1
|
Song SJ, Nam Y, Rim YA, Ju JH, Sohn Y. Comparative analysis of regulations and studies on stem cell therapies: focusing on induced pluripotent stem cell (iPSC)-based treatments. Stem Cell Res Ther 2024; 15:447. [PMID: 39574212 PMCID: PMC11583560 DOI: 10.1186/s13287-024-04065-9] [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: 08/16/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
Stem cell therapies have emerged as a promising approach in regenerative medicine, demonstrating potential in personalized medicine, disease modeling, and drug discovery. Therapies based on induced pluripotent stem cells (iPSCs) particularly stand out for their ability to differentiate into various cell types while avoiding ethical concerns. However, the development and application of these therapies are influenced by varying regulatory frameworks across countries. This study provides a comparative analysis of regulations and research on stem cell therapies in key regions: The European Union (EU), Switzerland, South Korea, Japan, and the United States. First, the study reviews the regulatory frameworks on stem cell therapies. The EU and Switzerland maintain rigorous guidelines that prioritize safety and ethical considerations, which can hinder innovation. In contrast, the United States adopts a more flexible regulatory stance, facilitating the rapid development of stem cell therapies. South Korea and Japan take a balanced approach by incorporating practices from both regimes. These regulatory differences reflect each country's unique priorities and impact the pace and scope of stem cell therapy development. Moreover, the study examines global trends in clinical trials on stem cell treatments based on data obtained from two sources: ClinicalTrials.gov and ICTRP. Findings indicate a significant growth in the number of clinical trials since 2008, particularly in that involving iPSCs. Therapeutic studies involving iPSCs predominantly target conditions affecting the cardiovascular and nervous systems which are considered vital. The results put emphasis on the safety of stem cell treatments. Meanwhile, the number of such trials also varies by country. The United States and Japan, where relatively flexible guidelines on stem cell research are adopted, are in a leading position. However, countries in the EU fall behind with rigorous regulations imposed. This reflects the need for more flexible regulatory guidance for active development of stem cell therapies. The findings underscore the importance of legal frameworks in facilitating innovation while ensuring safety. Regulatory agencies in different countries should collaborate to achieve a balanced global standard to ensure the safe and efficient advancement of stem cell therapies. Global regulatory convergence will promote international collaboration in research and the applicability of new treatments.
Collapse
Affiliation(s)
- Seohyun Jennie Song
- Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Yoojun Nam
- YiPSCELL Inc., L2 Omnibus Park, Banpo-Dearo 222, Seocho-Gu, Seoul, 06591, Republic of Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- YiPSCELL Inc., L2 Omnibus Park, Banpo-Dearo 222, Seocho-Gu, Seoul, 06591, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- YiPSCELL Inc., L2 Omnibus Park, Banpo-Dearo 222, Seocho-Gu, Seoul, 06591, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, #505, Banpo-Dong, Seocho-Gu, Seoul, 06591, Republic of Korea.
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
2
|
Jeyaraman M, Jeyaraman N, Ramasubramanian S, Balaji S, Muthu S. Evidence-based orthobiologic practice: Current evidence review and future directions. World J Orthop 2024; 15:908-917. [DOI: 10.5312/wjo.v15.i10.908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/31/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024] Open
Abstract
The field of orthopedic and regenerative medicine is rapidly evolving with the increasing utilization of orthobiologic. These biologically derived therapies, including platelet-rich plasma, mesenchymal stem cells, bone marrow aspirate concentrate, stromal vascular fraction (SVF), and autologous chondrocyte implantation, are gaining traction for their potential to enhance the body's natural healing processes. They offer a promising alternative to traditional surgical interventions for musculoskeletal injuries and degenerative conditions. Current evidence suggests significant benefits of orthobiologics in treating conditions like osteoarthritis, tendon injuries, and spinal disorders, yet inconsistencies in treatment protocols and outcomes persist. The global market for orthobiologics is projected to grow substantially, driven by advancements in biologic therapies such as adipose-derived stem cells and SVF, and the demand for minimally invasive treatments. Despite their promise, regulatory and ethical challenges, as well as the need for high-quality, standardized research, remain significant obstacles. Future directions in the field include advancements in delivery systems, personalized medicine approaches, and the exploration of novel sources like induced pluripotent stem cells, aiming for more targeted and effective treatments. Collaborative efforts are crucial to overcoming these challenges and ensuring the safe and effective application of orthobiologics in clinical practice.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Research Methods, Orthopedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Naveen Jeyaraman
- Department of Orthopedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of Orthopedic, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Sangeetha Balaji
- Department of Orthopedic, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Sathish Muthu
- Department of Research Methods, Orthopedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| |
Collapse
|
3
|
Mansoori S, Noei A, Maali A, Seyed-Motahari SS, Sharifzadeh Z. Recent updates on allogeneic CAR-T cells in hematological malignancies. Cancer Cell Int 2024; 24:304. [PMID: 39227937 PMCID: PMC11370086 DOI: 10.1186/s12935-024-03479-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 08/13/2024] [Indexed: 09/05/2024] Open
Abstract
CAR-T cell therapy is known as an effective therapy in patients with hematological malignancies. Since 2017, several autologous CAR-T cell (auto-CAR-T) drugs have been approved by the US Food and Drug Administration (FDA) for the treatment of some kinds of relapsed/refractory hematological malignancies. However, some patients fail to respond to these drugs due to high manufacturing time, batch-to-batch variation, poor quality and insufficient quantity of primary T cells, and their insufficient expansion and function. CAR-T cells prepared from allogeneic sources (allo-CAR-Ts) can be an alternative option to overcome these obstacles. Recently, several allo-CAR-Ts have entered into the early clinical trials. Despite their promising preclinical and clinical results, there are two main barriers, including graft-versus-host disease (GvHD) and allo-rejection that may decline the safety and efficacy of allo-CAR-Ts in the clinic. The successful development of these products depends on the starter cell source, the gene editing method, and the ability to escape immune rejection and prevent GvHD. Here, we summarize the gene editing technologies and the potential of various cell sources for developing allo-CAR-Ts and highlight their advantages for the treatment of hematological malignancies. We also describe preclinical and clinical data focusing on allo-CAR-T therapy in blood malignancies and discuss challenges and future perspectives of allo-CAR-Ts for therapeutic applications.
Collapse
Affiliation(s)
| | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | | |
Collapse
|
4
|
Yaqubi S, Karimian M. Stem cell therapy as a promising approach for ischemic stroke treatment. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 6:100183. [PMID: 38831867 PMCID: PMC11144755 DOI: 10.1016/j.crphar.2024.100183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Ischemia as the most common type of stroke is the main cause of death and disability in the world. However, there are few therapeutic approaches to treat ischemic stroke. The common approach to the treatment of ischemia includes surgery-cum-chemical drugs. Surgery and chemical drugs are used to remove blood clots to prevent the deterioration of the nervous system. Given the surgical hazards and the challenges associated with chemical drugs, these cannot be considered safe approaches to the treatment of brain ischemia. Besides surgery-cum-chemical drugs, different types of stem cells including mesenchymal stem cells and neurological stem cells have been considered to treat ischemic stroke. Therapeutic approaches utilizing stem cells to treat strokes are promising because of their neuroprotective and regenerative benefits. However, the mechanisms by which the transplanted stem cells perform their precisely actions are unknown. The purpose of this study is to critically review stem cell-based therapeutic approaches for ischemia along with related challenges.
Collapse
Affiliation(s)
- Sahar Yaqubi
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
5
|
Krishnan I, Chan AML, Law JX, Ng MH, Jayapalan JJ, Lokanathan Y. Proteomic Analysis of Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Systematic Review. Int J Mol Sci 2024; 25:5340. [PMID: 38791378 PMCID: PMC11121203 DOI: 10.3390/ijms25105340] [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: 03/26/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Numerous challenges remain within conventional cell-based therapy despite the growing trend of stem cells used to treat various life-debilitating diseases. These limitations include batch-to-batch heterogeneity, induced alloreactivity, cell survival and integration, poor scalability, and high cost of treatment, thus hindering successful translation from lab to bedside. However, recent pioneering technology has enabled the isolation and enrichment of small extracellular vesicles (EVs), canonically known as exosomes. EVs are described as a membrane-enclosed cargo of functional biomolecules not limited to lipids, nucleic acid, and proteins. Interestingly, studies have correlated the biological role of MSC-EVs to the paracrine activity of MSCs. This key evidence has led to rigorous studies on MSC-EVs as an acellular alternative. Using EVs as a therapy was proposed as a model leading to improvements through increased safety; enhanced bioavailability due to size and permeability; reduced heterogeneity by selective and quantifiable properties; and prolonged shelf-life via long-term freezing or lyophilization. Yet, the identity and potency of EVs are still relatively unknown due to various methods of preparation and to qualify the final product. This is reflected by the absence of regulatory strategies overseeing manufacturing, quality control, clinical implementation, and product registration. In this review, the authors review the various production processes and the proteomic profile of MSC-EVs.
Collapse
Affiliation(s)
- Illayaraja Krishnan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | | | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| |
Collapse
|
6
|
Xu J, Chen H, Wang C, Ma Y, Song Y. Raman Flow Cytometry and Its Biomedical Applications. BIOSENSORS 2024; 14:171. [PMID: 38667164 PMCID: PMC11048678 DOI: 10.3390/bios14040171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
Raman flow cytometry (RFC) uniquely integrates the "label-free" capability of Raman spectroscopy with the "high-throughput" attribute of traditional flow cytometry (FCM), offering exceptional performance in cell characterization and sorting. Unlike conventional FCM, RFC stands out for its elimination of the dependency on fluorescent labels, thereby reducing interference with the natural state of cells. Furthermore, it significantly enhances the detection information, providing a more comprehensive chemical fingerprint of cells. This review thoroughly discusses the fundamental principles and technological advantages of RFC and elaborates on its various applications in the biomedical field, from identifying and characterizing cancer cells for in vivo cancer detection and surveillance to sorting stem cells, paving the way for cell therapy, and identifying metabolic products of microbial cells, enabling the differentiation of microbial subgroups. Moreover, we delve into the current challenges and future directions regarding the improvement in sensitivity and throughput. This holds significant implications for the field of cell analysis, especially for the advancement of metabolomics.
