1
|
Terai S, Asonuma M, Hoshino A, Kino-oka M, Ochiya T, Okada K, Sato Y, Takahashi Y, Tobita M, Tsuchiya A. Guidance on the clinical application of extracellular vesicles. Regen Ther 2025; 29:43-50. [PMID: 40124467 PMCID: PMC11930518 DOI: 10.1016/j.reth.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Therapies using extracellular vesicles (EVs), exosomes, or cell culture supernatants containing EVs or exosomes (referred to as "EV therapies" in this Guidance) have garnered an increasing amount of interest. However, pharmaceutical products containing EVs as their main active ingredient are yet to receive regulatory approval. The "Basic points to consider regarding the preparation of extracellular vesicles and their clinical applications in Japan," was announced by the Japanese Society for Regenerative Medicine (JSRM) on March 10, 2021, with a specific focus on exosomes among EVs to promote high safety standards in this evolving field and facilitate the application of EV clinically. The Scientific Committee of the Pharmaceuticals and Medical Devices Agency and the Japanese Society for Extracellular Vesicles have issued reports and statements regarding treatment, resulting in a growing momentum toward treatment in Japan. This article summarized the basic items that should be recognized comprehensively for clinical practice in three categories: (1) risk profiling, (2) preparation (manufacturing) process and quality, and (3) verification of EVs checking items and effectiveness. This guideline will be revised in the future with technological innovations and new findings; nevertheless, it will serve as a guide for the development of treatments.
Collapse
Affiliation(s)
- Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- The Japanese Society for Regenerative Medicine, Japan1
| | - Motohiro Asonuma
- Visiting Professor Juntendo University (Information System Headquarters), Japan
- The Japanese Society for Regenerative Medicine, Japan1
| | - Ayuko Hoshino
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
- Research Base for Cell Manufacturability, Osaka University, Osaka, Japan
- The Japanese Society for Regenerative Medicine, Japan1
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Center for Future Medical Research, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
- The Japanese Society for Regenerative Medicine, Japan1
- The Japanese Society for Extracellular Vesicles, Japan2
| | - Kiyoshi Okada
- X-innovation Committee for Health and Medical, Osaka University Graduate School of Medicine, Osaka, Japan
- The Japanese Society for Regenerative Medicine, Japan1
| | - Yoji Sato
- Division of Drugs, National Institute of Health Sciences, Kanagawa, Japan
- The Japanese Society for Regenerative Medicine, Japan1
| | - Yuki Takahashi
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- The Japanese Society for Extracellular Vesicles, Japan2
| | - Morikuni Tobita
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
- The Japanese Society for Regenerative Medicine, Japan1
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Shimokato, 1110 Chuo, Yamanashi, Japan
- The Japanese Society for Regenerative Medicine, Japan1
- The Japanese Society for Extracellular Vesicles, Japan2
| |
Collapse
|
2
|
Gupta D, Shaz B. Surveying local CAR T-cell manufacturing processes to facilitate standardization and expand accessibility. J Transl Med 2025; 23:507. [PMID: 40329308 PMCID: PMC12057077 DOI: 10.1186/s12967-025-06400-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR T-cell) therapies have shown significant promise in treating cancers and other diseases. However, the manufacturing processes for CAR T-cell therapies exhibit considerable variability, which can affect treatment consistency and patient outcomes. While centralized manufacturing models dominate, local decentralized approaches, including point-of-care production, are being explored to address logistical and access challenges. This study aims to evaluate the current landscape of local CAR T-cell manufacturing at academic institutions. METHODS A comprehensive, cross-sectional survey was distributed to 130 FACT and/or JACIE accredited academic institutions globally. The survey, developed from semi-structured interviews with CAR T-cell manufacturing experts, assessed practices in cell modification methods, equipment protocols, and regulatory challenges. Data were analyzed using descriptive statistics, comparing responses across institutions and regions. RESULTS 45 of the 130 institutions (35 from the United States and 10 internationally, from the European Union, the United Kingdom, and Australia) responded to the survey (35% response rate). Of the 45 responding institutions, 40 were actively engaged or planning to engage in CAR T-cell production, while five had no plans to initiate manufacturing. Within the 40 institutions engaged in CAR T-cell production, 63% (25/40) reported active manufacturing, while 37% (15/40) were in the process of developing manufacturing capabilities. The most commonly reported barriers to local manufacturing were cost constraints (70%, 28/40), regulatory complexities (70%, 28/40), and facility requirements (57%, 17/40). Variability in product quality was cited by 73% (29/40) of institutions. Equipment costs and the need for specialized training emerged as major challenges, particularly for international institutions. Institutions also highlighted the need for automated platforms, with 60% (24/40) using the Miltenyi CliniMACS Prodigy and 50% (20/40) using the Lonza Cocoon. CONCLUSIONS This study highlights the widespread adoption of local CAR T-cell manufacturing and the significant variability in production processes across institutions. The findings emphasize the importance of establishing quality control benchmarks and data reporting frameworks to improve product consistency and access to CAR T-cell therapies. Addressing barriers such as cost, infrastructure, and regulatory challenges through standardization efforts and international collaboration could enhance the reproducibility, scalability, and accessibility of CAR T-cell therapies globally.
