1
|
Sasai M, Kitawaki Y, Umezawa A, Oie Y, Kamiya T, Kawamura T, Sato Y, Hosoya S, Sugawara H, Yamada K, Izumoto H, Mae Y, Kuroda T, Okada K. Challenges and opportunities in the compassionate use of out-of-specification products in autologous regenerative medicine. Stem Cell Res Ther 2025; 16:238. [PMID: 40361213 PMCID: PMC12076910 DOI: 10.1186/s13287-025-04343-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND In recent years, therapeutic preparations using patient-derived tissues have emerged as commercially approved regenerative medicine products for expanding treatment possibilities for patients with no other treatment options. Autologous cell-processed products, derived from the tissue of the patient, typically exhibit variability in raw material quality, resulting in the generation of out-of-specification (OOS) products. MAIN BODY The compassionate use of OOS products is also practiced by the Food and Drug Administration and European Medicines Agency; differences among the three regulatory authorities were investigated to identify challenges in Japan. For conditions with no alternative treatments and severe time constraints, OOS products are sometimes used under compassionate grounds, particularly in Japan, where they are administered within the framework of clinical trials. This approach, although ethical, imposes significant operational and administrative burdens on medical institutions and marketing authorisation holders, raising concerns about sustainability. We considered the rationalisation of the current system and reached the conclusion that it would not contribute to load reduction and sustainability; thus, we devised a new framework. CONCLUSION This study reviewed the compassionate use systems for OOS products in Japan, the United States, and Europe, highlighting current challenges and proposing a sustainable regulatory framework for future practice.
Collapse
Affiliation(s)
- Masao Sasai
- The University of Osaka Hospital, Osaka, Japan
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
| | - Yayoi Kitawaki
- Novartis Pharma K.K, Tokyo, Japan
- Forum for Innovative Regenerative Medicine, Tokyo, Japan
| | - Akihiro Umezawa
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
- National Center for Child Health and Development, Tokyo, Japan
| | - Yoshinori Oie
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
- Department of Ophthalmology, Graduate School of Medicine, The University of Osaka, Osaka, Japan
| | - Takahiro Kamiya
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
- Institute of Science Tokyo, Tokyo, Japan
| | - Takuji Kawamura
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
- Department of Cardiovascular Surgery, Graduate School of Medicine, The University of Osaka, Osaka, Japan
| | - Yoji Sato
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
- National Institute of Health Sciences, Kanagawa, Japan
| | - Satoshi Hosoya
- The Japanese Society for Regenerative Medicine, Tokyo, Japan
- National Center for Child Health and Development, Tokyo, Japan
| | - Hiroyuki Sugawara
- Forum for Innovative Regenerative Medicine, Tokyo, Japan
- Terumo Corp, Kanagawa, Japan
| | - Kazuto Yamada
- Forum for Innovative Regenerative Medicine, Tokyo, Japan
- Japan Tissue Engineering Co, Ltd, Aichi, Japan
| | - Hiroshi Izumoto
- Forum for Innovative Regenerative Medicine, Tokyo, Japan
- Nipro Corp, Osaka, Japan
| | - Yoshiyuki Mae
- Forum for Innovative Regenerative Medicine, Tokyo, Japan
- Nipro Corp, Osaka, Japan
| | - Tohru Kuroda
- Forum for Innovative Regenerative Medicine, Tokyo, Japan
| | - Kiyoshi Okada
- The Japanese Society for Regenerative Medicine, Tokyo, Japan.
- Department of Medical Innovation, The University of Osaka Hospital, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
2
|
Adel A, Abdul-Hamid M, Abdel-Kawi SH, A. Abdelaziz M, Sakr HI, Ahmed OM. Bone marrow-derived mesenchymal stem cells reduce CCl 4-induced kidney injury and fibrosis in male Wistar rats. Ren Fail 2024; 46:2319330. [PMID: 39049729 PMCID: PMC11275530 DOI: 10.1080/0886022x.2024.2319330] [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: 12/04/2023] [Revised: 01/25/2024] [Accepted: 02/11/2024] [Indexed: 07/27/2024] Open
Abstract
AIM This study explores the possible therapeutic role of rats and mice bone marrow-derived mesenchymal stem cells (BM-MSCs) on renal damage and toxicity brought on by carbon tetrachloride (CCl4) in Wistar rats. METHODS Following an intraperitoneal injection of CCl4 (0.5 mL/kg b.w. twice weekly) for eight weeks, male Wistar rats were intravenously treated with rats and mice BM-MSCs (1 × 106 cells in 0.2 mL Dulbecco's Modified Eagle Medium (DMEM)/rat/week) a week for four weeks. Kidney functions were evaluated and kidney samples were examined using hematoxylin and eosin (H&E), Masson's trichrome (MT) staining techniques, and electron microscopy analysis. Kidney cyclooxygenase-2 (COX-2), protein 53 (p53), and tumor necrosis factor-α (TNF-α) were detected by immunohistochemical staining techniques. Additionally, bioindicators of oxidative stress and antioxidant defense systems were identified in kidney tissue. RESULTS In CCl4-injected rats, serum creatinine, urea, and uric acid levels significantly increased, as did renal lipid peroxidation (LPO), while superoxide dismutase, glutathione peroxidase (GPx), glutathione (GSH) transferase, and GSH levels significantly dropped in the kidneys. Histologically, the kidneys displayed a wide range of structural abnormalities, such as glomerular shrinkage, tubular dilations, inflammatory leukocytic infiltration, fibroblast proliferation, and elevated collagen content. Inflammatory cytokines like COX-2 and TNF-α as well as the pro-apoptotic mediator p53 were considerably upregulated. Treatment of BM-MSCs from mice and rats with CCl4-injected rats considerably reduced the previously noted abnormalities. CONCLUSIONS By boosting antioxidant defense and reducing apoptosis and inflammation, BM-MSCs from mice and rats were able to enhance kidney function and histological integrity in rats that had received CCl4 injections.
Collapse
Affiliation(s)
- Asmaa Adel
- Histology, Cell Biology and Genetic Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Manal Abdul-Hamid
- Histology, Cell Biology and Genetic Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Samraa H. Abdel-Kawi
- Medical Histology and Cell Biology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A. Abdelaziz
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Hader I. Sakr
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Medical Physiology, General Medicine Practice Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Osama M. Ahmed
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
3
|
Soma T, Oie Y, Takayanagi H, Matsubara S, Yamada T, Nomura M, Yoshinaga Y, Maruyama K, Watanabe A, Takashima K, Mao Z, Quantock AJ, Hayashi R, Nishida K. Induced pluripotent stem-cell-derived corneal epithelium for transplant surgery: a single-arm, open-label, first-in-human interventional study in Japan. Lancet 2024; 404:1929-1939. [PMID: 39522528 PMCID: PMC11694011 DOI: 10.1016/s0140-6736(24)01764-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/06/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The loss of corneal epithelial stem cells from the limbus at the edge of the cornea has severe consequences for vision, with the pathological manifestations of a limbal stem-cell deficiency (LSCD) difficult to treat. Here, to the best of our knowledge, we report the world's first use of corneal epithelial cell sheets derived from human induced pluripotent stem cells (iPSCs) to treat LSCD. METHODS This non-randomised, single-arm, clinical study involved four eyes of four patients with LSCD at the Department of Ophthalmology, Osaka University Hospital. They comprised a woman aged 44 years with idiopathic LSCD (patient 1), a man aged 66 years with ocular mucous membrane pemphigoid (patient 2), a man aged 72 years with idiopathic LSCD (patient 3), and a woman aged 39 years with toxic epidermal necrosis (patient 4). Allogeneic human iPSC-derived corneal epithelial cell sheets (iCEPSs) were transplanted onto affected eyes. This was done sequentially in two sets of HLA-mismatched surgeries, with patients 1 and 2 receiving low-dose cyclosporin and patients 3 and 4 not. The primary outcome measure was safety, ascertained by adverse events. These were monitored continuously throughout the 52-week follow-up period, and during an additional 1-year safety monitoring period. Secondary outcomes, reflective of efficacy, were also recorded. This study is registered with UMIN, UMIN000036539 and is complete. FINDINGS Patients were enrolled between June 17, 2019 and Nov 16, 2020. We had 26 adverse events during the 52-week follow-up period (consisting of 18 mild and one moderate event in treated eyes, and seven mild non-ocular events), with nine recorded in the additional 1-year safety monitoring period. No serious adverse events, such as tumourigenesis or clinical rejection, occurred during the whole 2-year observational period. At 52 weeks, secondary measures of efficacy showed that the disease stage had improved, corrected distance visual acuity was enhanced, and corneal opacification had diminished in all treated eyes. Corneal epithelial defects, subjective symptoms, quality-of-life questionnaire scores and corneal neovascularisation mostly improved or were unchanged. Overall, the beneficial efficacy outcomes achieved for patients 1 and 2 were better than those achieved for patients 3 and 4. INTERPRETATION iCEPS transplantation for LSCD was found to be safe throughout the study period. A larger clinical trial is planned to further investigate the efficacy of the procedure. FUNDING The Japan Agency for Medical Research and Development, the Ministry of Education, Culture, Sports, Science, and Technology-Japan, and the UK Biotechnology and Biological Sciences Research Council.