Collapse
Affiliation(s)
- Jiayang Xu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou 310058, China;
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Hongyi Chen
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- Division of Life Sciences and Medicine, School of Biomedical Engineering (Suzhou), University of Science and Technology of China, Suzhou 215163, China
| | - Ce Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yuting Ma
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yizhi Song
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- Division of Life Sciences and Medicine, School of Biomedical Engineering (Suzhou), University of Science and Technology of China, Suzhou 215163, China
| |
Collapse
|
7
|
Bharuka T, Reche A. Advancements in Periodontal Regeneration: A Comprehensive Review of Stem Cell Therapy. Cureus 2024; 16:e54115. [PMID: 38487109 PMCID: PMC10938178 DOI: 10.7759/cureus.54115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Periodontal disease, characterized by inflammation and infection of the supporting structures of teeth, presents a significant challenge in dentistry and public health. Current treatment modalities, while effective to some extent, have limitations in achieving comprehensive periodontal tissue regeneration. This comprehensive review explores the potential of stem cell therapy in advancing the field of periodontal regeneration. Stem cells, including mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs), hold promise due to their immunomodulatory effects, differentiation potential into periodontal tissues, and paracrine actions. Preclinical studies using various animal models have revealed encouraging outcomes, though standardization and long-term assessment remain challenges. Clinical trials and case studies demonstrate the safety and efficacy of stem cell therapy in real-world applications, especially in personalized regenerative medicine. Patient selection criteria, ethical considerations, and standardized treatment protocols are vital for successful clinical implementation. Stem cell therapy is poised to revolutionize periodontal regeneration, offering more effective, patient-tailored treatments while addressing the systemic health implications of periodontal disease. This transformative approach holds the potential to significantly impact clinical practice and improve the overall well-being of individuals affected by this prevalent oral health concern. Responsible regulatory compliance and a focus on ethical considerations will be essential as stem cell therapy evolves in periodontal regeneration.
Collapse
Affiliation(s)
- Tanvi Bharuka
- Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Amit Reche
- Public Health Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
8
|
Song Y, Tian C, Lee Y, Yoon M, Yoon SE, Cho SY. Nanosensor Chemical Cytometry: Advances and Opportunities in Cellular Therapy and Precision Medicine. ACS MEASUREMENT SCIENCE AU 2023; 3:393-403. [PMID: 38145025 PMCID: PMC10740128 DOI: 10.1021/acsmeasuresciau.3c00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 12/26/2023]
Abstract
With the definition of therapeutics now encompassing transplanted or engineered cells and their molecular products, there is a growing scientific necessity for analytics to understand this new category of drugs. This Perspective highlights the recent development of new measurement science on label-free single cell analysis, nanosensor chemical cytometry (NCC), and their potential for cellular therapeutics and precision medicine. NCC is based on microfluidics integrated with fluorescent nanosensor arrays utilizing the optical lensing effect of a single cell to real-time extract molecular properties and correlate them with physical attributes of single cells. This new class of cytometry can quantify the heterogeneity of the multivariate physicochemical attributes of the cell populations in a completely label-free and nondestructive way and, thus, suggest the vein-to-vein conditions for the safe therapeutic applications. After the introduction of the NCC technology, we suggest the technological development roadmap for the maturation of the new field: from the sensor/chip design perspective to the system/software development level based on hardware automation and deep learning data analytics. The advancement of this new single cell sensing technology is anticipated to aid rich and multivariate single cell data setting and support safe and reliable cellular therapeutics. This new measurement science can lead to data-driven personalized precision medicine.
Collapse
Affiliation(s)
- Youngho Song
- School
of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Changyu Tian
- School
of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yullim Lee
- School
of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minyeong Yoon
- School
of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sang Eun Yoon
- Division
of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Soo-Yeon Cho
- School
of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
9
|
An S, Anwar K, Ashraf M, Lee H, Jung R, Koganti R, Ghassemi M, Djalilian AR. Wound-Healing Effects of Mesenchymal Stromal Cell Secretome in the Cornea and the Role of Exosomes. Pharmaceutics 2023; 15:1486. [PMID: 37242728 PMCID: PMC10221647 DOI: 10.3390/pharmaceutics15051486] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) and their secreted factors have been shown to have immunomodulatory and regenerative effects. In this study, we investigated human bone-marrow-derived MSC secretome (MSC-S) for the treatment of corneal epithelial wounds. Specifically, we evaluated the role of MSC extracellular vesicles (EV)/exosomes in mediating the wound-healing effects of the MSC-S. In vitro studies using human corneal epithelial cells showed that MSC-CM increased cell proliferation in HCEC and HCLE cells, while EV-depleted MSC-CM showed lower cell proliferation in both cell lines compared to the MSC-CM group. In vitro and in vivo experiments revealed that 1X MSC-S consistently promoted wound healing more effectively than 0.5X MSC-S, and MSC-CM promoted wound healing in a dose-dependent manner, while exosome deprivation delayed wound healing. We further evaluated the incubation period of MSC-CM on corneal wound healing and showed that MSC-S collected for 72 h is more effective than MSC-S collected for 48 h. Finally, we evaluated the stability of MSC-S under different storage conditions and found that after one cycle of freeze-thawing, MSC-S is stable at 4 °C for up to 4 weeks. Collectively, we identified the following: (i) MSC-EV/Exo as the active ingredient in MSC-S that mediates the wound-healing effects in the corneal epithelium, providing a measure to optimize its dosing for a potential clinical product; (ii) Treatment with EV/Exo-containing MSC-S resulted in an improved corneal barrier and decreased corneal haze/edema relative to EV/Exo-depleted MSC-S; (iii) The stability of MSC-CM for up to 4 weeks showed that the regular storage condition did not significantly impact its stability and therapeutic functions.
Collapse
Affiliation(s)
- Seungwon An
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Khandaker Anwar
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Mohammadjavad Ashraf
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Hyungjo Lee
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Rebecca Jung
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Mahmood Ghassemi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| |
Collapse
|
10
|
Zhang X, Wang D, Wang Z, Ling SKK, Yung PSH, Tuan RS, Ker DFE. Clinical perspectives for repairing rotator cuff injuries with multi-tissue regenerative approaches. J Orthop Translat 2022; 36:91-108. [PMID: 36090820 PMCID: PMC9428729 DOI: 10.1016/j.jot.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background In the musculoskeletal system, bone, tendon, and muscle form highly integrated multi-tissue units such as the rotator cuff complex, which facilitates functional and dynamic movement of the shoulder joint. Understanding the intricate interplay among these tissues within clinical, biological, and engineering contexts is vital for addressing challenging issues in treatment of musculoskeletal disorders and injuries. Methods A wide-ranging literature search was performed, and findings related to the socioeconomic impact of rotator cuff tears, the structure-function relationship of rotator cuff bone-tendon-muscle units, pathophysiology of injury, current clinical treatments, recent state-of-the-art advances (stem cells, growth factors, and exosomes) as well as their regulatory approval, and future strategies aimed at engineering bone-tendon-muscle musculoskeletal units are outlined. Results Rotator cuff injuries are a significant socioeconomic burden on numerous healthcare systems that may be addressed by treating the rotator cuff as a single complex, given its highly integrated structure-function relationship as well as degenerative pathophysiology and limited healing in bone-tendon-muscle musculoskeletal tissues. Current clinical practices for treating rotator cuff injuries, including the use of commercially available devices and evolving trends in surgical management have benefited patients while advances in application of stem/progenitor cells, growth factors, and exosomes hold clinical potential. However, such efforts do not emphasize targeted regeneration of bone-tendon-muscle units. Strategies aimed at regenerating bone-tendon-muscle units are thus expected to address challenging issues in rotator cuff repair. Conclusions The rotator cuff is a highly integrated complex of bone-tendon-muscle units that when injured, has severe consequences for patients and healthcare systems. State-of-the-art clinical treatment as well as recent advances have resulted in improved patient outcome and may be further enhanced by engineering bone-tendon-muscle multi-tissue grafts as a potential strategy for rotator cuff injuries. Translational Potential of this Article This review aims to bridge clinical, tissue engineering, and biological aspects of rotator cuff repair and propose a novel therapeutic strategy by targeted regeneration of multi-tissue units. The presentation of these wide-ranging and multi-disciplinary concepts are broadly applicable to regenerative medicine applications for musculoskeletal and non-musculoskeletal tissues.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Hong Kong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Zuyong Wang
- College of Materials Science and Engineering, Hunan University, Changsha, China
| | - Samuel Ka-kin Ling
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Patrick Shu-hang Yung
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Hong Kong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| |
Collapse
|
11
|
3D-Printing Graphene Scaffolds for Bone Tissue Engineering. Pharmaceutics 2022; 14:pharmaceutics14091834. [PMID: 36145582 PMCID: PMC9503344 DOI: 10.3390/pharmaceutics14091834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Graphene-based materials have recently gained attention for regenerating various tissue defects including bone, nerve, cartilage, and muscle. Even though the potential of graphene-based biomaterials has been realized in tissue engineering, there are significantly many more studies reporting in vitro and in vivo data in bone tissue engineering. Graphene constructs have mainly been studied as two-dimensional (2D) substrates when biological organs are within a three-dimensional (3D) environment. Therefore, developing 3D graphene scaffolds is the next clinical standard, yet most have been fabricated as foams which limit control of consistent morphology and porosity. To overcome this issue, 3D-printing technology is revolutionizing tissue engineering, due to its speed, accuracy, reproducibility, and overall ability to personalize treatment whereby scaffolds are printed to the exact dimensions of a tissue defect. Even though various 3D-printing techniques are available, practical applications of 3D-printed graphene scaffolds are still limited. This can be attributed to variations associated with fabrication of graphene derivatives, leading to variations in cell response. This review summarizes selected works describing the different fabrication techniques for 3D scaffolds, the novelty of graphene materials, and the use of 3D-printed scaffolds of graphene-based nanoparticles for bone tissue engineering.