Collapse
Affiliation(s)
- Deven Gupta
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA.
- Margolis Institute for Health Policy, Duke University, Durham, NC, USA.
| | - Beth Shaz
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Margolis Institute for Health Policy, Duke University, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
3
|
Shen L, Li C, Li Y, Guan X, Zou W, Liu J. Imaging technology in tracking the intravital fate of transplanted stem cells. Pharmacol Res 2025; 216:107752. [PMID: 40306602 DOI: 10.1016/j.phrs.2025.107752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Stem cell therapy emerges as a promising alternative strategy for diseases that currently lack effective treatment options. Investigating the pharmacokinetic properties of stem cells, such as their survival, migration, differentiation, and engraftment dynamics, offers valuable insights for elucidating therapeutic mechanisms, refining treatment protocols, and ultimately enhancing therapeutic efficacy. Moreover, the pharmaceutical research of stem cell products is an essential prerequisite for regulatory approval. This contribution focus on the development of advanced imaging technologies for noninvasive monitoring the intravital fate of implanted stem cells, as well as the advantages and challenges of each imaging approach. Through comprehensive analysis of stem cell metabolic pathway, we identify critical barriers to clinical translation of stem cell therapy. In the end, we discuss future perspectives and opportunities in stem cell tracking and functional assessment.
Collapse
Affiliation(s)
- Liming Shen
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Chengze Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yulian Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xin Guan
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Wei Zou
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116023, China
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116023, China.
| |
Collapse
|
4
|
Chen KY, Chan HC, Chan CM. Can Stem Cell Therapy Revolutionize Ocular Disease Treatment? A Critical Review of Preclinical and Clinical Advances. Stem Cell Rev Rep 2025:10.1007/s12015-025-10884-x. [PMID: 40266467 DOI: 10.1007/s12015-025-10884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Stem cell therapy in regenerative medicine has a scope for treating ocular diseases. Stem cell therapy aims to repair damaged tissue and restore vision. The present review focuses on the advancements in stem cell therapies for ocular disorders, their mechanism of action, and clinical applications while addressing some outstanding challenges. Stem cells that include embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and retinal progenitor cells have regenerative potential for ocular repair. They differentiate into specialized ocular cell types, conduct neuroprotection, and modulate immune responses. It is emphasized in preclinical and clinical studies that stem cell therapy can treat corneal disorders such as limbal stem cell deficiency, retinal diseases like dry age macular degeneration and retinitis pigmentosa, and diabetic retinopathy. Various studies suggested that stem cells have considerable scope in glaucoma treatment by supporting retinal ganglion cell survival and optic nerve regeneration. Advanced approaches such as gene editing, organoid generation, and artificial intelligence enhance these therapies. Effective delivery to target areas, engraftment, orientation, and long-term survival of transplanted cells need optimization. Issues such as immune rejection and tumorigenicity must be addressed. This approach is further hindered by regulatory issues and overly complicated approval processes and trials. Ethical issues related to sourcing embryonic stem cells and patient consent complicate the issue. The cost of manufacturing stem cells and their accessibility are other factors posing potential barriers to widespread application. These regulatory, ethical, and economic issues must be tackled if stem cell treatments are to be made safe, accessible, and effective. Future studies will include refining therapeutic protocols, scaling manufacturing processes, and overcoming socio-economic barriers, eventually improving clinical outcomes.