Collapse
Affiliation(s)
- Takeshi Soma
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yoshinori Oie
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Takayanagi
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shoko Matsubara
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomomi Yamada
- Department of Medical Innovation, Osaka University Hospital, Suita, Osaka, Japan
| | - Masaki Nomura
- Facility for iPS Cell Therapy, CiRA Foundation, Kyoto, Japan
| | - Yu Yoshinaga
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Vision Informatics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Atsushi Watanabe
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kayo Takashima
- Uehiro Research Division for iPS Cell Ethics, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Zaixing Mao
- R&D Division, Topcon Corporation, Tokyo, Japan
| | - Andrew J Quantock
- The School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - Ryuhei Hayashi
- Laboratory of Stem Cells and Applied Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
4
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Van Delen M, Derdelinckx J, Wouters K, Nelissen I, Cools N. A systematic review and meta-analysis of clinical trials assessing safety and efficacy of human extracellular vesicle-based therapy. J Extracell Vesicles 2024; 13:e12458. [PMID: 38958077 PMCID: PMC11220457 DOI: 10.1002/jev2.12458] [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: 12/22/2023] [Accepted: 05/03/2024] [Indexed: 07/04/2024] Open
Abstract
Nowadays, it has become clear that extracellular vesicles (EVs) are not a cellular waste disposal vesicle but are an essential part of an intercellular communication system. Besides the use of EVs in biomarker studies and diagnostics, the potential of EV-therapeutics has been seen by many. They provide unique properties for disease therapy, including strong immune-modulatory actions, the possibility of engineering, low immunogenicity, and the capability of crossing biological barriers. Proof-of-concept of EV-therapeutics for various pathologies has been achieved in preclinical studies. However, clinical trials with EVs have only been emerging slowly. Here, we aim to provide a comprehensive overview of the current state-of-the-art concerning clinical studies using EVs in human therapy. By approaching the current knowledge in a systematic manner, we were able to include 21 reports for meta-analysis of safety and evaluation of efficacy outcomes. Overall, we have shown that EV-based therapy is safe with a low incidence of serious adverse events (SAE; 0.7% (95%-CI: 0.1-5.2%), and adverse events (AE; 4.4% (95%-CI: 0.7-22.2%). Subgroup analysis showed no significant difference in SAE when comparing autologous versus allogeneic administration, as well as engineered versus non-engineered EV products. A significantly higher number of AE was seen in autologous versus allogeneic administration. However, the clinical relevance remains questionable. Evaluation of the clinical outcomes of immunostimulatory, immunosuppressive or regenerative EV-therapies indicated improvement in the majority of treated patients. Despite these promising results, data need to be approached with caution due to a high heterogeneity in the EVs manufacturing methods, study design, and reporting of (S)AE. Overall, we conclude that EV-based therapy is safe and presents a promising opportunity in therapy. More efforts are needed in the standardization and harmonization of reporting of EV isolation and characterization data as well as in the reporting of (S)AE to allow inter-study comparison.
Collapse
Affiliation(s)
- Mats Van Delen
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio)University of AntwerpAntwerpenBelgium
- Health DepartmentFlemish Institute for Technological Research (VITO)MolBelgium
| | - Judith Derdelinckx
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio)University of AntwerpAntwerpenBelgium
- Clinical Trial Center (CTC), CRC Antwerp, Antwerp University HospitalUniversity of AntwerpEdegemBelgium
| | | | - Inge Nelissen
- Health DepartmentFlemish Institute for Technological Research (VITO)MolBelgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio)University of AntwerpAntwerpenBelgium
- Center for Cell Therapy and Regenerative Medicine (CCRG)Antwerp University HospitalEdegemBelgium
| |
Collapse
|
6
|
Gupta M, Rathored J. Hyperbaric oxygen therapy: future prospects in regenerative therapy and anti-aging. FRONTIERS IN AGING 2024; 5:1368982. [PMID: 38757145 PMCID: PMC11097100 DOI: 10.3389/fragi.2024.1368982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024]
Abstract
Hyperbaric Oxygen Therapy (HBOT) utilizes 100% oxygen at high atmospheric pressure for clinical applications. HBOT has proven to be an effective supplementary treatment for a variety of clinical and pathological disorders. HBOT's therapeutic results are based on the physiological effects of increased tissue oxygenation, or improved oxygen bioavailability. HBOT's current indications in illnesses like as wound healing, thermal or radiation burns, and tissue necrosis point to its function in facilitating the regeneration process. Various research has revealed that HBOT plays a function in vascularization, angiogenesis, and collagen production augmentation. Individual regeneration capacity is influenced by both environmental and genetic factors. Furthermore, the regenerating ability of different types of tissues varies, and this ability declines with age. HBOT affects physiological processes at the genetic level by altering gene expression, delaying cell senescence, and assisting in telomere length enhancement. The positive results in a variety of indications, ranging from tissue regeneration to better cognitive function, indicate that it has enormous potential in regenerative and anti-aging therapy.
Collapse
Affiliation(s)
- Manoj Gupta
- Datta Meghe Institute of Medical Sciences, Wardha, India
| | - Jaishriram Rathored
- Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| |
Collapse
|
7
|
Tabatabaei Hosseini BS, Meadows K, Gabriel V, Hu J, Kim K. Biofabrication of Cellulose-based Hydrogels for Advanced Wound Healing: A Special Emphasis on 3D Bioprinting. Macromol Biosci 2024; 24:e2300376. [PMID: 38031512 DOI: 10.1002/mabi.202300376] [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: 08/17/2023] [Revised: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Even with the current advancements in wound management, addressing most skin injuries and wounds continues to pose a significant obstacle for the healthcare industry. As a result, researchers are now focusing on creating innovative materials utilizing cellulose and its derivatives. Cellulose, the most abundant biopolymer in nature, has unique properties that make it a promising material for wound healing, such as biocompatibility, tunable physiochemical characteristics, accessibility, and low cost. 3D bioprinting technology has enabled the production of cellulose-based wound dressings with complex structures that mimic the extracellular matrix. The inclusion of bioactive molecules such as growth factors offers the ability to aid in promoting wound healing, while cellulose creates an ideal environment for controlled release of these biomolecules and moisture retention. The use of 3D bioprinted cellulose-based wound dressings has potential benefits for managing chronic wounds, burns, and painful wounds by promoting wound healing and reducing the risk of infection. This review provides an up-to-date summary of cellulose-based dressings manufactured by 3D bioprinting techniques by looking into wound healing biology, biofabrication methods, cellulose derivatives, and the existing cellulose bioinks targeted toward wound healing.
Collapse
Affiliation(s)
| | - Kieran Meadows
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Vincent Gabriel
- Calgary Firefighters Burn Treatment Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Jinguang Hu
- Department of Petroleum and Chemical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Keekyoung Kim
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
8
|
Forteza-Genestra MA, Antich-Rosselló M, Ramis-Munar G, Calvo J, Gayà A, Monjo M, Ramis JM. Comparative effect of platelet- and mesenchymal stromal cell-derived extracellular vesicles on human cartilage explants using an ex vivo inflammatory osteoarthritis model. Bone Joint Res 2023; 12:667-676. [PMID: 37852621 PMCID: PMC10584413 DOI: 10.1302/2046-3758.1210.bjr-2023-0109.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Aims Extracellular vesicles (EVs) are nanoparticles secreted by all cells, enriched in proteins, lipids, and nucleic acids related to cell-to-cell communication and vital components of cell-based therapies. Mesenchymal stromal cell (MSC)-derived EVs have been studied as an alternative for osteoarthritis (OA) treatment. However, their clinical translation is hindered by industrial and regulatory challenges. In contrast, platelet-derived EVs might reach clinics faster since platelet concentrates, such as platelet lysates (PL), are already used in therapeutics. Hence, we aimed to test the therapeutic potential of PL-derived extracellular vesicles (pEVs) as a new treatment for OA, which is a degenerative joint disease of articular cartilage and does not have any curative or regenerative treatment, by comparing its effects to those of human umbilical cord MSC-derived EVs (cEVs) on an ex vivo OA-induced model using human cartilage explants. Methods pEVs and cEVs were isolated by size exclusion chromatography (SEC) and physically characterized by nanoparticle tracking analysis (NTA), protein content, and purity. OA conditions were induced in human cartilage explants (10 ng/ml oncostatin M and 2 ng/ml tumour necrosis factor alpha (TNFα)) and treated with 1 × 109 particles of pEVs or cEVs for 14 days. Then, DNA, glycosaminoglycans (GAG), and collagen content were quantified, and a histological study was performed. EV uptake was monitored using PKH26 labelled EVs. Results Significantly higher content of DNA and collagen was observed for the pEV-treated group compared to control and cEV groups. No differences were found in GAG quantification nor in EVs uptake within any treated group. Conclusion In conclusion, pEVs showed better performance than cEVs in our in vitro OA model. Although further studies are needed, pEVs are shown as a potential alternative to cEVs for cell-free regenerative medicine.
Collapse
Affiliation(s)
- Maria A. Forteza-Genestra
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Miquel Antich-Rosselló
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Guillem Ramis-Munar
- Microscopy Area, Serveis Cietificotècnics, University of the Balearic Islands, Palma, Spain
| | - Javier Calvo
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Fundació Banc de Sang i Teixits de les Illes Balears (FBSTIB), Palma, Spain
| | - Antoni Gayà
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Fundació Banc de Sang i Teixits de les Illes Balears (FBSTIB), Palma, Spain
| | - Marta Monjo
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Joana M. Ramis
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
9
|
Thampi P, Samulski RJ, Grieger JC, Phillips JN, McIlwraith CW, Goodrich LR. Gene therapy approaches for equine osteoarthritis. Front Vet Sci 2022; 9:962898. [PMID: 36246316 PMCID: PMC9558289 DOI: 10.3389/fvets.2022.962898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 01/24/2023] Open
Abstract
With an intrinsically low ability for self-repair, articular cartilage injuries often progress to cartilage loss and joint degeneration resulting in osteoarthritis (OA). Osteoarthritis and the associated articular cartilage changes can be debilitating, resulting in lameness and functional disability both in human and equine patients. While articular cartilage damage plays a central role in the pathogenesis of OA, the contribution of other joint tissues to the pathogenesis of OA has increasingly been recognized thus prompting a whole organ approach for therapeutic strategies. Gene therapy methods have generated significant interest in OA therapy in recent years. These utilize viral or non-viral vectors to deliver therapeutic molecules directly into the joint space with the goal of reprogramming the cells' machinery to secrete high levels of the target protein at the site of injection. Several viral vector-based approaches have demonstrated successful gene transfer with persistent therapeutic levels of transgene expression in the equine joint. As an experimental model, horses represent the pathology of human OA more accurately compared to other animal models. The anatomical and biomechanical similarities between equine and human joints also allow for the use of similar imaging and diagnostic methods as used in humans. In addition, horses experience naturally occurring OA and undergo similar therapies as human patients and, therefore, are a clinically relevant patient population. Thus, further studies utilizing this equine model would not only help advance the field of human OA therapy but also benefit the clinical equine patients with naturally occurring joint disease. In this review, we discuss the advancements in gene therapeutic approaches for the treatment of OA with the horse as a relevant patient population as well as an effective and commonly utilized species as a translational model.
