1
|
Matiukhova M, Ryapolova A, Andriianov V, Reshetnikov V, Zhuravleva S, Ivanov R, Karabelsky A, Minskaia E. A comprehensive analysis of induced pluripotent stem cell (iPSC) production and applications. Front Cell Dev Biol 2025; 13:1593207. [PMID: 40406420 PMCID: PMC12095295 DOI: 10.3389/fcell.2025.1593207] [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: 03/13/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
The ability to reprogram mature, differentiated cells into induced pluripotent stem cells (iPSCs) using exogenous pluripotency factors opened up unprecedented opportunities for their application in biomedicine. iPSCs are already successfully used in cell and regenerative therapy, as various drug discovery platforms and for in vitro disease modeling. However, even though already 20 years have passed since their discovery, the production of iPSC-based therapies is still associated with a number of hurdles due to low reprogramming efficiency, the complexity of accurate characterization of the resulting colonies, and the concerns associated with the safety of this approach. However, significant progress in many areas of molecular biology facilitated the production, characterization, and thorough assessment of the safety profile of iPSCs. The number of iPSC-based studies has been steadily increasing in recent years, leading to the accumulation of significant knowledge in this area. In this review, we aimed to provide a comprehensive analysis of methods used for reprogramming and subsequent characterization of iPSCs, discussed barriers towards achieving these goals, and various approaches to improve the efficiency of reprogramming of different cell populations. In addition, we focused on the analysis of iPSC application in preclinical and clinical studies. The accumulated breadth of data helps to draw conclusions about the future of this technology in biomedicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ekaterina Minskaia
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
2
|
Jiang Q, Yu W, Ma J, Zhao M, Zou J, Mir S, Zhang J, Germain RN, Hassan R. Robust differentiation of NK cells from MSLN.CAR-IL-15-engineered human iPSCs with enhanced antitumor efficacy against solid tumors. SCIENCE ADVANCES 2025; 11:eadt9932. [PMID: 40315330 PMCID: PMC12047432 DOI: 10.1126/sciadv.adt9932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Human induced pluripotent stem cells (iPSCs) offer a promising source for chimeric antigen receptor (CAR)-engineered natural killer (NK) products. However, complex iPSC-NK (iNK) manufacturing challenges clinical use. Here, we identified LiPSC-GR1.1 as a superior iPSC line for iNK production. By engineering LiPSC-GR1.1 with a mesothelin (MSLN)-targeting CAR and interleukin-15 (IL-15), we achieved robust differentiation of iPSCs into mature activated iNK cells with enhanced tumor killing efficacy, superior tumor homing, and vigorous proliferation. Single-cell transcriptomic analysis revealed that transforming growth factor-β (TGF-β)-producing tumor cells up-regulated major histocompatibility complex molecules and down-regulated MSLN post-CAR-IL-15 iNK treatment. Tumor-infiltrating CAR-IL-15 iNK cells exhibited high levels of CAR, IL-15, and NK-activating receptors, negligible checkpoint exhaustion markers, and extremely low levels of NK suppressive factors CISH, TGFBR2, and BATF, enabling them to sustain activation, metabolic fitness, and effective tumor killing within TGF-β-rich hypoxic tumor microenvironment. Overall, we developed MSLN.CAR-IL-15-engineered GR1.1-iNK therapy with enhanced antitumor efficacy for solid tumor treatment.
Collapse
Affiliation(s)
- Qun Jiang
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Weiming Yu
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging, NIAID and NCI, NIH, Bethesda, MD, USA
| | - James Ma
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mingming Zhao
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jizhong Zou
- iPSC Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Sameer Mir
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jingli Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging, NIAID and NCI, NIH, Bethesda, MD, USA
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
3
|
Soundararajan L, Surendran H, Patlolla N, Battu R, Stoddard J, Arrizabalaga S, Liu Z, Lingam G, Su X, Ryals RC, Pal R. Allogeneic RPE cell suspension manufactured at scale demonstrating preclinical safety and efficacy led to IND approval. NPJ Regen Med 2025; 10:19. [PMID: 40253438 PMCID: PMC12009284 DOI: 10.1038/s41536-025-00407-0] [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: 04/08/2024] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
Cell replacement therapy is a promising therapeutic option for dry age-related macular degeneration (AMD). In this study, we outline our design for scalable manufacture with appropriate quality gates and present in vivo data for establishing preclinical safety and efficacy of an induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) product, thus laying the foundation for Phase 1/2a trial approval in India (ClinicalTrials.gov ID: NCT06394232; date of registration: 23rd September 2024). Escalating doses of RPE cell suspension in immunocompromised animals demonstrated absence of tumor formation up to 9 months post-injection. Good Laboratory Practices (GLP) toxicology and tolerability studies in rabbits and non-human primates (NHP) respectively showed no major adverse events. RPE transplanted into immune suppressed RCS rats showed integration, neuroprotection and rescue of visual function. In addition, we provide a detailed description of the modifications in GMP manufacturing protocol to create a final product with a unique composition and Chemistry, Manufacturing and Controls (CMC) studies performed during product development.