Collapse
Affiliation(s)
- Kai-Yang Chen
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hoi-Chun Chan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chi-Ming Chan
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City, Taiwan.
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
5
|
Wang H, Ciccocioppo R, Terai S, Shoeibi S, Carnevale G, De Marchi G, Tsuchiya A, Ishii S, Tonouchi T, Furuyama K, Yang Y, Mito M, Abe H, Di Tinco R, Cardinale V. Targeted animal models for preclinical assessment of cellular and gene therapies in pancreatic and liver diseases: regulatory and practical insights. Cytotherapy 2025; 27:259-278. [PMID: 39755978 PMCID: PMC12068232 DOI: 10.1016/j.jcyt.2024.11.008] [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: 05/31/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 01/07/2025]
Abstract
Cellular and gene therapy (CGT) products have emerged as a popular approach in regenerative medicine, showing promise in treating various pancreatic and liver diseases in numerous clinical trials. Before these therapies can be tested in human clinical trials, it is essential to evaluate their safety and efficacy in relevant animal models. Such preclinical testing is often required to obtain regulatory approval for investigational new drugs. However, there is a lack of detailed guidance on selecting appropriate animal models for CGT therapies targeting specific pancreatic and liver conditions, such as pancreatitis and chronic liver diseases. In this review, the gastrointestinal committee for the International Society for Cell and Gene Therapy provides a summary of current recommendations for animal species and disease model selection, as outlined by the US Food and Drug Administration, with references to EU EMA and Japan PMDA. We discuss a range of small and large animal models, as well as humanized models, that are suitable for preclinical testing of CGT products aimed at treating pancreatic and liver diseases. For each model, we cover the associated pathophysiology, commonly used metrics for assessing disease status, the pros and limitations of the models, and the relevance of these models to human conditions. We also summarize the use and application of humanized mouse and other animal models in evaluating the safety and efficacy of CGT products. This review aims to provide comprehensive guidance for selecting appropriate animal species and models to help bridge the gap between the preclinical research and clinical trials using CGT therapies for specific pancreatic and liver diseases.
Collapse
Affiliation(s)
- Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph H Johnson Veteran Medical Center, Charleston, South Carolina, USA.
| | - Rachele Ciccocioppo
- Department of Medicine, Gastroenterology Unit, Pancreas Institute, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sara Shoeibi
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia De Marchi
- Department of Medicine, Gastroenterology Unit, Pancreas Institute, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Soichi Ishii
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takafumi Tonouchi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kaito Furuyama
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuan Yang
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaki Mito
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Rosanna Di Tinco
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Vincenzo Cardinale
- Department of Translational and Precision Medicine, University of Rome, Rome, Italy.
| |
Collapse
|
6
|
Patel D, Reese Koç J, Otegbeye F. Creating a GMP cell processing program: A focus on quality and regulation. Best Pract Res Clin Haematol 2025; 38:101614. [PMID: 40274340 DOI: 10.1016/j.beha.2025.101614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Implementing current Good Manufacturing Practice (GMP) regulations and principles even in early phases of cell-based therapy studies is crucial for ensuring safety and reproducible quality of these products. This paper outlines the comprehensive steps necessary to establish a robust GMP-compliant cell processing program in academic programs with emphases on adherence to regulatory and quality standards. While there are different regulatory agencies governing practice across the globe, the prevailing quality principles described here incorporate common requirements and guidelines from agencies such as the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA). The goal of this review is to provide guidance for developing a quality management program (QMP) that addresses all critical factors impacting each step in the cell therapy product lifecycle: from procurement and receipt of starter material, through manufacturing, testing, storage, distribution, and administration. The QMP should be designed to assure quality outcomes by maintaining qualified and trained staff at all levels as applicable to their job functions; establishing clear policies and procedures; ensuring the qualification of facilities and equipment; using qualified materials for human use; and providing a framework for detection of trends and implementing process improvement.