Collapse
Affiliation(s)
- Parvathy Thampi
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Joshua C. Grieger
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Jennifer N. Phillips
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - Laurie R. Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States,*Correspondence: Laurie R. Goodrich
| |
Collapse
|
10
|
Moghassemi S, Dadashzadeh A, de Azevedo RB, Amorim CA. Secure transplantation by tissue purging using photodynamic therapy to eradicate malignant cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 234:112546. [PMID: 36029759 DOI: 10.1016/j.jphotobiol.2022.112546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 12/17/2022]
Abstract
The field of photodynamic therapy (PDT) for treating various malignant neoplasms has been given researchers' attention due to its ability to be a selective and minimally invasive cancer therapy strategy. The possibility of tumor cell infection and hence high recurrence rates in cancer patients tends to restrict autologous transplantation. So, the photodynamic tissue purging process, which consists of selective photoinactivation of the malignant cells in the graft, is defined as a compromising strategy to purify contaminated tissues before transplantation. In this strategy, the direct malignant cells' death results from the reactive oxygen species (ROS) generation through the activation of a photosensitizer (PS) by light exposure in the presence of oxygen. Since new PS generations can effectively penetrate the tissue, PDT could be an ideal ex vivo tissue purging protocol that eradicates cancer cells derived from various malignancies. The challenge is that the applied pharmacologic ex vivo tissue purging should efficiently induce tumor cells with minor influence on normal tissue cells. This review aims to provide an overview of the current status of the most effective PDT strategies and PS development concerning their potential application in ex vivo purging before hematopoietic stem cell or ovarian tissue transplantation.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ricardo Bentes de Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília DF, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
11
|
Beheshtizadeh N, Gharibshahian M, Pazhouhnia Z, Rostami M, Zangi AR, Maleki R, Azar HK, Zalouli V, Rajavand H, Farzin A, Lotfibakhshaiesh N, Sefat F, Azami M, Webster TJ, Rezaei N. Commercialization and regulation of regenerative medicine products: Promises, advances and challenges. Biomed Pharmacother 2022; 153:113431. [DOI: 10.1016/j.biopha.2022.113431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/04/2022] [Accepted: 07/14/2022] [Indexed: 11/02/2022] Open
|
12
|
Clinical Trials Using Mesenchymal Stem Cells for Spinal Cord Injury: Challenges in Generating Evidence. Cells 2022; 11:cells11061019. [PMID: 35326470 PMCID: PMC8946989 DOI: 10.3390/cells11061019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) remains an important public health problem which often causes permanent loss of muscle strength, sensation, and function below the site of the injury, generating physical, psychological, and social impacts throughout the lives of the affected individuals, since there are no effective treatments available. The use of stem cells has been investigated as a therapeutic approach for the treatment of SCI. Although a significant number of studies have been conducted in pre-clinical and clinical settings, so far there is no established cell therapy for the treatment of SCI. One aspect that makes it difficult to evaluate the efficacy is the heterogeneity of experimental designs in the clinical trials that have been published. Cell transplantation methods vary widely among the trials, and there are still no standardized protocols or recommendations for the therapeutic use of stem cells in SCI. Among the different cell types, mesenchymal stem/stromal cells (MSCs) are the most frequently tested in clinical trials for SCI treatment. This study reviews the clinical applications of MSCs for SCI, focusing on the critical analysis of 17 clinical trials published thus far, with emphasis on their design and quality. Moreover, it highlights the need for more evidence-based studies designed as randomized controlled trials and potential challenges to be addressed in context of stem cell therapies for SCI.
Collapse
|
13
|
Liu D, Bobrovskaya L, Zhou XF. Cell Therapy for Neurological Disorders: The Perspective of Promising Cells. BIOLOGY 2021; 10:1142. [PMID: 34827135 PMCID: PMC8614777 DOI: 10.3390/biology10111142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022]
Abstract
Neurological disorders are big public health challenges that are afflicting hundreds of millions of people around the world. Although many conventional pharmacological therapies have been tested in patients, their therapeutic efficacies to alleviate their symptoms and slow down the course of the diseases are usually limited. Cell therapy has attracted the interest of many researchers in the last several decades and has brought new hope for treating neurological disorders. Moreover, numerous studies have shown promising results. However, none of the studies has led to a promising therapy for patients with neurological disorders, despite the ongoing and completed clinical trials. There are many factors that may affect the outcome of cell therapy for neurological disorders due to the complexity of the nervous system, especially cell types for transplantation and the specific disease for treatment. This paper provides a review of the various cell types from humans that may be clinically used for neurological disorders, based on their characteristics and current progress in related studies.
Collapse
Affiliation(s)
| | | | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; (D.L.); (L.B.)
| |
Collapse
|
14
|
Ghaffari S, Kazerooni H, Salehi-Najafabadi A. An overview of the recent findings of cell-based therapies for the treatment and management of COVID-19. Int Immunopharmacol 2021; 101:108226. [PMID: 34634685 PMCID: PMC8492917 DOI: 10.1016/j.intimp.2021.108226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/19/2021] [Accepted: 10/03/2021] [Indexed: 12/22/2022]
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic taking the lives of millions. The virus itself not only invades and destroys the angiotensin-converting enzyme 2 (ACE2)-expressing cells of the lungs, kidneys, liver, etc. but also elicits a hyperinflammatory immune response, further damaging the tissue leading to acute respiratory distress syndrome (ARDS) and death. Although vaccines, as a prime example of active immunotherapy, have clearly disrupted the transmission of virus and reduced mortality, hospitalization, and burden of disease, other avenues of immunotherapy are also being explored. One such approach would be to adoptively transfer modified/unmodified immune cells to the critically ill. Here, we compiled and summarized the immunopathogenesis of SARS-CoV-2 and the recent preclinical and clinical data on the potential of cell-based therapies in the fight against COVID-19.
Collapse
Affiliation(s)
- Sasan Ghaffari
- Department of Science and Technology, Supreme National Defense University, Tehran, Iran; Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran; Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| | - Hanif Kazerooni
- Department of Science and Technology, Supreme National Defense University, Tehran, Iran.
| | - Amir Salehi-Najafabadi
- Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran; Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
15
|
Motamedi S, Esfandpour A, Babajani A, Jamshidi E, Bahrami S, Niknejad H. The Current Challenges on Spray-Based Cell Delivery to the Skin Wounds. Tissue Eng Part C Methods 2021; 27:543-558. [PMID: 34541897 DOI: 10.1089/ten.tec.2021.0158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cell delivery through spray instruments is a promising and effective method in tissue engineering and regenerative medicine. It is used for treating different acute and chronic wounds, including burns with different etiologies, chronic diabetic or venous wounds, postcancer surgery, and hypopigmentation disorders. Cell spray can decrease the needed donor site area compared with conventional autologous skin grafting. Keratinocytes, fibroblasts, melanocytes, and mesenchymal stem cells are promising cell sources for cell spray procedures. Different spray instruments are designed and utilized to deliver the cells to the intended skin area. In an efficient spray instrument, cell viability and wound coverage are two determining parameters influenced by various physical and biological factors such as air pressure, spraying distance, viscosity of suspension, stiffness of the wound surface, and velocity of impact. Besides, to improve cell delivery by spray instruments, some matrices and growth factors can be added to cell suspensions. This review focuses on the different types of cells and spray instruments used in cell delivery procedures. It also discusses physical and biological parameters associated with cell viability and wound coverage in spray instruments. Moreover, the recent advances in codelivery of cells with biological glues and growth factors, as well as clinical translation of cell spraying, have been reviewed. Impact statement Skin wounds are a group of prevalent injuries that can lead to life-threatening complexities. As a focus of interest, stem cell therapy and spray-based cell delivery have effectively decreased associated morbidity and mortality. This review summarizes a broad scope of recent evidence related to spray-based cell therapy, instruments, and approaches adopted to make the process more efficient in treating skin wounds. An overview including utilized cell types, clinical cases, and current challenges is also provided.
Collapse
Affiliation(s)
- Shiva Motamedi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Esfandpour
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Jamshidi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Prat-Vidal C, Crisóstomo V, Moscoso I, Báez-Díaz C, Blanco-Blázquez V, Gómez-Mauricio G, Albericio G, Aguilar S, Fernández-Santos ME, Fernández-Avilés F, Sánchez-Margallo FM, Bayes-Genis A, Bernad A. Intracoronary Delivery of Porcine Cardiac Progenitor Cells Overexpressing IGF-1 and HGF in a Pig Model of Sub-Acute Myocardial Infarction. Cells 2021; 10:cells10102571. [PMID: 34685551 PMCID: PMC8534140 DOI: 10.3390/cells10102571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human cardiac progenitor cells (hCPC) are considered a good candidate in cell therapy for ischemic heart disease, demonstrating capacity to improve functional recovery after myocardial infarction (MI), both in small and large preclinical animal models. However, improvements are required in terms of cell engraftment and efficacy. Based on previously published reports, insulin-growth factor 1 (IGF-1) and hepatocyte growth factor (HGF) have demonstrated substantial cardioprotective, repair and regeneration activities, so they are good candidates to be evaluated in large animal model of MI. We have validated porcine cardiac progenitor cells (pCPC) and lentiviral vectors to overexpress IGF-1 (co-expressing eGFP) and HGF (co-expressing mCherry). pCPC were transduced and IGF1-eGFPpos and HGF-mCherrypos populations were purified by cell sorting and further expanded. Overexpression of IGF-1 has a limited impact on pCPC expression profile, whereas results indicated that pCPC-HGF-mCherry cultures could be counter selecting high expresser cells. In addition, pCPC-IGF1-eGFP showed a higher cardiogenic response, evaluated in co-cultures with decellularized extracellular matrix, compared with native pCPC or pCPC-HGF-mCherry. In vivo intracoronary co-administration of pCPC-IGF1-eGFP and pCPC-HFG-mCherry (1:1; 40 × 106/animal), one week after the induction of an MI model in swine, revealed no significant improvement in cardiac function.
Collapse
Affiliation(s)
- Cristina Prat-Vidal
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain; (C.P.-V.); (A.B.-G.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08908 L’Hospitalet de Llobregat, Spain
| | - Verónica Crisóstomo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Isabel Moscoso
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Claudia Báez-Díaz
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Virginia Blanco-Blázquez
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | | | - Guillermo Albericio
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
| | - Susana Aguilar
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
| | - María-Eugenia Fernández-Santos
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Laboratorio Investigación Traslacional en Cardiología (LITC), Unidad de Producción Celular-GMP (UPC-GMP), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), TERCEL, 28007 Madrid, Spain
| | - Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Laboratorio Investigación Traslacional en Cardiología (LITC), Unidad de Producción Celular-GMP (UPC-GMP), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), TERCEL, 28007 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Francisco M. Sánchez-Margallo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain; (C.P.-V.); (A.B.-G.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Cardiology Service, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
- Correspondence: ; Tel.: +34-915-855-424
| |
Collapse
|
17
|
Abdelgawad M, Bakry NS, Farghali AA, Abdel-Latif A, Lotfy A. Mesenchymal stem cell-based therapy and exosomes in COVID-19: current trends and prospects. Stem Cell Res Ther 2021; 12:469. [PMID: 34419143 PMCID: PMC8379570 DOI: 10.1186/s13287-021-02542-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Novel coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus-2. The virus causes an exaggerated immune response, resulting in a cytokine storm and acute respiratory distress syndrome, the leading cause of COVID-19-related mortality and morbidity. So far, no therapies have succeeded in circumventing the exacerbated immune response or cytokine storm associated with COVID-19. Mesenchymal stem cells (MSCs), through their immunomodulatory and regenerative activities, mostly mediated by their paracrine effect and extracellular vesicle production, have therapeutic potential in many autoimmune, inflammatory, and degenerative diseases. In this paper, we review clinical studies on the use of MSCs for COVID-19 treatment, including the salutary effects of MSCs on the pathophysiology of COVID-19 and the immunomodulation of the cytokine storm. Ongoing clinical trial designs, cell sources, dose and administration, and populations are summarized, and the paracrine mode of benefit is discussed. We also offer suggestions for optimizing MSC-based therapies, including genetic engineering, strategies for cell surface modification, nanotechnology applications, and combination therapies.