Collapse
|
12
|
Muthu S, Jeyaraman M, Kotner MB, Jeyaraman N, Rajendran RL, Sharma S, Khanna M, Rajendran SNS, Oh JM, Gangadaran P, Ahn BC. Evolution of Mesenchymal Stem Cell Therapy as an Advanced Therapeutic Medicinal Product (ATMP)-An Indian Perspective. Bioengineering (Basel) 2022; 9:bioengineering9030111. [PMID: 35324800 PMCID: PMC8945480 DOI: 10.3390/bioengineering9030111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023] Open
Abstract
Stem cells can be defined as the cells that have the capacity to both self-renew and give rise to differentiated cells. Under the right conditions and signals, depending on their origin and bio-plasticity, stem cells can differentiate into multiple cell lineages and develop into various mature cells. Stem cell therapy is a fast-developing branch of medicine that includes the most innovative regenerative therapies for the restoration of cell and tissue function in individuals with severe diseases. Stem cell research has resulted in the emergence of cell-based therapies for disorders that are resistant to conventional drugs and therapies, and they are considered under the category of an Advanced Therapeutic Medicinal Product (ATMP). The FDA and the European Medicines Agency (EMA) devised a new strategy in 2017 with the aim of unifying the standards for development of ATMPs such that it is easy to exchange information at the international level. In this review, we discuss the evolution of mesenchymal stem cell-based therapy as an ATMP in the global and Indian scenarios, along with the guidelines governing their usage and clinical application of these therapeutics.
Collapse
Affiliation(s)
- Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624001, India;
- Indian Stem Cell Study Group, Lucknow 226010, India; (M.B.K.); (N.J.); (M.K.)
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group, Lucknow 226010, India; (M.B.K.); (N.J.); (M.K.)
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India
- Department of Orthopaedics, Faculty of Medicine-Sri Lalithambigai Medical College and Hospital, Dr. MGR Educational and Research Institute University, Chennai 600095, India
- Correspondence: (M.J.); (P.G.); (B.-C.A.)
| | - Moinuddin Basha Kotner
- Indian Stem Cell Study Group, Lucknow 226010, India; (M.B.K.); (N.J.); (M.K.)
- Fellow in Orthopaedic Rheumatology, Dr. Ram Manohar Lohiya National Law University, Lucknow 226012, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group, Lucknow 226010, India; (M.B.K.); (N.J.); (M.K.)
- Fellow in Orthopaedic Rheumatology, Dr. Ram Manohar Lohiya National Law University, Lucknow 226012, India
- Fellow in Joint Replacement, Atlas Hospitals, The Tamil Nadu Dr. MGR Medical University, Tiruchirappalli 620002, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.)
| | - Shilpa Sharma
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Manish Khanna
- Indian Stem Cell Study Group, Lucknow 226010, India; (M.B.K.); (N.J.); (M.K.)
| | - Sree Naga Sowndary Rajendran
- Department of Medicine, Sri Venkateshwaraa Medical College Hospital and Research Centre, Puducherry 605107, India;
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.)
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (M.J.); (P.G.); (B.-C.A.)
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (M.J.); (P.G.); (B.-C.A.)
| |
Collapse
|
13
|
Mousaei Ghasroldasht M, Seok J, Park HS, Liakath Ali FB, Al-Hendy A. Stem Cell Therapy: From Idea to Clinical Practice. Int J Mol Sci 2022; 23:ijms23052850. [PMID: 35269990 PMCID: PMC8911494 DOI: 10.3390/ijms23052850] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
Regenerative medicine is a new and promising mode of therapy for patients who have limited or no other options for the treatment of their illness. Due to their pleotropic therapeutic potential through the inhibition of inflammation or apoptosis, cell recruitment, stimulation of angiogenesis, and differentiation, stem cells present a novel and effective approach to several challenging human diseases. In recent years, encouraging findings in preclinical studies have paved the way for many clinical trials using stem cells for the treatment of various diseases. The translation of these new therapeutic products from the laboratory to the market is conducted under highly defined regulations and directives provided by competent regulatory authorities. This review seeks to familiarize the reader with the process of translation from an idea to clinical practice, in the context of stem cell products. We address some required guidelines for clinical trial approval, including regulations and directives presented by the Food and Drug Administration (FDA) of the United States, as well as those of the European Medicine Agency (EMA). Moreover, we review, summarize, and discuss regenerative medicine clinical trial studies registered on the Clinicaltrials.gov website.
Collapse
|
14
|
Buja LM, Mitchell RN. Basic pathobiology of cell-based therapies and cardiac regenerative medicine. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00016-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
15
|
Aishwarya L, Arun D, Kannan S. Stem cells as a potential therapeutic option for treating neurodegenerative diseases. Curr Stem Cell Res Ther 2021; 17:590-605. [PMID: 35135464 DOI: 10.2174/1574888x16666210810105136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 11/22/2022]
Abstract
In future, neurodegenerative diseases will take over cancer's place and become the major cause of death in the world, especially in developed countries. Advancements in the medical field and its facilities have led to an increase in the old age population, and thus contributing to the increase in number of people suffering from neurodegenerative diseases. Economically it is of a great burden to society and the affected family. No current treatment aims to replace, protect, and regenerate lost neurons; instead, it alleviates the symptoms, extends the life span by a few months and creates severe side effects. Moreover, people who are affected are physically dependent for performing their basic activities, which makes their life miserable. There is an urgent need for therapy that could be able to overcome the deficits of conventional therapy for neurodegenerative diseases. Stem cells, the unspecialized cells with the properties of self-renewing and potency to differentiate into various cells types can become a potent therapeutic option for neurodegenerative diseases. Stem cells have been widely used in clinical trials to evaluate their potential in curing different types of ailments. In this review, we discuss the various types of stem cells and their potential use in the treatment of neurodegenerative disease based on published preclinical and clinical studies.
Collapse
Affiliation(s)
- Aishwarya L
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| | - Dharmarajan Arun
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| | - Suresh Kannan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| |
Collapse
|
16
|
Thej C, Balasubramanian S, Rengasamy M, Walvekar A, Swamynathan P, Raj SS, Shahani P, Siddikuzzaman, Kolkundkar U, Seetharam RN, Gupta PK, Majumdar AS. Human bone marrow-derived, pooled, allogeneic mesenchymal stromal cells manufactured from multiple donors at different times show comparable biological functions in vitro, and in vivo to repair limb ischemia. Stem Cell Res Ther 2021; 12:279. [PMID: 33971964 PMCID: PMC8108338 DOI: 10.1186/s13287-021-02330-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We have previously demonstrated that a pooled population of bone marrow-derived, allogeneic mesenchymal stromal cells (BMMSC), Stempeucel®-1, produced under good manufacturing practices (GMP) conditions, showed clinical efficacy and safety in patients suffering from critical limb ischemia (CLI) due to Buerger's disease. While Stempeucel®-1 is currently used for CLI and other clinical indications, we wanted to ensure that the product's continuity is addressed by developing and characterizing a second generation of pooled product (Stempeucel®-1A), manufactured identically from second BM aspirates of the same three donors after a 2-year interval. METHODS The two versions of Stempeucel® were manufactured and subjected to gene and protein expression analysis. The nature of various growth factors/cytokines secreted and immunomodulatory activity of these two cell populations were compared directly by various in vitro assays. The preclinical efficacy of these two cell types was compared in an experimental model of hind limb ischemia (HLI) in BALB/c nude mice. The reversal of ischemia, blood flow, and muscle regeneration were determined by functional scoring, laser Doppler imaging, and immunohistochemical analyses. RESULTS Qualitative and quantitative analyses of genes and proteins involved in promoting angiogenic activity and immune regulatory functions revealed high levels of correlation between Stempeucel®-1 and Stempeucel®-1A cell populations. Moreover, intramuscular (i.m) administration of these two cell products in the ischemic limbs of BALB/c nude mice showed significant repair (≥ 70%) of toe and foot necrosis, leading to improved ambulatory function and limb salvage. Furthermore, a biodistribution kinetics study showed that Stempeucel®-1 was mostly localized in the ischemic muscles of mice for a significantly longer time compared to normal muscles, thus playing an essential role in modulating and reversing HLI damage. CONCLUSIONS This study shows that with a reproducible manufacturing procedure, it is possible to generate large numbers of pooled mesenchymal stromal cells from human bone marrow samples to establish product equivalence. We conclude from these results that, for the first time, two pooled, allogeneic BMMSC products can be repeatedly manufactured at different time intervals using a two-tier cell banking process with robust and comparable angiogenic properties to treat ischemic diseases.
