences and Technology, ETH Zürich, Switzerland.
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, VIC 3800, Australia.
| |
Collapse
|
14
|
Laurent A, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh AS, Raffoul W, Applegate LA. Holistic Approach of Swiss Fetal Progenitor Cell Banking: Optimizing Safe and Sustainable Substrates for Regenerative Medicine and Biotechnology. Front Bioeng Biotechnol 2020; 8:557758. [PMID: 33195124 PMCID: PMC7644790 DOI: 10.3389/fbioe.2020.557758] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Safety, quality, and regulatory-driven iterative optimization of therapeutic cell source selection has constituted the core developmental bedrock for primary fetal progenitor cell (FPC) therapy in Switzerland throughout three decades. Customized Fetal Transplantation Programs were pragmatically devised as straightforward workflows for tissue procurement, traceability maximization, safety, consistency, and robustness of cultured progeny cellular materials. Whole-cell bioprocessing standardization has provided plethoric insights into the adequate conjugation of modern biotechnological advances with current restraining legislative, ethical, and regulatory frameworks. Pioneer translational advances in cutaneous and musculoskeletal regenerative medicine continuously demonstrate the therapeutic potential of FPCs. Extensive technical and clinical hindsight was gathered by managing pediatric burns and geriatric ulcers in Switzerland. Concomitant industrial transposition of dermal FPC banking, following good manufacturing practices, demonstrated the extensive potential of their therapeutic value. Furthermore, in extenso, exponential revalorization of Swiss FPC technology may be achieved via the renewal of integrative model frameworks. Consideration of both longitudinal and transversal aspects of simultaneous fetal tissue differential processing allows for a better understanding of the quasi-infinite expansion potential within multi-tiered primary FPC banking. Multiple fetal tissues (e.g., skin, cartilage, tendon, muscle, bone, lung) may be simultaneously harvested and processed for adherent cell cultures, establishing a unique model for sustainable therapeutic cellular material supply chains. Here, we integrated fundamental, preclinical, clinical, and industrial developments embodying the scientific advances supported by Swiss FPC banking and we focused on advances made to date for FPCs that may be derived from a single organ donation. A renewed model of single organ donation bioprocessing is proposed, achieving sustained standards and potential production of billions of affordable and efficient therapeutic doses. Thereby, the aim is to validate the core therapeutic value proposition, to increase awareness and use of standardized protocols for translational regenerative medicine, potentially impacting millions of patients suffering from cutaneous and musculoskeletal diseases. Alternative applications of FPC banking include biopharmaceutical therapeutic product manufacturing, thereby indirectly and synergistically enhancing the power of modern therapeutic armamentariums. It is hypothesized that a single qualifying fetal organ donation is sufficient to sustain decades of scientific, medical, and industrial developments, as technological optimization and standardization enable high efficiency.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Tec-Pharma SA, Bercher, Switzerland
- LAM Biotechnologies SA, Épalinges, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Anthony S. de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Wassim Raffoul
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Oxford Suzhou Center for Advanced Research, Science and Technology Co., Ltd., Oxford University, Suzhou, China
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Levinson C, Lee M, Applegate LA, Zenobi-Wong M. An injectable heparin-conjugated hyaluronan scaffold for local delivery of transforming growth factor β1 promotes successful chondrogenesis. Acta Biomater 2019; 99:168-180. [PMID: 31536840 DOI: 10.1016/j.actbio.2019.09.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/05/2019] [Accepted: 09/11/2019] [Indexed: 02/08/2023]
Abstract