Collapse
Affiliation(s)
- Mai Abdelgawad
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt
| | - Nourhan Saied Bakry
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt
| | - Ahmed A Farghali
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt
| | - Ahmed Abdel-Latif
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA. .,College of Medicine, University of Kentucky, Lexington, KY, 40506-0046, USA.
| | - Ahmed Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt.
| |
Collapse
|
18
|
Kregar Velikonja N, Erjavec K, Knežević M. Knowledge, Awareness, and Attitudes toward Umbilical Cord Blood Biobanking. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Umbilical Cord Blood (UCB) stem cells are a non-invasive, effective alternative source of hematopoietic stem cells for the treatment of a variety of diseases.
AIM: The aim of the study was to research knowledge, awareness, and attitudes of the general public and health professionals regarding the UCB storage.
MATERIALS AND METHODS: A cross-sectional study was conducted with an online survey (n = 408) using the snowball method.
RESULTS: A majority of respondents declared that they were not aware of UCB banking; however, people with an academic background in healthcare have a greater awareness of UCB banking. A subjective assessment found that their knowledge of UCB banking is better than that of non-professionals, however, they exhibited better knowledge and understanding of UCB use and banking in only a few aspects. People with an academic background in healthcare have a more polarized attitude toward cord blood banking and a higher percentage are unwilling to pay for UCB banking services. However, their preferences regarding public/private UCB banking do not differ significantly from those of non-professionals.
CONCLUSION: Here, we show that there is not much difference between the laic and professional categories in terms of knowledge about the specific purpose and characteristics of UCB storage.
Collapse
|
19
|
Kim K, Bou-Ghannam S, Kameishi S, Oka M, Grainger DW, Okano T. Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. J Control Release 2020; 330:696-704. [PMID: 33347942 DOI: 10.1016/j.jconrel.2020.12.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
The evolution of drug discovery exploded in the early 20th century with the advent of critical scientific advancements in organic chemistry, chemical analysis, and purification. Early drug generations focused largely on symptom control and pain management, effective targets for small-molecule drugs. Recently, the attention in drug discovery has shifted to pursuit of radical cures. Cell therapy presents the ideal attributes of a promising new drug, targeting specific tissues based on chemotactic cues and modulating secretion of instructive regenerative molecules in response to dynamic signaling from disease environments. To actuate the therapeutic potential of cell therapy toward worldwide clinical use, cell delivery methods that can effectively localize and engraft mesenchymal stem cells (MSCs) with high disease-site fidelity and enable dynamic MSC bioactive function are paramount. In this review, we discuss the evolution of cell therapies with a focus on stem cell advantages, as well as the limitations to these therapies. This review aims to introduce cell sheet technology as a breakthrough cell therapy with demonstrated therapeutic success across indications for heart, liver, and kidney tissue regeneration. Opportunities and anticipated clinical impacts of cell sheet technology using MSCs are discussed.
Collapse
Affiliation(s)
- Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA.
| | - Sophia Bou-Ghannam
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, 36 South, Wasatch Drive, Salt Lake City, UT 84112, USA
| | - Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Masatoshi Oka
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, 36 South, Wasatch Drive, Salt Lake City, UT 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
20
|
Gay MHP, Baldomero H, Farge-Bancel D, Robey PG, Rodeo S, Passweg J, Müller-Gerbl M, Martin I. The Survey on Cellular and Tissue-Engineered Therapies in Europe in 2016 and 2017. Tissue Eng Part A 2020; 27:336-350. [PMID: 32680446 DOI: 10.1089/ten.tea.2020.0092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
This report describes activity in Europe for the years 2016 and 2017 in the area of cellular and tissue-engineered therapies, excluding hematopoietic stem cell treatments for the reconstitution of hematopoiesis. It is the eighth of its kind and is supported by five established scientific organizations. In 2016 and 2017, a combined 234 teams from 29 countries responded to the cellular and engineered tissue therapy survey; 227 teams reported treating 8236 patients in these 2 years. Indications were categorized in hematology/oncology (40%; predominantly prevention or treatment of graft vs. host disease and hematopoietic graft enhancement), musculoskeletal/rheumatological disorders (29%), cardiovascular disorders (6%), neurological disorders (4%), gastrointestinal disorders (<1%), as well as miscellaneous disorders (20%), which were not assigned to the previous indications. The predominantly used cells were autologous (61%). The majority of autologous cells were used to treat musculoskeletal/rheumatological (44%) disorders, whereas allogeneic cells were mainly used for hematology/oncology (78%). The reported cell types were mesenchymal stem/stromal cells (MSCs) (56%), hematopoietic cells (21%), keratinocytes (7%), chondrocytes (6%) dermal fibroblasts (4%), dendritic cells (2%), and other cell types (4%). Cells were expanded in vitro in 62% of the treatments, sorted in 11% of the cases, and rarely transduced (2%). The processing of cells was outsourced to external facilities in 30% of the cases. Cells were delivered predominantly intravenously or intra-arterially [47%], as suspension [36%], or using a membrane/scaffold (16%). The data are compared with those from previous years to identify trends in a rapidly evolving field. In this edition, the report includes a critical discussion of data collected in the space of orthopedics and the use of MSCs.
Collapse
Affiliation(s)
- Max H P Gay
- Department of Biomedicine, Institute of Anatomy, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Helen Baldomero
- EBMT Activity Survey Office, Division of Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Dominique Farge-Bancel
- Unité de Médecine Interne, Maladies Auto-immunes et Pathologie Vasculaire, Université de Paris, Paris, France.,MATHEC, Centre de Référence des Maladies auto-immunes systémiques Rares d'Ile-de-France, Hôpital St-Louis, Paris, France
| | - Pamela G Robey
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Scott Rodeo
- Orthopaedic Surgery, Weill Medical College of Cornell University, New York, New York, USA.,The Hospital for Special Surgery, New York, New York, USA
| | - Jakob Passweg
- EBMT Activity Survey Office, Division of Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Magdalena Müller-Gerbl
- Department of Biomedicine, Institute of Anatomy, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
21
|
Liu MC, Chang ML, Wang YC, Chen WH, Wu CC, Yeh SD. Revisiting the Regenerative Therapeutic Advances Towards Erectile Dysfunction. Cells 2020; 9:E1250. [PMID: 32438565 PMCID: PMC7290763 DOI: 10.3390/cells9051250] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Erectile dysfunction (ED) is an inability to attain or maintain adequate penile erection for successful vaginal intercourse, leading to sexual and relationship dissatisfaction. To combat ED, various surgical and non-surgical approaches have been developed in the past to restore erectile functions. These therapeutic interventions exhibit significant impact in providing relief to patients; however, due to their associated adverse effects and lack of long-term efficacy, newer modalities such as regenerative therapeutics have gained attention due to their safe and prolonged efficacy. Stem cells and platelet-derived biomaterials contained in platelet-rich plasma (PRP) are thriving as some of the major therapeutic regenerative agents. In recent years, various preclinical and clinical studies have evaluated the individual, as well as combined of stem cells and PRP to restore erectile function. Being rich in growth factors, chemokines, and angiogenic factors, both stem cells and PRP play a crucial role in regenerating nerve cells, myelination of axons, homing and migration of progenitor cells, and anti-fibrosis and anti-apoptosis of damaged cavernous nerve in corporal tissues. Further, platelet-derived biomaterials have been proven to be a biological supplement for enhancing the proliferative and differentiation potential of stem cells towards neurogenic fate. Therefore, this article comprehensively analyzes the progresses of these regenerative therapies for ED.
Collapse
Affiliation(s)
- Ming-Che Liu
- Department of Urology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (M.-C.L.); (C.-C.W.)
- Clinical Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, school of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Meng-Lin Chang
- Department of Urology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 242, Taiwan;
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Ya-Chun Wang
- TCM Biotech International Corp., New Taipei City 22175, Taiwan; (Y.-C.W.); (W.-H.C.)
| | - Wei-Hung Chen
- TCM Biotech International Corp., New Taipei City 22175, Taiwan; (Y.-C.W.); (W.-H.C.)
| | - Chien-Chih Wu
- Department of Urology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (M.-C.L.); (C.-C.W.)
- Department of Education and Humanities in Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Shauh-Der Yeh
- Department of Urology and Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
22
|
Oshima T, Nakase J, Toratani T, Numata H, Takata Y, Nakayama K, Tsuchiya H. A Scaffold-Free Allogeneic Construct From Adipose-Derived Stem Cells Regenerates an Osteochondral Defect in a Rabbit Model. Arthroscopy 2019; 35:583-593. [PMID: 30612760 DOI: 10.1016/j.arthro.2018.08.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE To determine whether an osteochondral defect could be healed histologically by implanting allogeneic 3-dimensionally formed adipose-derived stem cells (ADSCs) in a rabbit model. METHODS Thirty Japanese white rabbits (aged 15-17 weeks) were assigned to 1 of 2 groups. An osteochondral defect (diameter, 4.8 mm; depth, 3 mm) was created in the trochlear groove of the knee using a drill. The defects were left empty in the control group and were filled with cylindrical plugs of allogeneic ADSCs extracted from adipose tissue in the experimental group. Macroscopic scoring, histologic scoring, and immunohistologic stainability of type II collagen were evaluated at 4, 8, and 12 weeks postoperatively. RESULTS The macroscopic scores of the healing tissue in the experimental group were significantly greater than those in the control group at 12 weeks (P = .031). Histologically, safranin O staining was noted at 4 weeks and increased gradually over time in the experimental group. The modified International Cartilage Repair Society histologic score in the experimental group was significantly higher than that in the controls at 8 and 12 weeks (14 vs 9 at 8 weeks [P = .008], 18 vs 10 at 12 weeks [P = .007]). The implanted tissue was positive for type II collagen, and stainability increased gradually over time. CONCLUSIONS The 3-dimensional scaffold-free allogeneic ADSCs implanted into the osteochondral defect survived, adhered to the defect, increased the stainability of type II collagen gradually over time, and promoted histologic healing in a rabbit model. CLINICAL RELEVANCE ADSC implantation designed to promote osteochondral healing may play an important role in osteochondral healing.