Collapse
Affiliation(s)
- Charan Thej
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Sudha Balasubramanian
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Mathiyazhagan Rengasamy
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Ankita Walvekar
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Priyanka Swamynathan
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Swathi Sundar Raj
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Pradnya Shahani
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Siddikuzzaman
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Udaykumar Kolkundkar
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Raviraja N Seetharam
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Pawan Kumar Gupta
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India
| | - Anish S Majumdar
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, K R Puram Hobli, Bengaluru, India.
| |
Collapse
|
17
|
Gala de Pablo J, Lindley M, Hiramatsu K, Goda K. High-Throughput Raman Flow Cytometry and Beyond. Acc Chem Res 2021; 54:2132-2143. [PMID: 33788539 DOI: 10.1021/acs.accounts.1c00001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Flow cytometry is a powerful tool with applications in diverse fields such as microbiology, immunology, virology, cancer biology, stem cell biology, and metabolic engineering. It rapidly counts and characterizes large heterogeneous populations of cells in suspension (e.g., blood cells, stem cells, cancer cells, and microorganisms) and dissociated solid tissues (e.g., lymph nodes, spleen, and solid tumors) with typical throughputs of 1,000-100,000 events per second (eps). By measuring cell size, cell granularity, and the expression of cell surface and intracellular molecules, it provides systematic insights into biological processes. Flow cytometers may also include cell sorting capabilities to enable subsequent additional analysis of the sorted sample (e.g., electron microscopy and DNA/RNA sequencing), cloning, and directed evolution. Unfortunately, traditional flow cytometry has several critical limitations as it mainly relies on fluorescent labeling for cellular phenotyping, which is an indirect measure of intracellular molecules and surface antigens. Furthermore, it often requires time-consuming preparation protocols and is incompatible with cell therapy. To overcome these difficulties, a different type of flow cytometry based on direct measurements of intracellular molecules by Raman spectroscopy, or "Raman flow cytometry" for short, has emerged. Raman flow cytometry obtains a chemical fingerprint of the cell in a nondestructive manner, allowing for single-cell metabolic phenotyping. However, its slow signal acquisition due to the weak light-molecule interaction of spontaneous Raman scattering prevents the throughput necessary to interrogate large cell populations in reasonable time frames, resulting in throughputs of about 1 eps. The remedy to this throughput limit lies in coherent Raman scattering methods such as stimulated Raman scattering (SRS) and coherent anti-Stokes Raman scattering (CARS), which offer a significantly enhanced light-sample interaction and hence enable high-throughput Raman flow cytometry, Raman imaging flow cytometry, and even Raman image-activated cell sorting (RIACS). In this Account, we outline recent advances, technical challenges, and emerging opportunities of coherent Raman flow cytometry. First, we review the principles of various types of SRS and CARS and introduce several techniques of coherent Raman flow cytometry such as CARS, multiplex CARS, Fourier-transform CARS, SRS, SRS imaging flow cytometry, and RIACS. Next, we discuss a unique set of applications enabled by coherent Raman flow cytometry, from microbiology and lipid biology to cancer detection and cell therapy. Finally, we describe future opportunities and challenges of coherent Raman flow cytometry including increasing sensitivity and throughput, integration with droplet microfluidics, utilizing machine learning techniques, or achieving in vivo flow cytometry. This Account summarizes the growing field of high-throughput Raman flow cytometry and the bright future it can bring.
Collapse
Affiliation(s)
- Julia Gala de Pablo
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Matthew Lindley
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Kotaro Hiramatsu
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
- Kanagawa Institute of Industrial Science and Technology, 705-1 Shimoimaizumi, Ebina, Kanagawa 243-0435, Japan
- Research Center for Spectrochemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
- Department of Bioengineering, University of California, 410 Westwood Plaza, Los Angeles, California 90095 United States
- Institute of Technological Sciences, Wuhan University, Wuchang District, Wuhan 430072, Hubei, China
| |
Collapse
|
18
|
Jha BS, Farnoodian M, Bharti K. Regulatory considerations for developing a phase I investigational new drug application for autologous induced pluripotent stem cells-based therapy product. Stem Cells Transl Med 2021; 10:198-208. [PMID: 32946199 PMCID: PMC7848308 DOI: 10.1002/sctm.20-0242] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/04/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
Induced pluripotent stem cells (iPSC)-based therapies have been hailed as the future of regenerative medicine because of their potential to provide treatment options for most degenerative diseases. A key promise of iPSC-based therapies is the possibility of an autologous transplant that may engraft better in the longer-term due to its compatibility with the patient's immune system. Despite over a decade of research, clinical translation of autologous iPSC-based therapies has been slow-partly due to a lacking pre-defined regulatory path. Here, we outline regulatory considerations for developing an autologous iPSC-based product and challenges associated with the clinical manufacturing of autologous iPSCs and their derivatives. These challenges include donor tissue source, reprogramming methods, heterogeneity of differentiated cells, controls for the manufacturing process, and preclinical considerations. A robust manufacturing process with appropriate quality controls and well-informed, prospectively designed preclinical studies provide a path toward successful approval of autologous iPSC-based therapies.
Collapse
Affiliation(s)
- Balendu Shekhar Jha
- Center for Cell Engineering, Department of Transfusion MedicineClinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research Section, Ophthalmic Genetics and Visual Function BranchNational Eye Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, Ophthalmic Genetics and Visual Function BranchNational Eye Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
19
|
Sekar MP, Budharaju H, Zennifer A, Sethuraman S, Vermeulen N, Sundaramurthi D, Kalaskar DM. Current standards and ethical landscape of engineered tissues-3D bioprinting perspective. J Tissue Eng 2021; 12:20417314211027677. [PMID: 34377431 PMCID: PMC8330463 DOI: 10.1177/20417314211027677] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/08/2021] [Indexed: 01/17/2023] Open
Abstract
Tissue engineering is an evolving multi-disciplinary field with cutting-edge technologies and innovative scientific perceptions that promise functional regeneration of damaged tissues/organs. Tissue engineered medical products (TEMPs) are biomaterial-cell products or a cell-drug combination which is injected, implanted or topically applied in the course of a therapeutic or diagnostic procedure. Current tissue engineering strategies aim at 3D printing/bioprinting that uses cells and polymers to construct living tissues/organs in a layer-by-layer fashion with high 3D precision. However, unlike conventional drugs or therapeutics, TEMPs and 3D bioprinted tissues are novel therapeutics and need different regulatory protocols for clinical trials and commercialization processes. Therefore, it is essential to understand the complexity of raw materials, cellular components, and manufacturing procedures to establish standards that can help to translate these products from bench to bedside. These complexities are reflected in the regulations and standards that are globally in practice to prevent any compromise or undue risks to patients. This review comprehensively describes the current legislations, standards for TEMPs with a special emphasis on 3D bioprinted tissues. Based on these overviews, challenges in the clinical translation of TEMPs & 3D bioprinted tissues/organs along with their ethical concerns and future perspectives are discussed.
Collapse
Affiliation(s)
- Muthu Parkkavi Sekar
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Allen Zennifer
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Niki Vermeulen
- Department of Science, Technology and Innovation Studies, School of Social and Political Science, University of Edinburgh, High School Yards, Edinburgh, UK
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | | |
Collapse
|
20
|
Challenges for Cell-Based Medicinal Products From a Pharmaceutical Product Perspective. J Pharm Sci 2020; 110:1900-1908. [PMID: 33307042 DOI: 10.1016/j.xphs.2020.11.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/19/2020] [Accepted: 11/30/2020] [Indexed: 11/22/2022]
Abstract
Advanced therapy medicinal products (ATMPs), such as somatic cell-therapy medicinal products or tissue-engineered products for human use, offer new and potentially curative opportunities to treat yet untreatable diseases or disorders. For cell-therapy medicinal products (CBMPs), multiple stability and quality challenges exist and relate to the cellular composition and unstable nature of these parenteral preparations. It is the aim of this review to discuss open questions and problems associated with the development, manufacturing and testing of CBMPs from a pharmaceutical drug product perspective. This includes safety, storage and handling, particulates, the choice of container closure systems and integrity. Analytical methods commonly used to evaluate the quality of the final CBMP to ensure patient's safety will be discussed. Particulate contamination in final products deserve special attention since CBMPs cannot be sterile filtered. Visible and sub-visible particles may represent environmental contaminations or may form during storage. They may be introduced from processing materials such as single use product contact materials, ancillary materials, or any components such as primary packaging used for the final product. Currently available analytical methods for detecting particulates may not be easily applicable to CBMPs due to their inherent particulate nature and appearance.