Collapse
Affiliation(s)
- Darshan Patel
- Therapeutic Products Program, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N, Seattle, WA, 98109, USA.
| | - Jane Reese Koç
- Cellular Therapy Service, Seidman Cancer Center Wesley Immunotherapy Center, 2103 Cornell Road, Cleveland, OH, 44106, USA.
| | - Folashade Otegbeye
- Translational Science and Therapeutics Division, Therapeutic Products Program, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N, Seattle, WA, 98109, USA.
| |
Collapse
|
7
|
Taei A, Sajadi FS, Salahi S, Enteshari Z, Falah N, Shiri Z, Abasalizadeh S, Hajizadeh-Saffar E, Hassani SN, Baharvand H. The cell replacement therapeutic potential of human pluripotent stem cells. Expert Opin Biol Ther 2025; 25:47-67. [PMID: 39679436 DOI: 10.1080/14712598.2024.2443079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION The remarkable ability of human pluripotent stem cells (hPSCs) to differentiate into specialized cells of the human body emphasizes their immense potential in treating various diseases. Advances in hPSC technology are paving the way for personalized and allogeneic cell-based therapies. The first-in-human studies showed improved treatment of diseases with no adverse effects, which encouraged the industrial production of this type of medicine. To ensure the quality, safety and efficacy of hPSC-based products throughout their life cycle, it is important to monitor and control their clinical translation through good practices (GxP) regulations. Understanding these rules in advance will help ensure that the industrial development of hPSC-derived products for widespread clinical implementation is feasible and progresses rapidly. AREAS COVERED In this review, we discuss the key translational obstacles of hPSCs, outline the current hPSC-based clinical trials, and present a workflow for putative clinical hPSC-based products. Finally, we highlight some future therapeutic opportunities for hPSC-derivatives. EXPERT OPINION hPSC-based products continue to show promise for the treatment of a variety of diseases. While clinical trials support the relative safety and efficacy of hPSC-based products, further investigation is required to explore the clinical challenges and achieve exclusive regulations for hPSC-based cell therapies.
Collapse
Affiliation(s)
- Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh-Sadat Sajadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Sarvenaz Salahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Enteshari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasrin Falah
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Shiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Abasalizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
8
|
Grattoni A, Korbutt G, Tomei AA, García AJ, Pepper AR, Stabler C, Brehm M, Papas K, Citro A, Shirwan H, Millman JR, Melero-Martin J, Graham M, Sefton M, Ma M, Kenyon N, Veiseh O, Desai TA, Nostro MC, Marinac M, Sykes M, Russ HA, Odorico J, Tang Q, Ricordi C, Latres E, Mamrak NE, Giraldo J, Poznansky MC, de Vos P. Harnessing cellular therapeutics for type 1 diabetes mellitus: progress, challenges, and the road ahead. Nat Rev Endocrinol 2025; 21:14-30. [PMID: 39227741 PMCID: PMC11938328 DOI: 10.1038/s41574-024-01029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a growing global health concern that affects approximately 8.5 million individuals worldwide. T1DM is characterized by an autoimmune destruction of pancreatic β cells, leading to a disruption in glucose homeostasis. Therapeutic intervention for T1DM requires a complex regimen of glycaemic monitoring and the administration of exogenous insulin to regulate blood glucose levels. Advances in continuous glucose monitoring and algorithm-driven insulin delivery devices have improved the quality of life of patients. Despite this, mimicking islet function and complex physiological feedback remains challenging. Pancreatic islet transplantation represents a potential functional cure for T1DM but is hindered by donor scarcity, variability in harvested cells, aggressive immunosuppressive regimens and suboptimal clinical outcomes. Current research is directed towards generating alternative cell sources, improving transplantation methods, and enhancing cell survival without chronic immunosuppression. This Review maps the progress in cell replacement therapies for T1DM and outlines the remaining challenges and future directions. We explore the state-of-the-art strategies for generating replenishable β cells, cell delivery technologies and local targeted immune modulation. Finally, we highlight relevant animal models and the regulatory aspects for advancing these technologies towards clinical deployment.