Collapse
Affiliation(s)
- Takeshi Oshima
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Junsuke Nakase
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| | - Tatsuhiro Toratani
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hitoaki Numata
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yasushi Takata
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
23
|
Kargozar S, Mozafari M, Hamzehlou S, Brouki Milan P, Kim HW, Baino F. Bone Tissue Engineering Using Human Cells: A Comprehensive Review on Recent Trends, Current Prospects, and Recommendations. APPLIED SCIENCES 2019; 9:174. [DOI: 10.3390/app9010174] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of proper cells for bone tissue engineering remains a major challenge worldwide. Cells play a pivotal role in the repair and regeneration of the bone tissue in vitro and in vivo. Currently, a large number of differentiated (somatic) and undifferentiated (stem) cells have been used for bone reconstruction alone or in combination with different biomaterials and constructs (e.g., scaffolds). Although the results of the cell transplantation without any supporting or adjuvant material have been very effective with regard to bone healing. Recent advances in bone scaffolding are now becoming new players affecting the osteogenic potential of cells. In the present study, we have critically reviewed all the currently used cell sources for bone reconstruction and discussed the new horizons that are opening up in the context of cell-based bone tissue engineering strategies.
Collapse
Affiliation(s)
- Saeid Kargozar
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), Tehran 14155-4777, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran 144961-4535, Iran
| | - Sepideh Hamzehlou
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran 14155-6447, Iran
- Medical Genetics Network (MeGeNe), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran 144961-4535, Iran
| | - Hae-Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Korea
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan 31116, Korea
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
24
|
Liadi M, Collins A, Li Y, Li D. The Impact of Tissue Storage Conditions on Rat Olfactory Ensheathing Cell Yield and the Future Clinical Implications. Cell Transplant 2018; 27:1320-1327. [PMID: 30095003 PMCID: PMC6168985 DOI: 10.1177/0963689718787762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Trauma causes spinal cord injury, and the devastating consequences of the injury are due
to the failure of the damaged central nervous system (CNS) axons to regenerate. Previous
studies have shown that olfactory ensheathing cells (OECs) are a unique type of glial cell
and they can promote regeneration of CNS axons to aid recovery after spinal cord injury.
Transplantation of OECs, in particular from the olfactory bulb (OB), is considered one of
the most promising therapeutic strategies for the repair of CNS injuries, including spinal
cord injury. Transplantation of OECs can be autologous or allogenic. Here we focused on
the less invasive and more error-proof allograft approach which needs a collection of
donor OB tissue for OEC production. In this study, we investigated the effects on the
yield and proportions of OECs and olfactory nerve fibroblasts (ONFs) from storing OB
tissue in various media for periods of 24 and 48 hours. The OEC yield contributes to the
viability of a successful cell transplant. We concluded that storing OB tissue for a
period longer than 24 hours negatively impacted the total cell number and subsequently the
OEC population. This study provides useful information for future clinical
applications.
Collapse
Affiliation(s)
- Modinat Liadi
- 1 Department of Brain Repair and Rehabilitation, Spinal Repair Unit, UCL Institute of Neurology, Queen Square, London, UK
| | - Andrew Collins
- 1 Department of Brain Repair and Rehabilitation, Spinal Repair Unit, UCL Institute of Neurology, Queen Square, London, UK
| | - Ying Li
- 1 Department of Brain Repair and Rehabilitation, Spinal Repair Unit, UCL Institute of Neurology, Queen Square, London, UK
| | - Daqing Li
- 1 Department of Brain Repair and Rehabilitation, Spinal Repair Unit, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
25
|
Marei MK, El Backly RM. Dental Mesenchymal Stem Cell-Based Translational Regenerative Dentistry: From Artificial to Biological Replacement. Front Bioeng Biotechnol 2018; 6:49. [PMID: 29770323 PMCID: PMC5941981 DOI: 10.3389/fbioe.2018.00049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Dentistry is a continuously changing field that has witnessed much advancement in the past century. Prosthodontics is that branch of dentistry that deals with replacing missing teeth using either fixed or removable appliances in an attempt to simulate natural tooth function. Although such "replacement therapies" appear to be easy and economic they fall short of ever coming close to their natural counterparts. Complications that arise often lead to failures and frequent repairs of such devices which seldom allow true physiological function of dental and oral-maxillofacial tissues. Such factors can critically affect the quality of life of an individual. The market for dental implants is continuously growing with huge economic revenues. Unfortunately, such treatments are again associated with frequent problems such as peri-implantitis resulting in an eventual loss or replacement of implants. This is particularly influential for patients having co-morbid diseases such as diabetes or osteoporosis and in association with smoking and other conditions that undoubtedly affect the final treatment outcome. The advent of tissue engineering and regenerative medicine therapies along with the enormous strides taken in their associated interdisciplinary fields such as stem cell therapy, biomaterial development, and others may open arenas to enhancing tissue regeneration via designing and construction of patient-specific biological and/or biomimetic substitutes. This review will overview current strategies in regenerative dentistry while overviewing key roles of dental mesenchymal stem cells particularly those of the dental pulp, until paving the way to precision/translational regenerative medicine therapies for future clinical use.
Collapse
Affiliation(s)
- Mona K Marei
- Department of Removable Prosthodontics, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.,Tissue Engineering Laboratories, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Rania M El Backly
- Tissue Engineering Laboratories, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.,Endodontics, Conservative Dentistry Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| |
Collapse
|
26
|
Lewis FC, Cottle BJ, Shone V, Marazzi G, Sassoon D, Tseng CCS, Dankers PYW, Chamuleau SAJ, Nadal-Ginard B, Ellison-Hughes GM. Transplantation of Allogeneic PW1 pos/Pax7 neg Interstitial Cells Enhance Endogenous Repair of Injured Porcine Skeletal Muscle. ACTA ACUST UNITED AC 2017; 2:717-736. [PMID: 30062184 PMCID: PMC6059014 DOI: 10.1016/j.jacbts.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 01/07/2023]
Abstract
Allogeneic PICs express and secrete an array of pro-regenerative paracrine factors that stimulate a regenerative response in a preclinical muscle injury model applicable to humans. Paracrine factors secreted by allogeneic PICs stimulate endogenous progenitor cell activation and differentiation, leading to accelerated and improved myofiber regeneration and microvessel formation. Allogeneic PICs survive long enough to exert their action before being cleared by the host immune system. Therefore, the cells transplanted are allogeneic but the regeneration is completely autologous. Administration of HGF and IGF-1 improves skeletal muscle regeneration, but not to the same extent as PIC transplantation.
Skeletal muscle-derived PW1pos/Pax7neg interstitial cells (PICs) express and secrete a multitude of proregenerative growth factors and cytokines. Utilizing a porcine preclinical skeletal muscle injury model, delivery of allogeneic porcine PICs (pPICs) significantly improved and accelerated myofiber regeneration and neocapillarization, compared with saline vehicle control-treated muscles. Allogeneic pPICs did not contribute to new myofibers or capillaries and were eliminated by the host immune system. In conclusion, allogeneic pPIC transplantation stimulated the endogenous stem cell pool to bring about enhanced autologous skeletal muscle repair and regeneration. This allogeneic cell approach is considered a cost-effective, easy to apply, and readily available regenerative therapeutic strategy.
Collapse
Key Words
- BrdU, 5-bromo-2′-deoxyuridine
- CM, pPIC conditioned medium
- CSA, cross sectional area
- CSC, cardiac stem cell
- CTRL, control
- CTX, cardiotoxin
- DAPI, 4′,6-diamidino-2-phenylindole
- DMEM, Dulbecco’s Modified Eagle's medium
- FBS, fetal bovine serum
- GFPpPIC, GFP-positive porcine PW1pos/Pax7neg interstitial cell
- GM, growth medium
- HUVEC, human umbilical vein endothelial cell
- HVG, hematoxylin and van Gieson
- ICM, heat-inactivated conditioned medium
- IV, intravenous
- MHC, myosin heavy chain
- MI, myocardial infarction
- P, passage
- PBMC, peripheral blood mononuclear cell
- PBS, phosphate buffered saline
- PIC, PW1pos/Pax7neg interstitial cell
- PICs
- TA, tibialis anterior
- UM, unconditioned medium
- allogeneic progenitor cells
- growth factors
- nMHC, neonatal myosin heavy chain
- pPIC, porcine PW1pos/Pax7neg interstitial cell
- porcine preclinical model
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
- regeneration
- skeletal muscle
- vWF, Von Willebrand factor
Collapse
Affiliation(s)
- Fiona C Lewis
- School of Basic & Medical Biosciences, Centre of Human & Aerospace Physiological Sciences & Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London, United Kingdom
| | - Beverley J Cottle
- School of Basic & Medical Biosciences, Centre of Human & Aerospace Physiological Sciences & Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London, United Kingdom
| | - Victoria Shone
- School of Basic & Medical Biosciences, Centre of Human & Aerospace Physiological Sciences & Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London, United Kingdom
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine UMRS 1166, Institute of Cardiometabolism and Nutrition, Université de Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - David Sassoon
- Stem Cells and Regenerative Medicine UMRS 1166, Institute of Cardiometabolism and Nutrition, Université de Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Cheyenne C S Tseng
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Patricia Y W Dankers
- Supramolecular Biomaterials for Translational Biomedical Science, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Steven A J Chamuleau
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bernardo Nadal-Ginard
- School of Basic & Medical Biosciences, Centre of Human & Aerospace Physiological Sciences & Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London, United Kingdom
| | - Georgina M Ellison-Hughes
- School of Basic & Medical Biosciences, Centre of Human & Aerospace Physiological Sciences & Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London, United Kingdom
| |
Collapse
|
27
|
Bayat M, Jalalifirouzkouhi A. Presenting a Method to Improve Bone Quality Through Stimulation of Osteoporotic Mesenchymal Stem Cells by Low-Level Laser Therapy. Photomed Laser Surg 2017; 35:622-628. [PMID: 28621568 DOI: 10.1089/pho.2016.4245] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE This review aims to present a method to improve bone quality through stimulation of osteoporotic mesenchymal stem cells (MSCs) by low-level laser therapy (LLLT). BACKGROUND Osteoporosis (OP) is characterized by decreased bone mass and bone strength, which results in an increased incidence of bone fractures. These fractures often lead to additional disability and mortality. Osteoporotic MSCs have reduced osteogenic differentiation when cultured in their standard differentiation media. LLLT has a biostimulatory effect on fibroblasts and osteoblasts. MSCs have the ability to generate cells of connective tissue lineages, which includes the bones. Recently, transplantation of in vitro cultured bone marrow (BM) MSCs into sites at risk for development of osteoporotic bone has resulted in improved bone structure. METHODS Comprehensive research was performed using PubMed, and biostimulatory effect of LLLT on bony cells and MSCs were studied. RESULTS LLLT can stimulate growth, proliferation, and differentiation of SCs in vitro and in vivo. This ability of LLLT is an essential prerequisite for performing experiments related to disease control in humans. Thus, laser-treated osteoporotic autologous BMMSCs may represent a promising therapeutic method to protect the bones in patients with OP and prevent fractures in these patients. Therefore, researchers hypothesize that transplantation of in vitro laser-treated autologous cultured osteoporotic BMMSCs that have the appropriate osteogenic phenotype into sites at risk for development of osteoporotic bone may result in improved bone structure. In this respect, investigators have successfully used LLLT to restore autologous osteoporotic MSCs in vitro. Subsequently, these cells have been differentiated into osteoblast cell lines with the use of laser treatment after which they were transplanted into osteoporotic animal models. CONCLUSIONS This technique might improve bone quality and structure. However, additional research must be undertaken to understand the underlying mechanisms of this treatment, validate its effectiveness, and assess the feasibility for clinical application of LLLT to treat MSCs in regeneration of osteoporotic bone.