Collapse
|
21
|
Whitney KE, Briggs KK, Chamness C, Bolia IK, Huard J, Philippon MJ, Evans TA. Bone Marrow Concentrate Injection Treatment Improves Short-term Outcomes in Symptomatic Hip Osteoarthritis Patients: A Pilot Study. Orthop J Sports Med 2020; 8:2325967120966162. [PMID: 33344667 PMCID: PMC7731709 DOI: 10.1177/2325967120966162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/12/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is one of the leading causes of disability in the United States, the hip being the second most affected weightbearing joint. Autologous bone marrow concentrate (BMC) is a promising alternative therapy to conventional treatments, with the potential to mitigate inflammation and improve joint function. PURPOSE To investigate the effectiveness of a single intra-articular BMC injection for patients with symptomatic hip OA. STUDY DESIGN Case series; Level of evidence, 4. METHODS A total of 24 patients diagnosed with symptomatic hip OA who elected to undergo a single BMC injection were prospectively enrolled in the study. Patients were excluded if they reported a preinjection Numeric Rating Scale (NRS) score for pain with activity of <6 points out of 10. The Western Ontario and McMaster Universities Arthritis Index (WOMAC), modified Harris Hip Score (mHHS), Hip Outcome Score-Activities of Daily Living (HOS-ADL), 12-Item Short Form Health Survey (SF-12), and NRS pain scores were collected before and after the procedure (6 weeks, 3 months, and 6 months). Joint space and Tönnis OA grade scores were recorded on preinjection anteroposterior pelvis radiographs. RESULTS A total of 18 hips from 16 patients (7 male and 9 female) (mean age, 57.6 ± 11; mean body mass index, 25.9 ± 3.6 kg/m2) were used in the final analysis. Significant improvements were observed in NRS pain with activity (from 8 to 4.5; P < .001) and without activity (from 5 to 1; P < .001), WOMAC (from 31 to 16; P = .006), mHHS (from 63 to 80; P = .004), and HOS-ADL (from 71 to 85; P = .014) over 6 months. At 6 months, all patients maintained their improvements and did not return to preprocedure status. BMI significantly correlated with baseline WOMAC scores (P = .012) and inversely correlated with 6-month SF-12 Physical Component Summary (P = .038). Tönnis grades 2 and 3 were inversely correlated with 6-week SF-12 Mental Component Summary (P = .008) and 3-month pain with activity (P = .032). No serious adverse events were reported from the BMC harvest or injection procedure. CONCLUSION A single BMC injection can significantly improve subjective pain and function scores up to 6 months in patients with symptomatic hip OA. Further studies are warranted to evaluate BMC treatment against other therapeutics in a larger sample size and compare the biological signature profiles that may be responsible for the therapeutic effect.
Collapse
Affiliation(s)
- Kaitlyn E. Whitney
- The Steadman Clinic, Vail, Colorado, USA
- Steadman Philippon Research Institute, Vail, Colorado, USA
| | | | | | | | - Johnny Huard
- Steadman Philippon Research Institute, Vail, Colorado, USA
| | | | | |
Collapse
|
22
|
Abstract
Small molecule inhibitors targeting BCL2 are explored as anticancer therapeutics. Previously, we have reported identification and characterization of a novel BCL2 inhibitor, Disarib. Disarib induced cancer cell death in a BCL2 dependent manner in different cancer cell lines and mouse tumor models when it was administered intraperitoneally. In the present study, using two syngeneic mouse models, breast adenocarcinoma (EAC) and Dalton’s lymphoma (DLA), we show that oral administration of Disarib resulted in significant tumor regression in a concentration dependent manner. Importantly, tumor developed in both female and male mice were equally sensitive to Disarib. Further, we have investigated the toxicity of Disarib in normal cells. Single dose toxicity analysis of Disarib in male and female mice after oral administration revealed no significant variations compared to control group for parameters such as body weight, food and water consumption and behavioural changes which were analysed for the entire period of study. Haematological and histopathological analyses also did not show any significant difference from the control groups. Thus, our results reveal safe use of Disarib as a small molecule inhibitor and provide the foundation for investigation of other preclinical studies.
Collapse
|
23
|
Bacon K, Lavoie A, Rao BM, Daniele M, Menegatti S. Past, Present, and Future of Affinity-based Cell Separation Technologies. Acta Biomater 2020; 112:29-51. [PMID: 32442784 PMCID: PMC10364325 DOI: 10.1016/j.actbio.2020.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Progress in cell purification technology is critical to increase the availability of viable cells for therapeutic, diagnostic, and research applications. A variety of techniques are now available for cell separation, ranging from non-affinity methods such as density gradient centrifugation, dielectrophoresis, and filtration, to affinity methods such as chromatography, two-phase partitioning, and magnetic-/fluorescence-assisted cell sorting. For clinical and analytical procedures that require highly purified cells, the choice of cell purification method is crucial, since every method offers a different balance between yield, purity, and bioactivity of the cell product. For most applications, the requisite purity is only achievable through affinity methods, owing to the high target specificity that they grant. In this review, we discuss past and current methods for developing cell-targeting affinity ligands and their application in cell purification, along with the benefits and challenges associated with different purification formats. We further present new technologies, like stimuli-responsive ligands and parallelized microfluidic devices, towards improving the viability and throughput of cell products for tissue engineering and regenerative medicine. Our comparative analysis provides guidance in the multifarious landscape of cell separation techniques and highlights new technologies that are poised to play a key role in the future of cell purification in clinical settings and the biotech industry. STATEMENT OF SIGNIFICANCE: Technologies for cell purification have served science, medicine, and industrial biotechnology and biomanufacturing for decades. This review presents a comprehensive survey of this field by highlighting the scope and relevance of all known methods for cell isolation, old and new alike. The first section covers the main classes of target cells and compares traditional non-affinity and affinity-based purification techniques, focusing on established ligands and chromatographic formats. The second section presents an excursus of affinity-based pseudo-chromatographic and non-chromatographic technologies, especially focusing on magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting (FACS). Finally, the third section presents an overview of new technologies and emerging trends, highlighting how the progress in chemical, material, and microfluidic sciences has opened new exciting avenues towards high-throughput and high-purity cell isolation processes. This review is designed to guide scientists and engineers in their choice of suitable cell purification techniques for research or bioprocessing needs.
Collapse
Affiliation(s)
- Kaitlyn Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA
| | - Ashton Lavoie
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA
| | - Balaji M Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695-7928, USA
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University - University of North Carolina Chapel Hill, North Carolina, United States
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695-7928, USA.
| |
Collapse
|
24
|
Jose J, George T, Thomas AM. Regulation of Stem Cell-Based Research in India in Comparison with the US, EU and other Asian Countries: Current Issues and Future Perspectives. Curr Stem Cell Res Ther 2020; 15:492-508. [PMID: 32250233 DOI: 10.2174/1574888x15666200402134750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/20/2019] [Accepted: 11/29/2019] [Indexed: 11/22/2022]
Abstract
Stem cell therapy is applicable for repair and replacement of damaged cells and tissues. Apart from transplanting cells to the body, the stem cell therapy directs them to grow new and healthy tissues. Stem cells in the area of regenerative medicines hold tremendous promise that may help to regenerate the damaged tissues and heal various diseases like multiple sclerosis, heart diseases, Parkinson's disease, and so on. To prove the safety, efficacy, and for the requirement of a licence for manufacturing and sale, all the stem cell therapies should pass the required criteria and undergo certain examinations of the regulatory agencies. The regulatory authorities review the manufacturing procedures of products to assure its purity and potency. This review summarizes the comparative critical evaluations of existing regulations and developments on the stem cells research in India, USA, EU and Asian regions and also discusses the challenges that have to be overcome and the important points that should be understood to position India as a source of the perspective nation in stem cells around the world.
Collapse
Affiliation(s)
- Jobin Jose
- Department of Pharmaceutical Regulatory Affairs and Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, NITTE Deemed to be University, Paneer, Mangalore 575018, Karnataka, India
| | - Teena George
- Department of Pharmaceutical Regulatory Affairs and Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, NITTE Deemed to be University, Paneer, Mangalore 575018, Karnataka, India
| | - Aaron M Thomas
- Department of Pharmaceutical Regulatory Affairs and Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, NITTE Deemed to be University, Paneer, Mangalore 575018, Karnataka, India
| |
Collapse
|
25
|
Stine SJ, Popowski KD, Su T, Cheng K. Exosome and Biomimetic Nanoparticle Therapies for Cardiac Regenerative Medicine. Curr Stem Cell Res Ther 2020; 15:674-684. [PMID: 32148200 PMCID: PMC7805022 DOI: 10.2174/1574888x15666200309143924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Exosomes and biomimetic nanoparticles have great potential to develop into a wide-scale therapeutic platform within the regenerative medicine industry. Exosomes, a subgroup of EVs with diameter ranging from 30-100 nm, have recently gained attention as an innovative approach for the treatment of various diseases, including heart disease. Their beneficial factors and regenerative properties can be contrasted with various cell types. Various biomimetic nanoparticles have also emerged as a unique platform in regenerative medicine. Biomimetic nanoparticles are a drug delivery platform, which have the ability to contain both biological and fabricated components to improve therapeutic efficiency and targeting. The novelty of these platforms holds promise for future clinical translation upon further investigation. In order for both exosome therapeutics and biomimetic nanoparticles to translate into large-scale clinical treatment, numerous factors must first be considered and improved. Standardization of different protocols, from exosome isolation to storage conditions, must be optimized to ensure batches are pure. Standardization is also important to ensure no variability in this process across studies, thus making it easier to interpret data across different disease models and treatments. Expansion of clinical trials incorporating both biomimetic nanoparticles and exosomes will require a standardization of fabrication and isolation techniques, as well as stricter regulations to ensure reproducibility across various studies and disease models. This review will summarize current research on exosome therapeutics and the application of biomimetic nanoparticles in cardiac regenerative medicine, as well as applications for exosome expansion and delivery on a large clinical scale.