Collapse
Affiliation(s)
- Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| | - Gregory Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrew R Pepper
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Cherie Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Michael Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Klearchos Papas
- Department of Surgery, The University of Arizona, Tucson, AZ, USA
| | - Antonio Citro
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Haval Shirwan
- Department of Pediatrics, Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Juan Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Melanie Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Michael Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Norma Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Tejal A Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA
- Brown University, School of Engineering, Providence, RI, USA
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Megan Sykes
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University, New York, NY, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jon Odorico
- UW Health Transplant Center, Madison, WI, USA
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA, US
- Gladstone Institute of Genomic Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Esther Latres
- Research Department, Breakthrough T1D, New York, NY, USA
| | | | - Jaime Giraldo
- Research Department, Breakthrough T1D, New York, NY, USA.
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
9
|
Mishra P, Biesiada I, Gupta P, Ghavami S, Markowski J, Łos MJ. Unraveling the Complexity and Advancements of Transdifferentiation Technologies in the Biomedical Field and Their Potential Clinical Relevance. Arch Immunol Ther Exp (Warsz) 2025; 73:aite-2025-0001. [PMID: 39637369 DOI: 10.2478/aite-2025-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Chronic diseases such as cancer, autoimmunity, and organ failure currently depend on conventional pharmaceutical treatment, which may cause detrimental side effects in the long term. In this regard, cell-based therapy has emerged as a suitable alternative for treating these chronic diseases. Transdifferentiation technologies have evolved as a suitable therapeutic alternative that converts one differentiated somatic cell into another phenotype by using transcription factors (TFs), small molecules, or small, single-stranded, non-coding RNA molecules (miRNA). The transdifferentiation techniques rely on simple, fast, standardized, and versatile protocols with minimal chance of tumorigenicity and genotoxicity. However, there are still challenges and limitations that need to be addressed to enhance their clinical translation percentage in the near future. Taking this into account, we have delineated the features and strategies used in the transdifferentiation techniques. Then, we delved into different intermediate states that were attained during transdifferentiation. Advancements in transdifferentiation techniques in the field of tissue engineering, autoimmunity, and cancer therapy were dissected. Furthermore, limitations, challenges, and future perspectives are outlined in this review to provide a whole new picture of the transdifferentiation techniques. Advancements in molecular biology, interdisciplinary research, bioinformatics, and artificial intelligence will push the frontiers of this technology further to establish new avenues for biomedical research.
Collapse
Affiliation(s)
- Purusottam Mishra
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Izabella Biesiada
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Payal Gupta
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland
| | - Jarosław Markowski
- Department of Laryngology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
10
|
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
|
11
|
Yasuda S, Bando K, Henry MP, Libertini S, Watanabe T, Bando H, Chen C, Fujimori K, Harada K, Kuroda T, Lemmens M, Marginean D, Moss D, Pereira Mouriès L, Nicholas NS, Smart MJK, Terai O, Sato Y. Detection of residual pluripotent stem cells in cell therapy products utilizing droplet digital PCR: an international multisite evaluation study. Stem Cells Transl Med 2024; 13:1001-1014. [PMID: 39120125 PMCID: PMC11465167 DOI: 10.1093/stcltm/szae058] [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: 02/28/2024] [Accepted: 06/08/2024] [Indexed: 08/10/2024] Open
Abstract
The presence of residual undifferentiated pluripotent stem cells (PSCs) in PSC-derived cell therapy products (CTPs) is a major safety issue for their clinical application, due to the potential risk of PSC-derived tumor formation. An international multidisciplinary multisite study to evaluate a droplet digital PCR (ddPCR) approach to detect residual undifferentiated PSCs in PSC-derived CTPs was conducted as part of the Health and Environmental Sciences Institute Cell Therapy-TRAcking, Circulation & Safety Technical Committee. To evaluate the use of ddPCR in quantifying residual iPSCs in a cell sample, different quantities of induced pluripotent stem cells (iPSCs) were spiked into a background of iPSC-derived cardiomyocytes (CMs) to mimic different concentrations of residual iPSCs. A one step reverse transcription ddPCR (RT-ddPCR) was performed to measure mRNA levels of several iPSC-specific markers and to evaluate the assay performance (precision, sensitivity, and specificity) between and within laboratories. The RT-ddPCR assay variability was initially assessed by measuring the same RNA samples across all participating facilities. Subsequently, each facility independently conducted the entire process, incorporating the spiking step, to discern the parameters influencing potential variability. Our results show that a RT-ddPCR assay targeting ESRG, LINC00678, and LIN28A genes offers a highly sensitive and robust detection of impurities of iPSC-derived CMs and that the main contribution to variability between laboratories is the iPSC-spiking procedure, and not the RT-ddPCR. The RT-ddPCR assay would be generally applicable for tumorigenicity evaluation of PSC-derived CTPs with appropriate marker genes suitable for each CTP.