Collapse
Affiliation(s)
- Mohammad Bayat
- 1 Cellular and Molecular Biology Research Center, and Biology and Anatomical Sciences Department, Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | | |
Collapse
|
28
|
Abbasalizadeh S, Pakzad M, Cabral JMS, Baharvand H. Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin Biol Ther 2017; 17:1201-1219. [PMID: 28699788 DOI: 10.1080/14712598.2017.1354982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Currently, promising outcomes from clinical trials of allogeneic cells, especially allogeneic mesenchymal stromal cells, fibroblasts, keratinocytes, and human cardiac stem cells, have encouraged research institutions, small and medium enterprises (SMEs), and big pharmaceutical companies to invest and focus on developing allogeneic cell therapy products. Commercial and large-scale production of allogeneic cell therapy products requires unique capabilities to develop technologies that generate safe and effective allogeneic cells/cell lines and their fully characterized master/working banks. In addition, it is necessary to design robust upstream and downstream manufacturing processes, and establish integrated, well-designed manufacturing facilities to produce high quality affordable products in accordance with current GMP regulations for the production of cell therapy products. Areas covered: The authors highlight: the recent advances in the development of allogeneic products, the available options to develop robust manufacturing processes, and facility design considerations. Expert opinion: Currently, there are multiple challenges in development of allogeneic cell therapy products. Indeed, the field is still in its infancy; with technologies and regulations still under development, as is our understanding of the mechanisms of action in the body and their interaction with the host immune system. Their characterization and testing is also an emerging and very complex area.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Mohammad Pakzad
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Joaquim M S Cabral
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Hossein Baharvand
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- c Department of Developmental Biology , University of Science and Culture , Tehran , Iran
| |
Collapse
|
29
|
Maartens JH, De-Juan-Pardo E, Wunner FM, Simula A, Voelcker NH, Barry SC, Hutmacher DW. Challenges and opportunities in the manufacture and expansion of cells for therapy. Expert Opin Biol Ther 2017; 17:1221-1233. [DOI: 10.1080/14712598.2017.1360273] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Joachim H. Maartens
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Elena De-Juan-Pardo
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Felix M. Wunner
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Antonio Simula
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Nicolas H. Voelcker
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Simon C. Barry
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
- Molecular Immunology, Department of Gastroenterology, Women’s and Children’s Hospital, Adelaide, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Dietmar W. Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
- ARC Centre in Additive Biomanufacturing, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
30
|
Corbett MS, Webster A, Hawkins R, Woolacott N. Innovative regenerative medicines in the EU: a better future in evidence? BMC Med 2017; 15:49. [PMID: 28270209 PMCID: PMC5341436 DOI: 10.1186/s12916-017-0818-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/14/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite a steady stream of headlines suggesting they will transform the future of healthcare, high-tech regenerative medicines have, to date, been quite inaccessible to patients, with only eight having been granted an EU marketing licence in the last 7 years. Here, we outline some of the historical reasons for this paucity of licensed innovative regenerative medicines. We discuss the challenges to be overcome to expedite the development of this complex and rapidly changing area of medicine, together with possible reasons to be more optimistic for the future. DISCUSSION Several factors have contributed to the scarcity of cutting-edge regenerative medicines in clinical practice. These include the great expense and difficulties involved in planning how individual therapies will be developed, manufactured to commercial levels and ultimately successfully delivered to patients. Specific challenges also exist when evaluating the safety, efficacy and cost-effectiveness of these therapies. Furthermore, many treatments are used without a licence from the European Medicines Agency, under "Hospital Exemption" from the EC legislation. For products which are licensed, alternative financing approaches by healthcare providers may be needed, since many therapies will have significant up-front costs but uncertain benefits and harms in the long-term. However, increasing political interest and more flexible mechanisms for licensing and financing of therapies are now evident; these could be key to the future growth and development of regenerative medicine in clinical practice. CONCLUSIONS Recent developments in regulatory processes, coupled with increasing political interest, may offer some hope for improvements to the long and often difficult routes from laboratory to marketplace for leading-edge cell or tissue therapies. Collaboration between publicly-funded researchers and the pharmaceutical industry could be key to the future development of regenerative medicine in clinical practice; such collaborations might also offer a possible antidote to the innovation crisis in the pharmaceutical industry.
Collapse
Affiliation(s)
- Mark S Corbett
- Centre for Reviews and Dissemination, University of York, Heslington, York, YO10 5DD, UK.
| | - Andrew Webster
- Science and Technology Studies Unit, Department of Sociology, University of York, Heslington, York, YO10 5DD, UK
| | - Robert Hawkins
- Medical Oncology, The Christie Hospital and University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | - Nerys Woolacott
- Centre for Reviews and Dissemination, University of York, Heslington, York, YO10 5DD, UK
| |
Collapse
|
31
|
Noiseux N, Mansour S, Weisel R, Stevens LM, Der Sarkissian S, Tsang K, Crean AM, Larose E, Li SH, Wintersperger B, Vu MQ, Prieto I, Li RK, Roy DC, Yau TM. The IMPACT-CABG trial: A multicenter, randomized clinical trial of CD133+ stem cell therapy during coronary artery bypass grafting for ischemic cardiomyopathy. J Thorac Cardiovasc Surg 2016; 152:1582-1588.e2. [DOI: 10.1016/j.jtcvs.2016.07.067] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/15/2016] [Accepted: 07/01/2016] [Indexed: 11/26/2022]
|
32
|
Abstract
The intrinsic regenerative capacity of avascular cartilage is limited. Cartilage injuries result in chronic, non-healing lesions requiring surgical management. Frequently, these surgical techniques make use of allogeneic cells and tissues. This review discusses the immune status of these materials. Cartilage allografts, often used in orthopedic and plastic surgeries, have rarely provoked a significant immune response. In whole cartilage transplants, the dense matrix produced by chondrocytes inhibits lymphocyte migration, preventing immune detection rendering them "antigen sequestered." It is unclear whether isolated chondrocytes are immune-privileged; chondrocytes express immune inhibitory B7 molecules, indicating that they have some ability to modulate immune reactions. Allogeneic cartilage grafts often involve a bony portion often retaining immunogenic cells and proteins-to facilitate good surgical attachment and concern that this may enhance inflammation and immune rejection. However, studies of failed cartilage grafts have not found immune responses to be a contributing factor. Meniscus allografts, which also retain a bony portion, raise similar concerns as cartilage allografts. Despite this, the plugs improved patient outcomes, indicating that the immunological effects were not clinically significant. Finally, allogeneic mesenchymal stromal cells (MSCs) also are being investigated as a treatment for cartilage damage. MSCs have been demonstrated to have unique immunomodulatory properties including their ability to reduce immune cell infiltration and to modulate inflammation. In summary, the immunogenic properties of cartilage vary with the type of allograft used: Cartilage allografts demonstrate active immune-suppressive mechanisms as evidenced by lack of allograft rejection, while MSC allografts appear to be safe for transplantation.
Collapse
Affiliation(s)
- Benjamin Smith
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Ian R Sigal
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Daniel A Grande
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA. .,Department of Orthopaedic Surgery, North Shore/LIJ Health System, Manhasset, NY, 11030, USA.
| |
Collapse
|
33
|
Archibald PRT, Williams DJ. Using the cost–effectiveness of allogeneic islet transplantation to inform induced pluripotent stem cell-derived β-cell therapy reimbursement. Regen Med 2015; 10:959-73. [DOI: 10.2217/rme.15.59] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aims: In the present study a cost–effectiveness analysis of allogeneic islet transplantation was performed and the financial feasibility of a human induced pluripotent stem cell-derived β-cell therapy was explored. Methods: Previously published cost and health benefit data for islet transplantation were utilized to perform the cost–effectiveness and sensitivity analyses. Results & conclusion: It was determined that, over a 9-year time horizon, islet transplantation would become cost saving and ‘dominate’ the comparator. Over a 20-year time horizon, islet transplantation would incur significant cost savings over the comparator (GB£59,000). Finally, assuming a similar cost of goods to islet transplantation and a lack of requirement for immunosuppression, a human induced pluripotent stem cell-derived β-cell therapy would dominate the comparator over an 8-year time horizon.
Collapse
Affiliation(s)
- Peter RT Archibald
- Centre for Biological Engineering, Loughborough University, Loughborough, UK
| | - David J Williams
- Centre for Biological Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
34
|
Heathman TRJ, Nienow AW, McCall MJ, Coopman K, Kara B, Hewitt CJ. The translation of cell-based therapies: clinical landscape and manufacturing challenges. Regen Med 2015; 10:49-64. [PMID: 25562352 DOI: 10.2217/rme.14.73] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapies have the potential to make a large contribution toward currently unmet patient need and thus effective manufacture of these products is essential. Many challenges must be overcome before this can become a reality and a better definition of the manufacturing requirements for cell-based products must be obtained. The aim of this study is to inform industry and academia of current cell-based therapy clinical development and to identify gaps in their manufacturing requirements. A total of 1342 active cell-based therapy clinical trials have been identified and characterized based on cell type, target indication and trial phase. Multiple technologies have been assessed for the manufacture of these cell types in order to facilitate product translation and future process development.