Cartilage lacks basic repair mechanisms and thus surgical interventions are necessary to treat lesions. Minimally-invasive arthroscopic procedures require the development of injectable biomaterials to support chondrogenesis of implanted cells. However, most cartilage tissue engineering approaches rely on pre-culture of scaffolds in media containing growth factors (GFs) such as transforming growth factor (TGF)-β1, which are crucial for cartilage formation and homeostasis. GFs media-supplementation is incompatible with injectable approaches and has led to a knowledge gap about optimal dose of GFs and release profiles needed to achieve chondrogenesis. This study aims to determine the optimal loading and release kinetics of TGF-β1 bound to an engineered GAG hydrogel to promote optimal cartilaginous matrix production in absence of TGF-β1 media-supplementation. We show that heparin, a GAG known to bind a wide range of GFs, covalently conjugated to a hyaluronan hydrogel, leads to a sustained release of TGF-β1. Using this heparin-conjugated hyaluronan hydrogel, 0.25 to 50 ng TGF-β1 per scaffold was loaded and cell viability, proliferation and cartilaginous matrix deposition of the encapsulated chondroprogenitor cells were measured. Excellent chondrogenesis was found when 5 ng TGF-β1 per scaffold and higher were used. We also demonstrate the necessity of a sustained release of TGF-β1, as no matrix deposition is observed upon a burst release. In conclusion, our biomaterial loaded with an optimal initial dose of 5 ng/scaffold TGF-β1 is a promising injectable material for cartilage repair, with potentially increased safety due to the low, locally administered GF dose. STATEMENT OF SIGNIFICANCE: Cartilage cell-based products are dependent on exogenous growth factor supplementation in order for proper tissue maturation. However, for a one-step repair of defects without need for expensive tissue maturation, an injectable, growth factor loaded formulation is required. Here we show development of an injectable hyaluronan hydrogel, which achieves a sustained release of TGF-β1 due to covalent conjugation of heparin. These grafts matured into cartilaginous tissue in the absence of growth factor supplementation. Additionally, this system allowed us to screen TGF-β1 concentrations to determine the mimimum amount of growth factor required for chondrogenesis. This study represents a critical step towards development of a minimally-invasive, arthroscopic treatment for cartilage lesions.
Collapse
|
16
|
Nasrollahzadeh N, Karami P, Pioletti DP. Control of Dissipation Sources: A Central Aspect for Enhancing the Mechanical and Mechanobiological Performances of Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2019; 11:39662-39671. [PMID: 31565916 DOI: 10.1021/acsami.9b15450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Development of mechanically durable and biologically inductive hydrogels is a major challenge for load-bearing applications such as engineered cartilage. Dissipative capacity of articular cartilage is central to its functional behavior when submitted to loading. While fluid frictional drag is playing a significant role in this phenomenon, the flow-dependent source of dissipation is mostly overlooked in the design of hydrogel scaffolds. Herein, we propose an original strategy based on the combination of fluidic and polymeric dissipation sources to simultaneously enhance hydrogel mechanical and mechanobiological performances. The nondestructive dissipation processes were carefully designed by hybrid cross-linking of the hydrogel network and low permeability of the porous structure. It was found that intrachain and pore water distribution in the porous hydrogels improves the mechanical properties in high water fractions. In contrast to widely reported tough hydrogels presenting limited load support capability at low strain values, we obtained stiff and dissipative hydrogels with unique fatigue behavior. We showed that the fatigue resistance capability is not a function of morphology, dissipation level, and stiffness of the viscoelastic hydrogels but rather depends on the origin of the dissipation. Moreover, the preserved dissipation source under mechanical stimulation maintained a mechanoinductive niche for enhancing chondrogenesis owing to fluid frictional drag contribution. The proposed strategy can be widely used to design functional scaffolds in high loading demands for enduring physiological stimuli and generating regulatory cues to cells.