Collapse
Affiliation(s)
- Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | | | | | | | | | | | - Connie Chen
- Health and Environmental Sciences Institute, Washington, DC, United States
| | | | - Kosuke Harada
- Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | | | | | - David Moss
- Cell and Gene Therapy Catapult, London, United Kingdom
| | | | | | | | | | - Yoji Sato
- Division of Drugs, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
12
|
Shan Y, Zhang M, Tao E, Wang J, Wei N, Lu Y, Liu Q, Hao K, Zhou F, Wang G. Pharmacokinetic characteristics of mesenchymal stem cells in translational challenges. Signal Transduct Target Ther 2024; 9:242. [PMID: 39271680 PMCID: PMC11399464 DOI: 10.1038/s41392-024-01936-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/15/2024] Open
Abstract
Over the past two decades, mesenchymal stem/stromal cell (MSC) therapy has made substantial strides, transitioning from experimental clinical applications to commercial products. MSC therapies hold considerable promise for treating refractory and critical conditions such as acute graft-versus-host disease, amyotrophic lateral sclerosis, and acute respiratory distress syndrome. Despite recent successes in clinical and commercial applications, MSC therapy still faces challenges when used as a commercial product. Current detection methods have limitations, leaving the dynamic biodistribution, persistence in injured tissues, and ultimate fate of MSCs in patients unclear. Clarifying the relationship between the pharmacokinetic characteristics of MSCs and their therapeutic effects is crucial for patient stratification and the formulation of precise therapeutic regimens. Moreover, the development of advanced imaging and tracking technologies is essential to address these clinical challenges. This review provides a comprehensive analysis of the kinetic properties, key regulatory molecules, different fates, and detection methods relevant to MSCs and discusses concerns in evaluating MSC druggability from the perspective of integrating pharmacokinetics and efficacy. A better understanding of these challenges could improve MSC clinical efficacy and speed up the introduction of MSC therapy products to the market.
Collapse
Affiliation(s)
- Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Mengying Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Enxiang Tao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Ning Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Yi Lu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qing Liu
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Kun Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
13
|
Raniga K, Nasir A, Vo NTN, Vaidyanathan R, Dickerson S, Hilcove S, Mosqueira D, Mirams GR, Clements P, Hicks R, Pointon A, Stebbeds W, Francis J, Denning C. Strengthening cardiac therapy pipelines using human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2024; 31:292-311. [PMID: 38366587 DOI: 10.1016/j.stem.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/27/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024]
Abstract
Advances in hiPSC isolation and reprogramming and hPSC-CM differentiation have prompted their therapeutic application and utilization for evaluating potential cardiovascular safety liabilities. In this perspective, we showcase key efforts toward the large-scale production of hiPSC-CMs, implementation of hiPSC-CMs in industry settings, and recent clinical applications of this technology. The key observations are a need for traceable gender and ethnically diverse hiPSC lines, approaches to reduce cost of scale-up, accessible clinical trial datasets, and transparent guidelines surrounding the safety and efficacy of hiPSC-based therapies.
Collapse
Affiliation(s)
- Kavita Raniga
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK.
| | - Aishah Nasir
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nguyen T N Vo
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | | - Diogo Mosqueira
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter Clements
- Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London WC2R 2LS, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | | | - Jo Francis
- Mechanstic Biology and Profiling, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Chris Denning
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|