Collapse
Affiliation(s)
- Thomas R J Heathman
- Centre for Biological Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | | | | | | | | | | |
Collapse
|
35
|
Berner A, Henkel J, Woodruff MA, Steck R, Nerlich M, Schuetz MA, Hutmacher DW. Delayed minimally invasive injection of allogenic bone marrow stromal cell sheets regenerates large bone defects in an ovine preclinical animal model. Stem Cells Transl Med 2015; 4:503-12. [PMID: 25834121 DOI: 10.5966/sctm.2014-0244] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/28/2015] [Indexed: 12/23/2022] Open
Abstract
Cell-based tissue engineering approaches are promising strategies in the field of regenerative medicine. However, the mode of cell delivery is still a concern and needs to be significantly improved. Scaffolds and/or matrices loaded with cells are often transplanted into a bone defect immediately after the defect has been created. At this point, the nutrient and oxygen supply is low and the inflammatory cascade is incited, thus creating a highly unfavorable microenvironment for transplanted cells to survive and participate in the regeneration process. We therefore developed a unique treatment concept using the delayed injection of allogenic bone marrow stromal cell (BMSC) sheets to regenerate a critical-sized tibial defect in sheep to study the effect of the cells' regeneration potential when introduced at a postinflammatory stage. Minimally invasive percutaneous injection of allogenic BMSCs into biodegradable composite scaffolds 4 weeks after the defect surgery led to significantly improved bone regeneration compared with preseeded scaffold/cell constructs and scaffold-only groups. Biomechanical testing and microcomputed tomography showed comparable results to the clinical reference standard (i.e., an autologous bone graft). To our knowledge, we are the first to show in a validated preclinical large animal model that delayed allogenic cell transplantation can provide applicable clinical treatment alternatives for challenging bone defects in the future.
Collapse
Affiliation(s)
- Arne Berner
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| | - Jan Henkel
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| | - Maria A Woodruff
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| | - Roland Steck
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| | - Michael Nerlich
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| | - Michael A Schuetz
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation and Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Queensland, Australia; Department of Trauma Surgery, University of Regensburg, Regensburg, Germany
| |
Collapse
|
36
|
Sourcing of an alternative pericyte-like cell type from peripheral blood in clinically relevant numbers for therapeutic angiogenic applications. Mol Ther 2014; 23:510-22. [PMID: 25582709 DOI: 10.1038/mt.2014.232] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/26/2014] [Indexed: 02/06/2023] Open
Abstract
Autologous cells hold great potential for personalized cell therapy, reducing immunological and risk of infections. However, low cell counts at harvest with subsequently long expansion times with associated cell function loss currently impede the advancement of autologous cell therapy approaches. Here, we aimed to source clinically relevant numbers of proangiogenic cells from an easy accessible cell source, namely peripheral blood. Using macromolecular crowding (MMC) as a biotechnological platform, we derived a novel cell type from peripheral blood that is generated within 5 days in large numbers (10-40 million cells per 100 ml of blood). This blood-derived angiogenic cell (BDAC) type is of monocytic origin, but exhibits pericyte markers PDGFR-β and NG2 and demonstrates strong angiogenic activity, hitherto ascribed only to MSC-like pericytes. Our findings suggest that BDACs represent an alternative pericyte-like cell population of hematopoietic origin that is involved in promoting early stages of microvasculature formation. As a proof of principle of BDAC efficacy in an ischemic disease model, BDAC injection rescued affected tissues in a murine hind limb ischemia model by accelerating and enhancing revascularization. Derived from a renewable tissue that is easy to collect, BDACs overcome current short-comings of autologous cell therapy, in particular for tissue repair strategies.
Collapse
|
37
|
Mittra J, Tait J, Mastroeni M, Turner ML, Mountford JC, Bruce K. Identifying viable regulatory and innovation pathways for regenerative medicine: a case study of cultured red blood cells. N Biotechnol 2014; 32:180-90. [PMID: 25094050 DOI: 10.1016/j.nbt.2014.07.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 07/16/2014] [Accepted: 07/16/2014] [Indexed: 11/25/2022]
Abstract
The creation of red blood cells for the blood transfusion markets represents a highly innovative application of regenerative medicine with a medium term (5-10 year) prospect for first clinical studies. This article describes a case study analysis of a project to derive red blood cells from human embryonic stem cells, including the systemic challenges arising from (i) the selection of appropriate and viable regulatory protocols and (ii) technological constraints related to stem cell manufacture and scale up to clinical Good Manufacturing Practice (GMP) standard. The method used for case study analysis (Analysis of Life Science Innovation Systems (ALSIS)) is also innovative, demonstrating a new approach to social and natural science collaboration to foresight product development pathways. Issues arising along the development pathway include cell manufacture and scale-up challenges, affected by regulatory demands emerging from the innovation ecosystem (preclinical testing and clinical trials). Our discussion reflects on the efforts being made by regulators to adapt the current pharmaceuticals-based regulatory model to an allogeneic regenerative medicine product and the broader lessons from this case study for successful innovation and translation of regenerative medicine therapies, including the role of methodological and regulatory innovation in future development in the field.
Collapse
Affiliation(s)
- J Mittra
- ESRC Innogen Centre, Old Surgeons' Hall, High School Yards, University of Edinburgh, UK.
| | - J Tait
- ESRC Innogen Centre, Old Surgeons' Hall, High School Yards, University of Edinburgh, UK
| | - M Mastroeni
- ESRC Innogen Centre, Old Surgeons' Hall, High School Yards, University of Edinburgh, UK
| | - M L Turner
- Scottish National Blood Transfusion Service, UK; Roslin Cells Ltd., UK
| | - J C Mountford
- Scottish National Blood Transfusion Service, UK; Institute of Cardiovascular and Medical Services, University of Glasgow, UK
| | | |
Collapse
|
38
|
Henkel J, Woodruff MA, Epari DR, Steck R, Glatt V, Dickinson IC, Choong PFM, Schuetz MA, Hutmacher DW. Bone Regeneration Based on Tissue Engineering Conceptions - A 21st Century Perspective. Bone Res 2013; 1:216-48. [PMID: 26273505 PMCID: PMC4472104 DOI: 10.4248/br201303002] [Citation(s) in RCA: 511] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/20/2013] [Indexed: 12/18/2022] Open
Abstract
The role of Bone Tissue Engineering in the field of Regenerative Medicine has been the topic of substantial research over the past two decades. Technological advances have improved orthopaedic implants and surgical techniques for bone reconstruction. However, improvements in surgical techniques to reconstruct bone have been limited by the paucity of autologous materials available and donor site morbidity. Recent advances in the development of biomaterials have provided attractive alternatives to bone grafting expanding the surgical options for restoring the form and function of injured bone. Specifically, novel bioactive (second generation) biomaterials have been developed that are characterised by controlled action and reaction to the host tissue environment, whilst exhibiting controlled chemical breakdown and resorption with an ultimate replacement by regenerating tissue. Future generations of biomaterials (third generation) are designed to be not only osteoconductive but also osteoinductive, i.e. to stimulate regeneration of host tissues by combining tissue engineering and in situ tissue regeneration methods with a focus on novel applications. These techniques will lead to novel possibilities for tissue regeneration and repair. At present, tissue engineered constructs that may find future use as bone grafts for complex skeletal defects, whether from post-traumatic, degenerative, neoplastic or congenital/developmental "origin" require osseous reconstruction to ensure structural and functional integrity. Engineering functional bone using combinations of cells, scaffolds and bioactive factors is a promising strategy and a particular feature for future development in the area of hybrid materials which are able to exhibit suitable biomimetic and mechanical properties. This review will discuss the state of the art in this field and what we can expect from future generations of bone regeneration concepts.
Collapse
Affiliation(s)
- Jan Henkel
- Institute of Health & Biomedical Innovation, Queensland University of Technology , Brisbane, Queensland, Australia
| | - Maria A Woodruff
- Institute of Health & Biomedical Innovation, Queensland University of Technology , Brisbane, Queensland, Australia
| | - Devakara R Epari
- Institute of Health & Biomedical Innovation, Queensland University of Technology , Brisbane, Queensland, Australia
| | - Roland Steck
- Institute of Health & Biomedical Innovation, Queensland University of Technology , Brisbane, Queensland, Australia
| | - Vaida Glatt
- Institute of Health & Biomedical Innovation, Queensland University of Technology , Brisbane, Queensland, Australia
| | - Ian C Dickinson
- Orthopaedic Oncology Service, Princess Alexandra Hospital , Brisbane, Australia
| | - Peter F M Choong
- Department of Surgery, University of Melbourne, St. Vincent's Hospital , Melbourne, Australia ; Department of Orthopaedics, St. Vincent's Hospital , Melbourne, Australia ; Bone and Soft Tissue Sarcoma Service, Peter MacCallum Cancer Centre , Melbourne, Australia
| | - Michael A Schuetz
- Institute of Health & Biomedical Innovation, Queensland University of Technology , Brisbane, Queensland, Australia ; Orthopaedic and Trauma Services, Princess Alexandra Hospital , Brisbane, Australia
| | - Dietmar W Hutmacher
- Orthopaedic Oncology Service, Princess Alexandra Hospital , Brisbane, Australia ; George W Woodruff School of Mechanical Engineering, Georgia Institute of Technology , Atlanta, GA, USA
| |
Collapse
|
39
|
Philippart P, Meuleman N, Stamatopoulos B, Najar M, Pieters K, De Bruyn C, Bron D, Lagneaux L. In vivo production of mesenchymal stromal cells after injection of autologous platelet-rich plasma activated by recombinant human soluble tissue factor in the bone marrow of healthy volunteers. Tissue Eng Part A 2013; 20:160-70. [PMID: 23924315 DOI: 10.1089/ten.tea.2013.0244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Autologous mesenchymal stromal cell (MSC)-based therapies offer one of the most promising and safe methods for regeneration or reconstruction of tissues and organs. Routine procedures to obtain adequate amount of autologous stem cells need their expansion through culture, with risks of contamination and cell differentiation, leading to the loss of cell ability for therapies. We suggest the use of human bone marrow (BM) as a physiological bioreactor to produce autologous MSC by injection of autologous platelet-rich plasma activated by recombinant human soluble tissue factor (rhsTF) in iliac crest. A trial on 13 healthy volunteers showed the feasibility and harmlessness of the procedure. The phenotype and cellularity of BM cells were not modified, on day 3 after injection. Endothelial progenitor cells (EPC) were mobilized to the bloodstream, without stimulation of hematopoietic stem cells (HSC). MSC level in BM increased with a specific commitment to preosteoblastic cell population both in vivo and in vitro. This self-stimulation system of BM seems thus to be a promising feasible process 3 days before clinical cell therapy applications.