Collapse
Affiliation(s)
- Naser Nasrollahzadeh
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering , EPFL , 1015 Lausanne , Switzerland
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering , EPFL , 1015 Lausanne , Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering , EPFL , 1015 Lausanne , Switzerland
| |
Collapse
|
17
|
A comparative study of cartilage engineered constructs in immunocompromised, humanized and immunocompetent mice. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.regen.2018.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
18
|
Nasrollahzadeh N, Applegate LA, Pioletti DP. Development of an Effective Cell Seeding Technique: Simulation, Implementation, and Analysis of Contributing Factors. Tissue Eng Part C Methods 2017; 23:485-496. [PMID: 28602135 DOI: 10.1089/ten.tec.2017.0108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cell seeding in a biomaterial is an important process for tissue engineering applications. It helps to modulate tissue formation or to control initial conditions for mechanobiological studies. The compression release-induced suction (CRIS) seeding technique leads to active infiltration of the cell suspension toward the central region of the scaffold. Its effectiveness, however, may significantly vary depending on several controlling factors such as the rate of loading and unloading or scaffold architecture. We utilized a 2D axisymmetric finite element model to numerically evaluate the influence of a seeding loading regime on suction pressure and infiltration velocity of the cell suspension. The in vitro application of optimized CRIS seeding obtained from simulation showed significant effectiveness over a static seeding method. As simulation results predicted, the permeability of the scaffold directly influenced CRIS seeding effectiveness in vitro. By supplementing an optimized CRIS loading regime with slow rotation after seeding treatment, cell distribution through thickness was improved especially for scaffolds showing low permeability. Finally, we systematically analyzed the relative importance of permeability, thickness, or coating on cell seeding efficiency and uniformity using a full factorial design of experiments. We observed that permeability has a higher impact on the CRIS seeding than scaffold coating and thickness.
Collapse
Affiliation(s)
- Naser Nasrollahzadeh
- 1 Laboratory of Biomechanical Orthopedics, Institute of Bioengineering , EPFL, Lausanne, Switzerland
| | - Lee Ann Applegate
- 2 Regenerative Therapy Unit, Plastic and Reconstructive Surgery, University Hospital of Lausanne (CHUV) , Lausanne, Switzerland
| | - Dominique P Pioletti
- 1 Laboratory of Biomechanical Orthopedics, Institute of Bioengineering , EPFL, Lausanne, Switzerland
| |
Collapse
|
19
|
Broguiere N, Cavalli E, Salzmann GM, Applegate LA, Zenobi-Wong M. Factor XIII Cross-Linked Hyaluronan Hydrogels for Cartilage Tissue Engineering. ACS Biomater Sci Eng 2016; 2:2176-2184. [DOI: 10.1021/acsbiomaterials.6b00378] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicolas Broguiere
- ETH Zürich, Cartilage Engineering and
Regeneration Laboratory, HPL J20, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Emma Cavalli
- ETH Zürich, Cartilage Engineering and
Regeneration Laboratory, HPL J20, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | | | - Lee Ann Applegate
- Department
of Musculoskeletal Medicine, Regenerative Therapy Unit, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| | - Marcy Zenobi-Wong
- ETH Zürich, Cartilage Engineering and
Regeneration Laboratory, HPL J20, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
20
|
Studer D, Cavalli E, Formica FA, Kuhn GA, Salzmann G, Mumme M, Steinwachs MR, Laurent-Applegate LA, Maniura-Weber K, Zenobi-Wong M. Human chondroprogenitors in alginate-collagen hybrid scaffolds produce stable cartilage in vivo. J Tissue Eng Regen Med 2016; 11:3014-3026. [PMID: 27373220 DOI: 10.1002/term.2203] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 02/26/2016] [Accepted: 03/27/2016] [Indexed: 12/17/2022]
Abstract