Collapse
Affiliation(s)
- Sydney J. Stine
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC USA
| | - Kristen D. Popowski
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC USA
| | - Teng Su
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| |
Collapse
|
26
|
Stem cell/cellular interventions in human spinal cord injury: Is it time to move from guidelines to regulations and legislations? Literature review and Spinal Cord Society position statement. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2019; 28:1837-1845. [PMID: 31098715 DOI: 10.1007/s00586-019-06003-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 05/05/2019] [Indexed: 01/01/2023]
Abstract
PURPOSE In preclinical studies, many stem cell/cellular interventions demonstrated robust regeneration and/or repair in case of SCI and were considered a promising therapeutic candidate. However, data from clinical studies are not robust. Despite lack of substantial evidence for the efficacy of these interventions in spinal cord injury (SCI), many clinics around the world offer them as "therapy." These "clinics" claim efficacy through patient testimonials and self-advertisement without any scientific evidence to validate their claims. Thus, SCS established a panel of experts to review published preclinical studies, clinical studies and current global guidelines/regulations on usage of cellular transplants and make recommendations for their clinical use. METHODS The literature review and draft position statement was compiled and circulated among the panel and relevant suggestions incorporated to reach consensus. This was discussed and finalized in an open forum during the SCS Annual Meeting, ISSICON. RESULTS Preclinical evidence suggests safety and clinical potency of cellular interventions after SCI. However, evidence from clinical studies consisted of mostly case reports or uncontrolled case series/studies. Data from animal studies cannot be generalized to human SCI with regard to toxicity prediction after auto/allograft transplantation. CONCLUSIONS Currently, cellular/stem cell transplantation for human SCI is experimental and needs to be tested through a valid clinical trial program. It is not ethical to provide unproven transplantation as therapy with commercial implications. To stop the malpractice of marketing such "unproven therapies" to a vulnerable population, it is crucial that all countries unite to form common, well-defined regulations/legislation on their use in SCI. These slides can be retrieved from Electronic Supplementary Material.
Collapse
|
27
|
Buja LM. Cardiac repair and the putative role of stem cells. J Mol Cell Cardiol 2019; 128:96-104. [DOI: 10.1016/j.yjmcc.2019.01.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/30/2018] [Accepted: 01/24/2019] [Indexed: 01/05/2023]
|
28
|
Larijani B, Goodarzi P, Payab M, Tayanloo-Beik A, Sarvari M, Gholami M, Gilany K, Nasli-Esfahani E, Yarahmadi M, Ghaderi F, Arjmand B. The Design and Application of an Appropriate Parkinson's Disease Animal Model in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1341:89-105. [PMID: 31485993 DOI: 10.1007/5584_2019_422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Aging as an inevitable and complex physiological process occurs through a progressive decrease in the potential of tissue regeneration. Given the increasing global outbreak of aging and age-related disorders, it is important to control this phenomenon. Parkinson's disease (one of the age-related neurodegenerative and progressive disorders) resulted from predominant dopaminergic neurons deficiency. Usual Parkinson's disease treatments just can lead to symptomatically relieving. Recently, cell therapy and regenerative medicine a great promise in the treatment of several types of disorders including Parkinson's disease. Herein, before starting clinical trials, preclinical studies should be performed to answer some fundamental questions about the safety and efficacy of various treatments. Additionally, developing a well-designed and approved study is required to provide an appropriate animal model with strongly reliable validation methods. Hereupon, this review will discuss about the design and application of an appropriate Parkinson's disease animal model in regenerative medicine. EVIDENCE ACQUISITION In order to conduct the present review, numbers of Parkinson's disease preclinical studies, as well as literatures related to the animal modeling, were considered. RESULTS Appropriate animal models which approved by related authorize committees should have a high similarity to humans from anatomical, physiological, behavioral, and genetic characteristics view of point. CONCLUSION It is concluded that animal studies before starting clinical trials have an important role in answering the crucial questions about the various treatments safety and efficacy. Therein, it is recommended that all of animal modeling stages be assessed by animal ethics and welfare guidelines and also evaluated by different validation tests. However, it is better to find some alternatives to replacement, refinement, and, reduction of animals. Nowadays, some novel technologies such as using imaging methods have been introduced.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Gilany
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Yarahmadi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Firoozeh Ghaderi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Improved Long-Term Volume Retention of Stromal Vascular Fraction Gel Grafting with Enhanced Angiogenesis and Adipogenesis. Plast Reconstr Surg 2018; 141:676e-686e. [PMID: 29334574 DOI: 10.1097/prs.0000000000004312] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The apoptosis of mature adipocytes after fat grafting can result in chronic inflammation, absorption, and fibrosis, leading to unpredictable outcomes. Selective elimination of mature adipocytes may result in better outcomes and a different underlying retention mode. The authors previously developed a mature adipocyte-free product, stromal vascular fraction gel, derived from lipoaspirate, which eliminates adipocytes and preserves the stromal vascular fraction. This study investigated the retention and regeneration mode of stromal vascular fraction gel grafting. METHODS Nude mice were grafted with human-derived stromal vascular fraction gel or Coleman fat. Detailed cellular events over 3 months were investigated histologically and immunohistochemically. RESULTS The retention rate 90 days after grafting was significantly higher for stromal vascular fraction gel grafts than for standard Coleman fat (82 ± 15 percent versus 42 ± 9 percent; p < 0.05). Histologic analysis suggested that, unlike Coleman fat grafts, stromal vascular fraction gel grafts did not include significant necrotic areas. Moreover, although adipose tissue regeneration was found in grafts of both groups, rapid angiogenesis and macrophage infiltration were observed at a very early stage after stromal vascular fraction gel grafting. The presence of small preadipocytes with multiple intracellular lipid droplets in stromal vascular fraction gel grafts on day 3 also suggested very early adipogenesis. Although some of the cells in the stromal vascular fraction survived in stromal vascular fraction gel grafts, most of the newly formed adipose tissue was host-derived. CONCLUSION Stromal vascular fraction gel has a high long-term retention rate and a unique adipose regeneration mode, involving prompt inflammation and infiltration of immune cells, stimulating rapid angiogenesis and inducing host cell-mediated adipogenesis.
Collapse
|
30
|
Liu W, Xie Y, Gao T, Huang F, Wang L, Ding L, Wang W, Liu S, Dai J, Wang B. Reflection and observation: cell-based screening failing to detect HBV in HUMSCs derived from HBV-infected mothers underscores the importance of more stringent donor eligibility to reduce risk of transmission of infectious diseases for stem cell-based medical products. Stem Cell Res Ther 2018; 9:177. [PMID: 29973264 PMCID: PMC6030788 DOI: 10.1186/s13287-018-0920-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/31/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
Abstract
Background In cell-based therapy, the transmission of communicable diseases imposes a substantial threat to recipients. In this study, we investigated whether cell-based screening could detect hepatitis B virus (HBV) in human umbilical cord-derived mesenchymal stem cells (HUMSCs) isolated from HBV-infected donors to understand the susceptibility of HUMSCs to HBV infection. Methods HBV assay was performed in HUMSCs derived from healthy and HBV-infected donors with enzyme-linked immunosorbent assay (ELISA), fluorescence quantitative PCR (FQ-PCR) assay, and droplet digital PCR (ddPCR) assay. Further, HBV DNA was assayed in HUMSCs derived from healthy donors after incubation with human sera containing a high titer of HBV using FQ-PCR. Results HBV antigen/antibody and DNA failed to be detected using ELISA, FQ-PCR, and ddPCR. After incubation with HBV infection sera, HBV DNA could be detected, but below the valid titer of the assay kit. The HBV DNA levels in HBV-incubated HUMSCs gradually decreased with medium change every 2 days and then significantly decreased, not even detected after passage. Conclusions The current cell-based screening methods could not detect HBV in HUMSCs derived from HBV-infected donors, indicating the importance of more stringent donor eligibility to reduce the risk of transmission of communicable diseases in cell-based therapy. To solve the problem of an occult HBV window period in donor eligibility determination, we recommend that the donors undergo another HBV serological test 3 months after the first serological communicable disease screening.
Collapse
Affiliation(s)
- Wei Liu
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yuanyuan Xie
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Tianyun Gao
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Feifei Huang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Liudi Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Lijun Ding
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Wenqing Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Shuo Liu
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jianwu Dai
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Beijing, 100190, China.
| | - Bin Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
31
|
Bedford P, Jy J, Collins L, Keizer S. Considering Cell Therapy Product "Good Manufacturing Practice" Status. Front Med (Lausanne) 2018; 5:118. [PMID: 29761103 PMCID: PMC5936751 DOI: 10.3389/fmed.2018.00118] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/09/2018] [Indexed: 11/17/2022] Open
Affiliation(s)
- Patrick Bedford
- Centre for Commercialization of Regenerative Medicine (CCRM), Toronto, ON, Canada
| | - Juliana Jy
- Centre for Commercialization of Regenerative Medicine (CCRM), Toronto, ON, Canada
| | - Lucas Collins
- Centre for Commercialization of Regenerative Medicine (CCRM), Toronto, ON, Canada
| | - Steven Keizer
- Centre for Commercialization of Regenerative Medicine (CCRM), Toronto, ON, Canada
| |
Collapse
|
32
|
Chhabra HS, Sarda K. Clinical translation of stem cell based interventions for spinal cord injury - Are we there yet? Adv Drug Deliv Rev 2017; 120:41-49. [PMID: 28964881 DOI: 10.1016/j.addr.2017.09.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/14/2017] [Accepted: 09/20/2017] [Indexed: 01/01/2023]
Abstract
Recent advances in basic science in research related to spinal cord injury (SCI) and regeneration have led to a variety of novel experimental therapeutics designed to promote functionally effective axonal regrowth and sprouting. Stem cell and other cellular interventions have gained lot of attention due to their immense potential of regeneration. These interventions have been tested for their efficacy in case of SCI both at the pre-clinical and clinical level. In this review we critically discuss the published literature on the cellular interventions for SCI and their clinical applications with respect to the strength of evidence established by these studies. The need to curb unethical practice of offering unproven stem cell "therapies" for SCI at a global level is also discussed.
Collapse
|
33
|
Abstract
Microparticles with controlled size and morphology are of significant interest in the field of drug delivery. Although advanced nanoparticles have been the object of a substantial number of reviews, fewer have focused on microparticles, especially for the delivery of drugs and growth factors to the wound site. Microparticles show distinct advantages, including ease of production and characterization, extended release properties, high drug loading and little concern about the toxicity as compared with the nanosized systems. This review presents an introduction to the pathophysiology of wound healing and provides an overview of some of the recent advances in microparticle-based drugs and growth factors delivery to wound sites.