Collapse
Affiliation(s)
- Pierre Philippart
- 1 Department of Stomatology and Maxillo-Facial Surgery, HIS Site Bracops , Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kuratnik A, Giardina C. Intestinal organoids as tissue surrogates for toxicological and pharmacological studies. Biochem Pharmacol 2013; 85:1721-6. [PMID: 23623789 DOI: 10.1016/j.bcp.2013.04.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 04/17/2013] [Accepted: 04/18/2013] [Indexed: 12/12/2022]
Abstract
Recently developed cell culture protocols have allowed for the derivation of multi-cellular structures dubbed intestinal "organoids" from embryonic stem cells (ESCs), induced pluripotent stem cells (IPSCs), and adult intestinal stem cells (ISCs). These structures resemble in vivo intestinal crypts, both in structure and developmental processes, and can be grown quickly and in relatively large quantities. Although much research has focused on developing intestinal organoids for tissue repair, more immediate applications include high-throughput screening for agents that target intestinal epithelium. Here we describe current methods for deriving mouse and human intestinal organoids and discuss some applications aimed at developing novel therapies or preventive agents for diseases of the lower GI tract such as inflammatory bowel diseases and colorectal cancer.
Collapse
Affiliation(s)
- Anton Kuratnik
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, U3125, Storrs, CT 06269, USA.
| | | |
Collapse
|
41
|
Raschzok N, Langer CM, Schmidt C, Lerche KH, Billecke N, Nehls K, Schlüter NB, Leder A, Rohn S, Mogl MT, Lüdemann L, Stelter L, Teichgräber UK, Neuhaus P, Sauer IM. Functionalizable silica-based micron-sized iron oxide particles for cellular magnetic resonance imaging. Cell Transplant 2013; 22:1959-70. [PMID: 23294541 DOI: 10.3727/096368912x661382] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cellular therapies require methods for noninvasive visualization of transplanted cells. Micron-sized iron oxide particles (MPIOs) generate a strong contrast in magnetic resonance imaging (MRI) and are therefore ideally suited as an intracellular contrast agent to image cells under clinical conditions. However, MPIOs were previously not applicable for clinical use. Here, we present the development and evaluation of silica-based micron-sized iron oxide particles (sMPIOs) with a functionalizable particle surface. Particles with magnetite content of >40% were composed using the sol-gel process. The particle surfaces were covered with COOH groups. Fluorescein, poly-L-lysine (PLL), and streptavidin (SA) were covalently attached. Monodisperse sMPIOs had an average size of 1.18 µm and an iron content of about 1.0 pg Fe/particle. Particle uptake, toxicity, and imaging studies were performed using HuH7 cells and human and rat hepatocytes. sMPIOs enabled rapid cellular labeling within 4 h of incubation; PLL-modified particles had the highest uptake. In T2*-weighted 3.0 T MRI, the detection threshold in agarose was 1,000 labeled cells, whereas in T1-weighted LAVA sequences, at least 10,000 cells were necessary to induce sufficient contrast. Labeling was stable and had no adverse effects on labeled cells. Silica is a biocompatible material that has been approved for clinical use. sMPIOs could therefore be suitable for future clinical applications in cellular MRI, especially in settings that require strong cellular contrast. Moreover, the particle surface provides the opportunity to create multifunctional particles for targeted delivery and diagnostics.
Collapse
Affiliation(s)
- Nathanael Raschzok
- General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Henkel J, Hutmacher DW. Design and fabrication of scaffold-based tissue engineering. ACTA ACUST UNITED AC 2013. [DOI: 10.1515/bnm-2013-0021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
43
|
Bercu MM, Arien-Zakay H, Stoler D, Lecht S, Lelkes PI, Samuel S, Or R, Nagler A, Lazarovici P, Elchalal U. Enhanced survival and neurite network formation of human umbilical cord blood neuronal progenitors in three-dimensional collagen constructs. J Mol Neurosci 2012; 51:249-61. [PMID: 23233347 DOI: 10.1007/s12031-012-9933-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 11/28/2012] [Indexed: 02/07/2023]
Abstract
Umbilical cord blood (CB) stem cells have been proposed for cell-based therapeutic applications for diverse diseases of the CNS. We hypothesized that tissue-engineering strategies may extend the efficacy of these approaches by improving the long-term viability and function of stem cell-derived neuronal progenitors. To test our hypothesis, we explored the survival and differentiation of human CB-derived neuronal progenitors (HUCBNP) in a three-dimensional (3D) collagen construct. In contrast to two-dimensional culture conditions, the cells survived in 3D for an extended period of time of more than 2 months. Under 3D conditions, HUCBNP underwent spontaneous neuronal differentiation, which was further enhanced by treatment with neuronal conditioned medium (CM) and nerve growth factor (NGF). Neurite outgrowth, quantified by assessing the fractal dimension (D f) of the complex neuronal networks, was significantly enhanced under 3D conditions in the presence of CM/NGF, concomitant with a reduced expression of the early neuronal marker nestin (1.9-fold), and increased levels of mature neuronal markers such as MAP-2 (3.6-fold), β-tubulin (1.5-fold), and neuronal specific enolase (6.6-fold) and the appearance of the synaptic marker synaptophysin. To assess the feasibility for clinical usage, HUCBNP were also isolated from frozen CB samples and cultured under 3D conditions. The data indicate the essential complete preservation of neurotrophic (survival) and neurotropic (neurite outgrowth) properties. In conclusion, 3D culture conditions are proposed as an essential step for both maintenance of CB neuronal progenitors in vitro and for investigating specific features of neuronal differentiation towards future use in regenerative therapy.
Collapse
Affiliation(s)
- Marian M Bercu
- School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Smith DM. Assessing commercial opportunities for autologous and allogeneic cell-based products. Regen Med 2012; 7:721-32. [DOI: 10.2217/rme.12.40] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The two primary cell sources used to produce cell-based therapies are autologous (self-derived) and allogeneic (derived from a donor). This analysis attempts to compare and contrast the two approaches in order to understand whether there is an emerging preference in the market. While the current clinical trials underway are slightly biased to autologous approaches, it is clear that both cell-based approaches are being aggressively pursued. This analysis also breaks down the commercial advantages of each cell-based approach, comparing both cost of goods and the ideal indication type for each. While allogeneic therapies have considerable advantages over autologous therapies, they do have a distinct disadvantage regarding potential immunogenicity. The introduction of the hybrid autologous business model provides the ability for autologous-based therapies to mitigate some of the advantages that allogeneic cell-based therapies enjoy, including cost of goods. Finally, two case studies are presented that demonstrate that there is sufficient space for both autologous and allogeneic cell-based therapies within a single disease area.
Collapse
Affiliation(s)
- Devyn M Smith
- Pfizer Worldwide R&D, The Portway Building, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| |
Collapse
|
45
|
Abstract
We identify three dimensions with which to classify heuristically the routes to widespread adoption of cellular therapies. The first dimension is based on the relative involvement of clinicians and companies in a particular cellular therapy. The second dimension is based on cell type and consequent scale of manufacture. The third dimension classifies the therapeutic intervention as a procedure or product and has perhaps received less attention. We suggest that for those cellular therapies that require therapeutic procedures, close collaboration between companies and clinicians will reduce the time to widespread adoption. For selected cellular therapies we make predictions of the likely time to widespread adoption.
Collapse
Affiliation(s)
- Lucy Foley
- Newcastle University Business School, Citywall, Citygate, Newcastle Upon Tyne, United Kingdom
| | | |
Collapse
|
46
|
Nonhuman primate induced pluripotent stem cells in regenerative medicine. Stem Cells Int 2012; 2012:767195. [PMID: 22577396 PMCID: PMC3345260 DOI: 10.1155/2012/767195] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 01/27/2012] [Indexed: 01/01/2023] Open
Abstract
Among the various species from which induced pluripotent stem cells have been derived, nonhuman primates (NHPs) have a unique role as preclinical models. Their relatedness to humans and similar physiology, including central nervous system, make them ideal for translational studies. We review here the progress made in deriving and characterizing iPS cell lines from different NHP species. We focus on iPS cell lines from the marmoset, a small NHP in which several human disease states can be modeled. The marmoset can serve as a model for the implementation of patient-specific autologous cell therapy in regenerative medicine.
Collapse
|
47
|
Duan B, Wang M. Customized Ca-P/PHBV nanocomposite scaffolds for bone tissue engineering: design, fabrication, surface modification and sustained release of growth factor. J R Soc Interface 2010; 7 Suppl 5:S615-29. [PMID: 20504805 DOI: 10.1098/rsif.2010.0127.focus] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Integrating an advanced manufacturing technique, nanocomposite material and controlled delivery of growth factor to form multifunctional tissue engineering scaffolds was investigated in this study. Based on calcium phosphate (Ca-P)/poly(hydroxybutyrate-co-hydroxyvalerate) (PHBV) nanocomposite microspheres, three-dimensional Ca-P/PHBV nanocomposite scaffolds with customized architecture, controlled porosity and totally interconnected porous structure were successfully fabricated using selective laser sintering (SLS), one of the rapid prototyping technologies. The cytocompatibility of sintered Ca-P/PHBV nanocomposite scaffolds, as well as PHBV polymer scaffolds, was studied. For surface modification of nanocomposite scaffolds, gelatin was firstly physically entrapped onto the scaffold surface and heparin was subsequently immobilized on entrapped gelatin. The surface-modification improved the wettability of scaffolds and provided specific binding site between conjugated heparin and the growth factor recombinant human bone morphogenetic protein-2 (rhBMP-2). The surface-modified Ca-P/PHBV nanocomposite scaffolds loaded with rhBMP-2 significantly enhanced the alkaline phosphatase activity and osteogenic differentiation markers in gene expression of C3H10T1/2 mesenchymal stem cells. Together with osteoconductive nanocomposite material and controlled growth factor delivery strategies, the use of SLS technique to form complex scaffolds will provide a promising route towards individualized bone tissue regeneration.
Collapse
Affiliation(s)
- Bin Duan
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong
| | | |
Collapse
|
48
|
Mason C, Manzotti E. Regen: the industry responsible for cell-based therapies. Regen Med 2009; 4:783-5. [DOI: 10.2217/rme.09.72] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Chris Mason
- Advanced Centre for Biochemical Engineering, University College London, Roberts Building, Torrington Place, London, WC1E 7JE, UK
| | - Elisa Manzotti
- Future Medicine Ltd, Unitec House, 2 Albert Place, Finchley Central, London, N3 1QB, UK
| |
Collapse
|