The goal of this study was to evaluate human epiphyseal chondroprogenitor cells (ECPs) as a potential new cell source for cartilage regeneration. ECPs were compared to human bone marrow stromal cells (MSCs) and human adult articular chondrocytes (ACs) for their chondrogenic potential and phenotypic stability in vitro and in vivo. The cells were seeded in Optimaix-3D scaffolds at 5 × 104 cells/mm3 and gene expression, matrix production and mechanical properties were analysed up to 6 weeks. In vitro, ECPs synthesized consistently high collagen 2 and low collagen 10. AC-seeded constructs exhibited high donor variability in GAG/DNA values as well as in collagen 2 staining, but showed low collagen 10 production. MSCs, on the other hand, expressed high levels of collagen 2 but also of collagens 1 and 10, and were therefore not considered further. In vivo, there was considerable loss of matrix proteins in ECPs compared to in vitro cultured samples. To overcome this, a second implantation study investigated the effect of mixing cells with alginate prior to seeding in the scaffold. ECPs in alginate maintained their cartilage matrix and resisted mineralization and vessel infiltration better 6 weeks after subcutaneous implantation, whereas ACs lost their chondrogenic matrix completely. This study shows the great potential of ECPs as an off-the-shelf, highly chondrogenic cell type that produces stable cartilage in vivo. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Deborah Studer
- Cartilage Engineering and Regeneration, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Zürich, Switzerland.,Cellular and Molecular Bioengineering Research Laboratory, Department of Chemical and Petroleum Engineering, University of Calgary, Canada
| | - Emma Cavalli
- Cartilage Engineering and Regeneration, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Zürich, Switzerland
| | - Florian A Formica
- Cartilage Engineering and Regeneration, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Zürich, Switzerland
| | - Gisela Anne Kuhn
- Institute for Biomechanics and ETH Phenomics Centre (EPIC), ETH Zürich, Switzerland
| | | | - Marcus Mumme
- Department of Biomedicine, University Hospital Basel, Switzerland
| | | | - Lee Ann Laurent-Applegate
- Department of Musculoskeletal Medicine, Regenerative Therapy Unit, University Hospital of Lausanne (CHUV), Epalinges, Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Testing and Research, St.Gallen, Switzerland
| | - Marcy Zenobi-Wong
- Cartilage Engineering and Regeneration, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Zürich, Switzerland
| |
Collapse
|
21
|
Kim M, Kim J, Park SR, Park DY, Kim YJ, Choi BH, Min BH. Comparison of fetal cartilage-derived progenitor cells isolated at different developmental stages in a rat model. Dev Growth Differ 2016; 58:167-79. [PMID: 26889876 DOI: 10.1111/dgd.12267] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 12/24/2015] [Accepted: 01/05/2016] [Indexed: 11/30/2022]
Abstract
Fetal cartilage-derived progenitor cells (FCPCs) could be a useful cell source in cell-based therapies for cartilage disorders. However, their characteristics can vary depending on the developmental stages. The aim of this study was to compare the characteristics of rat FCPCs from the hind limb on embryonic day 14 (E14), E16 and E20 regarding proliferation, pluripotency, and differentiation. Morphologically, rat fetal cartilage tissue showed an increase in cartilaginous differentiation features (Safranin-O, type II collagen) and decrease in pluripotency marker (Sox2) in the order of E14, E16 and E20. E14 FCPCs showed significantly higher doubling time compared to E16 and E20 FCPCs. While the E14 FCPCs expressed pluripotent genes (Sox2, Oct4, Nanog), the E16 and E20 FCPCs expressed chondrogenic markers (Sox9, Col2a1, Acan). E20 FCPCs showed the highest ability to both chondrogenic and adipogenic differentiation and E14 FCPCs showed relatively better activity in osteogenic differentiation. Further analysis showed that E20 FCPCs expressed both adipogenic (C/ebpß) and osteogenic (Runx2, Sp7, Taz) transcription factors as well as chondrogenic transcription factors. Our results show an inverse relationship overall between the expression of pluripotency genes and that of chondrogenic and lineage-specific genes in FCPCs under development. Due to its exceptional proliferation and chondrogenic differentiation ability, fetal cells from epiphyseal cartilage (E20 in rats) may be a suitable cell source for cartilage regeneration.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Molecular Science & Technology, 206 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea.,Cell Therapy Center, Ajou University Hospital, 164 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea
| | - Jiyoung Kim
- Inha Research Institute for Medical Sciences, Inha University College of Medicine, 100 Inha-ro Nam-gu, Incheon, 22212, Korea
| | - So Ra Park
- Inha Research Institute for Medical Sciences, Inha University College of Medicine, 100 Inha-ro Nam-gu, Incheon, 22212, Korea
| | - Do Young Park
- Department of Anatomy, School of Medicine, 206 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea
| | - Young Jick Kim
- Cell Therapy Center, Ajou University Hospital, 164 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, 100 Inha-ro Nam-gu, Incheon, 22212, Korea
| | - Byoung-Hyun Min
- Department of Molecular Science & Technology, 206 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea.,Cell Therapy Center, Ajou University Hospital, 164 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea.,Department of Orthopedic Surgery, School of Medicine, 206 Worldcup-ro Yongtong-gu, Suwon, 16499, Korea
| |
Collapse
|
22
|
Abdel-Sayed P, Pioletti DP. Strategies for improving the repair of focal cartilage defects. Nanomedicine (Lond) 2015; 10:2893-905. [PMID: 26377158 DOI: 10.2217/nnm.15.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Articular cartilage, together with skin, was predicted to be one of the first tissues to be successfully engineered. However cartilage repair remains nowadays still elusive, as we are still not able to overcome the hurdles of creating biomaterials corresponding to the native properties of the tissue, and which operate in joints environment that is not favorable for regeneration. In this review, we give an overview of the outcome of current cartilage treatment techniques. Furthermore we present current research strategies for improving cartilage tissue engineering.
Collapse
Affiliation(s)
- Philippe Abdel-Sayed
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| |
Collapse
|
23
|
Moghadam MN, Pioletti DP. Biodegradable HEMA-based hydrogels with enhanced mechanical properties. J Biomed Mater Res B Appl Biomater 2015; 104:1161-9. [DOI: 10.1002/jbm.b.33469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/30/2015] [Accepted: 05/22/2015] [Indexed: 01/10/2023]
Affiliation(s)
| | - Dominique P. Pioletti
- Laboratory of Biomechanical Orthopedics; Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL)
| |
Collapse
|
24
|
Abdel-Sayed P, Darwiche SE, Kettenberger U, Pioletti DP. The role of energy dissipation of polymeric scaffolds in the mechanobiological modulation of chondrogenic expression. Biomaterials 2013; 35:1890-7. [PMID: 24331703 DOI: 10.1016/j.biomaterials.2013.11.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/17/2013] [Indexed: 10/25/2022]
Abstract
Mechanical stimulation has been proposed to induce chondrogenesis in cell-seeded scaffolds. However, the effects of mechanical stimuli on engineered cartilage may vary substantially between different scaffolds. This advocates for the need to identify an overarching mechanobiological variable. We hypothesize that energy dissipation of scaffolds subjected to dynamic loading may be used as a mechanobiology variable. The energy dissipation would furnish a general criterion to adjust the mechanical stimulation favoring chondrogenesis in scaffold. Epiphyseal chondro-progenitor cells were then subject to unconfined compression 2 h per day during four days in different scaffolds, which differ only by the level of dissipation they generated while keeping the same loading conditions. Scaffolds with higher dissipation levels upregulated the mRNA of chondrogenic markers. In contrast lower dissipation of scaffolds was associated with downregulation of chondrogenic markers. These results showed that energy dissipation could be considered as a mechanobiology variable in cartilage. This study also indicated that scaffolds with energy dissipation level close to the one of cartilage favors chondrogenic expression when dynamical loading is present.
Collapse
Affiliation(s)
- Philippe Abdel-Sayed
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPF Lausanne, Switzerland
| | - Salim E Darwiche
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPF Lausanne, Switzerland
| | - Ulrike Kettenberger
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPF Lausanne, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPF Lausanne, Switzerland.
| |
Collapse
|