Collapse
|
34
|
Gu Q, Wang J, Wang L, Liu ZX, Zhu WW, Tan YQ, Han WF, Wu J, Feng CJ, Fang JH, Liu L, Wang L, Li W, Zhao XY, Hu BY, Hao J, Zhou Q. Accreditation of Biosafe Clinical-Grade Human Embryonic Stem Cells According to Chinese Regulations. Stem Cell Reports 2017; 9:366-380. [PMID: 28506532 PMCID: PMC5511037 DOI: 10.1016/j.stemcr.2017.04.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022] Open
Abstract
Human embryonic stem cells (hESCs) are promising in regenerative medicine. Although several hESC-based clinical trials are under way, a widely accepted standard of clinical-grade cells remains obscure. To attain a completely xeno-free clinical-grade cell line, the system must be free of xenogenic components, the cells must have a comprehensive set of functions, and good manufacturing practice conditions must be used. In this study, following these criteria, we successfully derived two hESC lines, which were thereby considered “clinical-grade embryonic stem cells”. In addition to the primary capacity for pluripotency, these two cell lines were efficiently differentiated into various types of clinical-grade progeny. Importantly, the cells were recognized by the National Institutes for Food and Drug Control of China for further eligible accreditation. These data indicate that we have established completely xeno-free clinical-grade hESC lines and their derivatives, which will be valuable for the foundation of an international standard for clinical-grade cells for therapy. Two clinical-grade hESC lines were generated The hESCs were demonstrated to have sustained pluripotency The hESCs could be differentiated into functional cells The hESCs passed biosafety tests and were recognized by the NIFDC
Collapse
Affiliation(s)
- Qi Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Juan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng-Xin Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wan-Wan Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuan-Qing Tan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei-Fang Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chun-Jing Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jin-Hui Fang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Lei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Yang Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bao-Yang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Hao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
35
|
Mishra R, Bishop T, Valerio IL, Fisher JP, Dean D. The potential impact of bone tissue engineering in the clinic. Regen Med 2016; 11:571-87. [PMID: 27549369 DOI: 10.2217/rme-2016-0042] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bone tissue engineering (BTE) intends to restore structural support for movement and mineral homeostasis, and assist in hematopoiesis and the protective functions of bone in traumatic, degenerative, cancer, or congenital malformation. While much effort has been put into BTE, very little of this research has been translated to the clinic. In this review, we discuss current regenerative medicine and restorative strategies that utilize tissue engineering approaches to address bone defects within a clinical setting. These approaches involve the primary components of tissue engineering: cells, growth factors and biomaterials discussed briefly in light of their clinical relevance. This review also presents upcoming advanced approaches for BTE applications and suggests a probable workpath for translation from the laboratory to the clinic.
Collapse
Affiliation(s)
- Ruchi Mishra
- Department of Plastic Surgery, The Ohio State University, Columbus, OH, USA
| | - Tyler Bishop
- Department of Plastic Surgery, The Ohio State University, Columbus, OH, USA
| | - Ian L Valerio
- Department of Plastic Surgery, The Ohio State University, Columbus, OH, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - David Dean
- Department of Plastic Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
36
|
Shafiq M, Jung Y, Kim SH. Insight on stem cell preconditioning and instructive biomaterials to enhance cell adhesion, retention, and engraftment for tissue repair. Biomaterials 2016; 90:85-115. [PMID: 27016619 DOI: 10.1016/j.biomaterials.2016.03.020] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/09/2016] [Accepted: 03/13/2016] [Indexed: 12/13/2022]
Abstract
Stem cells are a promising solution for the treatment of a variety of diseases. However, the limited survival and engraftment of transplanted cells due to a hostile ischemic environment is a bottleneck for effective utilization and commercialization. Within this environment, the majority of transplanted cells undergo apoptosis prior to participating in lineage differentiation and cellular integration. Therefore, in order to maximize the clinical utility of stem/progenitor cells, strategies must be employed to increase their adhesion, retention, and engraftment in vivo. Here, we reviewed key strategies that are being adopted to enhance the survival, retention, and engraftment of transplanted stem cells through the manipulation of both the stem cells and the surrounding environment. We describe how preconditioning of cells or cell manipulations strategies can enhance stem cell survival and engraftment after transplantation. We also discuss how biomaterials can enhance the function of stem cells for effective tissue regeneration. Biomaterials can incorporate or mimic extracellular function (ECM) function and enhance survival or differentiation of transplanted cells in vivo. Biomaterials can also promote angiogenesis, enhance engraftment and differentiation, and accelerate electromechanical integration of transplanted stem cells. Insight gained from this review may direct the development of future investigations and clinical trials.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Korea University of Science and Technology, 176 Gajeong-dong, Yuseong-gu, Daejeon, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul 130-650, Republic of Korea
| | - Youngmee Jung
- Korea University of Science and Technology, 176 Gajeong-dong, Yuseong-gu, Daejeon, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul 130-650, Republic of Korea
| | - Soo Hyun Kim
- Korea University of Science and Technology, 176 Gajeong-dong, Yuseong-gu, Daejeon, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul 130-650, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea.
| |
Collapse
|
37
|
Kawase T, Hayama K, Tsuchimochi M, Nagata M, Okuda K, Yoshie H, Burns DM, Nakata K. Evaluating the Safety of Somatic Periosteal Cells by Flow-Cytometric Analysis Monitoring the History of DNA Damage. Biopreserv Biobank 2016; 14:129-37. [PMID: 26828697 DOI: 10.1089/bio.2015.0072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In preparing cell-based products for regenerative therapy, cell quality should be strictly controlled. Methodologies for evaluating cell viability, identity, and purity are established and used routinely, whereas current methodologies for evaluating cell safety, particularly genetic integrity or tumorigenicity, are time-consuming and relatively insensitive. As part of developing a more practical screening system, the authors previously demonstrated that γ-H2AX and p53 were useful markers for evaluating the history of DNA damage. To validate these markers further and develop a more quantitative methodology, single cell-based expression of these markers and two additional candidates have now been examined using flow cytometry (FCM). FCM analysis and immunofluorescent staining demonstrated that γ-ray-irradiation suppressed proliferation, enlarged cells, and cell nuclei, and immediately upregulated γ-H2AX and p21(waf1) in large numbers of cells for up to 12 days. Gamma-H2AX foci were formed in the nuclei of many affected cells. An initial sharp increase in p53 expression declined slowly over 12 days, while Rb expression increased linearly. The present findings suggest that this high-throughput, cell-based, combinational evaluation of protein markers and cell size enables a small number of cells with a history of DNA damage to be detected quickly and routinely from within a very large cell population. Using this screening methodology will improve the ability to verify the quality of cell-based products used in regenerative therapy.
Collapse
Affiliation(s)
- Tomoyuki Kawase
- 1 Division of Oral Bioengineering, Institute of Medicine and Dentistry, Niigata University , Niigata, Japan .,2 Advanced Research Center, The Nippon Dental University School of Life Dentistry at Niigata , Niigata, Japan
| | - Kazuhide Hayama
- 3 Department of Oral and Maxillofacial Radiology, The Nippon Dental University School of Life Dentistry at Niigata , Niigata, Japan
| | - Makoto Tsuchimochi
- 3 Department of Oral and Maxillofacial Radiology, The Nippon Dental University School of Life Dentistry at Niigata , Niigata, Japan
| | - Masaki Nagata
- 4 Division of Oral and Maxillofacial Surgery, Institute of Medicine and Dentistry, Niigata University , Niigata, Japan
| | - Kazuhiro Okuda
- 5 Division of Periodontology, Institute of Medicine and Dentistry, Niigata University , Niigata, Japan
| | - Hiromasa Yoshie
- 5 Division of Periodontology, Institute of Medicine and Dentistry, Niigata University , Niigata, Japan
| | - Douglas M Burns
- 6 Midwest Biomedical Research Foundation, Research Service, Department of Veterans Affairs Medical Center , Kansas City, Missouri
| | - Koh Nakata
- 7 Bioscience Medical Research Center, Niigata University Medical and Dental Hospital , Niigata, Japan
| |
Collapse
|
38
|
Banovic M, Loncar Z, Behfar A, Vanderheyden M, Beleslin B, Zeiher A, Metra M, Terzic A, Bartunek J. Endpoints in stem cell trials in ischemic heart failure. Stem Cell Res Ther 2015; 6:159. [PMID: 26319401 PMCID: PMC4552990 DOI: 10.1186/s13287-015-0143-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Despite multimodal regimens and diverse treatment options alleviating disease symptoms, morbidity and mortality associated with advanced ischemic heart failure remain high. Recently, technological innovation has led to the development of regenerative therapeutic interventions aimed at halting or reversing the vicious cycle of heart failure progression. Driven by the unmet patient need and fueled by encouraging experimental studies, stem cell-based clinical trials have been launched over the past decade. Collectively, these trials have enrolled several thousand patients and demonstrated the clinical feasibility and safety of cell-based interventions. However, the totality of evidence supporting their efficacy in ischemic heart failure remains limited. Experience from the early randomized stem cell clinical trials underscores the key points in trial design ranging from adequate hypothesis formulation to selection of the optimal patient population, cell type and delivery route. Importantly, to translate the unprecedented promise of regenerative biotherapies into clinical benefit, it is crucial to ensure the appropriate choice of endpoints along the regulatory path. Accordingly, we here provide considerations relevant to the choice of endpoints for regenerative clinical trials in the ischemic heart failure setting.
Collapse
Affiliation(s)
- Marko Banovic
- Cardiology Department, University Clinical Center of Serbia, Belgrade Medical School, 11000, Belgrade, Serbia.
| | - Zlatibor Loncar
- Cardiology Department, University Clinical Center of Serbia, Belgrade Medical School, 11000, Belgrade, Serbia.
| | | | | | - Branko Beleslin
- Cardiology Department, University Clinical Center of Serbia, Belgrade Medical School, 11000, Belgrade, Serbia.
| | - Andreas Zeiher
- Cardiology Department, Goethe University of Frankfurt, 60590, Frankfurt, Germany.
| | - Marco Metra
- Cardiology Department, University of Brescia, 25123, Brescia, Italy.
| | | | - Jozef Bartunek
- Cardiovascular Center, OLV Hospital, 9300, Aalst, Belgium.
| |
Collapse
|
39
|
|
40
|
Kanherkar RR, Bhatia-Dey N, Makarev E, Csoka AB. Cellular reprogramming for understanding and treating human disease. Front Cell Dev Biol 2014; 2:67. [PMID: 25429365 PMCID: PMC4228919 DOI: 10.3389/fcell.2014.00067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022] Open
Abstract
In the last two decades we have witnessed a paradigm shift in our understanding of cells so radical that it has rewritten the rules of biology. The study of cellular reprogramming has gone from little more than a hypothesis, to applied bioengineering, with the creation of a variety of important cell types. By way of metaphor, we can compare the discovery of reprogramming with the archeological discovery of the Rosetta stone. This stone slab made possible the initial decipherment of Egyptian hieroglyphics because it allowed us to see this language in a way that was previously impossible. We propose that cellular reprogramming will have an equally profound impact on understanding and curing human disease, because it allows us to perceive and study molecular biological processes such as differentiation, epigenetics, and chromatin in ways that were likewise previously impossible. Stem cells could be called “cellular Rosetta stones” because they allow also us to perceive the connections between development, disease, cancer, aging, and regeneration in novel ways. Here we present a comprehensive historical review of stem cells and cellular reprogramming, and illustrate the developing synergy between many previously unconnected fields. We show how stem cells can be used to create in vitro models of human disease and provide examples of how reprogramming is being used to study and treat such diverse diseases as cancer, aging, and accelerated aging syndromes, infectious diseases such as AIDS, and epigenetic diseases such as polycystic ovary syndrome. While the technology of reprogramming is being developed and refined there have also been significant ongoing developments in other complementary technologies such as gene editing, progenitor cell production, and tissue engineering. These technologies are the foundations of what is becoming a fully-functional field of regenerative medicine and are converging to a point that will allow us to treat almost any disease.
Collapse
Affiliation(s)
- Riya R Kanherkar
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Naina Bhatia-Dey
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Evgeny Makarev
- InSilico Medicine, Emerging Technology Center, Johns Hopkins University Eastern Baltimore, MD, USA
| | - Antonei B Csoka
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| |
Collapse
|
41
|
Kawase T, Kamiya M, Hayama K, Nagata M, Okuda K, Yoshie H, Burns DM, Tsuchimochi M, Nakata K. X-ray and ultraviolet C irradiation-induced γ-H2AX and p53 formation in normal human periosteal cells in vitro: markers for quality control in cell therapy. Cytotherapy 2014; 17:112-23. [PMID: 25293814 DOI: 10.1016/j.jcyt.2014.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 08/11/2014] [Accepted: 08/12/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND AIMS For successful cell transplantation therapy, the quality of cells must be strictly controlled. Unfortunately, to exclude inappropriate cells that possess structurally abnormal chromosomes, currently only karyotyping functions as an assessment. Unfortunately, this methodology is time-consuming and only effective for metaphasic cells. To develop a more efficient, inclusive and sensitive methodology, we examined the phosphorylation of histone H2AX and the p53 levels in normal human periosteal cells exposed to x-rays or other oxidative stressors. METHODS Periosteal cells were obtained from human alveolar bone before being exposed to x-rays, ultraviolet C or hydrogen peroxide. The cell cycle, electric nuclear volume and CD44 expression were evaluated using flow cytometry, and the phosphorylated H2AX (γ-H2AX), p53, p21 and proliferating cell nuclear antigen (PCNA) levels were evaluated by Western blot analyses. RESULTS Each oxidative stress dose-dependently arrested cell growth and partially induced premature cellular senescence. In parallel, each oxidative stress rapidly phosphorylated H2AX and stabilized p53, and intense stress sustained these high levels for at least 8 days. CONCLUSIONS Intensive oxidative stress induces sustained high levels of γ-H2AX and p53, which force cells toward senescence or non-apoptotic cell death. Lower doses of oxidative stress induced more modest and transient increases in γ-H2AX and p53, and these cells eventually survive. However, because DNA is repaired without a template in the majority of these cells, G1 mutations accumulate. Therefore, we recommend that any cell population expressing elevated γ-H2AX and p53 levels be excluded from cell transplantation therapy.
Collapse
Affiliation(s)
- Tomoyuki Kawase
- Division of Oral Bioengineering, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan; Advanced Research Center, The Nippon Dental University School of Life Dentistry at Niigata, Niigata, Japan.
| | - Mana Kamiya
- Division of Oral Bioengineering, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan; Division of Periodontology, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| | - Kazuhide Hayama
- Department of Oral and Maxillofacial Radiology, The Nippon Dental University School of Life Dentistry at Niigata, Niigata, Japan
| | - Masaki Nagata
- Division of Oral and Maxillofacial Surgery, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| | - Kazuhiro Okuda
- Division of Periodontology, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| | - Hiromasa Yoshie
- Division of Periodontology, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| | - Douglas M Burns
- Midwest Biomedical Research Foundation, Research Service, Department of Veterans Affairs Medical Center, Kansas City, Missouri, USA
| | - Makoto Tsuchimochi
- Department of Oral and Maxillofacial Radiology, The Nippon Dental University School of Life Dentistry at Niigata, Niigata, Japan
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| |
Collapse
|
42
|
Yuan BZ, Wang J. The regulatory sciences for stem cell-based medicinal products. Front Med 2014; 8:190-200. [PMID: 24733351 DOI: 10.1007/s11684-014-0323-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 12/28/2013] [Indexed: 11/25/2022]
Abstract
Over the past few years, several new achievements have been made from stem cell studies, many of which have moved up from preclinical stages to early, or from early to middle or late, stages thanks to relatively safe profile and preliminary evidence of effectiveness. Moreover, some stem cell-based products have been approved for marketing by different national regulatory authorities. However, many critical issues associated mainly with incomplete understanding of stem cell biology and the relevant risk factors, and lack of effective regulations still exist and need to be urgently addressed, especially in countries where establishment of appropriate regulatory system just commenced. More relevantly, the stem cell regulatory sciences need to be established or improved to more effectively evaluate quality, safety and efficacy of stem cell products, and for building up the appropriate regulatory framework. In this review, we summarize some new achievements in stem cell studies, especially the preclinical and clinical studies, the existing regulations, and the associated challenges, and we then propose some considerations for improving stem cell regulatory sciences with a goal of promoting the steadfast growth of the well-regulated stem cell therapies abreast of evolvement of stem cell sciences and technologies.
Collapse
Affiliation(s)
- Bao-Zhu Yuan
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | | |
Collapse
|
43
|
Trofin EA, Monsarrat P, Kémoun P. Cell therapy of periodontium: from animal to human? Front Physiol 2013; 4:325. [PMID: 24298258 PMCID: PMC3828527 DOI: 10.3389/fphys.2013.00325] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/19/2013] [Indexed: 12/30/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease affecting the soft and hard tissues supporting the teeth, which often leads to tooth loss. Its significant impact on the patient's general health and quality of life point to a need for more effective management of this condition. Existing treatments include scaling/root planning and surgical approaches but their overall effects are relatively modest and restricted in application. The goal of regenerative therapy of periodontal defects is to enhance endogenous progenitors and thus promote optimal wound healing. Considering that the host or tissue might be defective in the periodontitis context, it has been proposed that grafting exogenous stem cells would produce new tissues and create a suitable microenvironment for tissue regeneration. Thus, cell therapy of periodontium has been assessed in many animal models and promising results have been reported. However, the methodological diversity of these studies makes the conversion to clinical practice difficult. The aim of this review is to highlight the primary requirements to be satisfied before the leap to clinical trials can be made. We therefore review cell therapy applications for periodontal regeneration in animal models and the concerns to be addressed before undertaking human experiments.
Collapse
Affiliation(s)
- Elena A Trofin
- Department of Biology, Toulouse Faculty of Dentistry - Paul Sabatier University, Toulouse University Hospital - CHU de Toulouse Toulouse, France ; Department of Pathology, Faculty of Medicine and Odontology, University of Valencia Valencia, Spain
| | | | | |
Collapse
|
44
|
Abstract
Direct-to-consumer (DTC) advertising of suspect goods and services has burgeoned because of the Internet. Despite very limited approval for use, DTC stem cell-marketed "treatments" have emerged for an array of conditions, creating global public health and safety risks. However, it remains unclear whether such use of stem cells is subject to drugs or biologics regulations. To address this gap, regulatory agencies should be given clear authority, and the international community should create a framework for appropriate stem cell use. In addition, consumer protection laws should be used to scrutinize providers.
Collapse
Affiliation(s)
- Bryan A Liang
- Institute of Health Law Studies, California Western School of Law, San Diego, CA 92101, USA.
| | | |
Collapse
|
45
|
Peptidhydrogele für den Einsatz von Stammzellen in der regenerativen Medizin. GYNAKOLOGISCHE ENDOKRINOLOGIE 2012. [DOI: 10.1007/s10304-012-0493-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|