Collapse
Affiliation(s)
- Lalitha Soundararajan
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru, India
| | - Harshini Surendran
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru, India
| | - Niharika Patlolla
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru, India
| | - Rajani Battu
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru, India
| | - Jonathan Stoddard
- Casey Eye Institute, Oregon Health and Science University (OHSU), Portland, Oregon, USA
| | | | - Zengping Liu
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Gopal Lingam
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University Hospital, Singapore, Singapore
| | - Xinyi Su
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University Hospital, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Renee C Ryals
- Casey Eye Institute, Oregon Health and Science University (OHSU), Portland, Oregon, USA
| | - Rajarshi Pal
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru, India.
- The University of Transdisciplinary Health Sciences and Technology (TDU), Bengaluru, India.
| |
Collapse
|
4
|
Ji S, Jin C, Cui X. Enhancing the physiological characteristics of chimeric antigen receptor natural killer cells by synthetic biology. Front Immunol 2025; 16:1592121. [PMID: 40313937 PMCID: PMC12043574 DOI: 10.3389/fimmu.2025.1592121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Chimeric antigen receptor natural Killer (CAR-NK) cells therapy represents a next-generation immunotherapeutic approach following CAR-T cells therapy, offering inherent "off-the-shelf" compatibility and mitigated off-tumor toxicity. Despite these advantages, clinical translation remains constrained by poor in vivo persistence and functional exhaustion in immunosuppressive tumor microenvironments (TME). This review examines recent advancements in synthetic biology aimed at enhancing the physiological characteristics of CAR-NK cells. By delineating the synergy between NK cells and synthetic biology toolkits, this work provides a roadmap for developing next-generation CAR-NK therapies capable of addressing solid tumor challenges while maintaining favorable safety profiles.
Collapse
Affiliation(s)
- Shuochao Ji
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Cheng Jin
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xinjiang Cui
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
5
|
Tiper Y, Xie Z, Hofemeier A, Lad H, Luber M, Krawetz R, Betz T, Zimmermann WH, Morton AB, Segal SS, Gilbert PM. Optimizing electrical field stimulation parameters reveals the maximum contractile function of human skeletal muscle microtissues. Am J Physiol Cell Physiol 2025; 328:C1160-C1176. [PMID: 40019026 DOI: 10.1152/ajpcell.00308.2024] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Skeletal muscle microtissues are engineered to develop therapies for restoring muscle function in patients. However, optimal electrical field stimulation (EFS) parameters to evaluate the function of muscle microtissues remain unestablished. This study reports a protocol to optimize EFS parameters for eliciting contractile force of muscle microtissues cultured in micropost platforms. Muscle microtissues were produced across an opposing pair of microposts in polydimethylsiloxane and polymethyl methacrylate culture platforms using primary, immortalized, and induced pluripotent stem cell-derived myoblasts. In response to EFS between needle electrodes, contraction deflects microposts proportional to developed force. At 5 V, pulse durations used for native muscle (0.1-1 ms) failed to elicit contraction of microtissues; durations reported for engineered muscle (5-10 ms) failed to elicit peak force. Instead, pulse durations of 20-80 ms were required to elicit peak twitch force across microtissues derived from five myoblast lines. Similarly, although peak tetanic force occurs at 20-50 Hz for native human muscles, it varied across microtissues depending on the cell line type, ranging from 7 to 60 Hz. A new parameter, the dynamic oscillation of force, captured trends during rhythmic contractions, whereas quantifying the duration-at-peak force provides an extended kinetics parameter. Our findings indicate that muscle microtissues have cell line type-specific contractile properties, yet all contract and relax more slowly than native muscle, implicating underdeveloped excitation-contraction coupling. Failure to optimize EFS parameters can mask the functional potential of muscle microtissues by underestimating force production. Optimizing and reporting EFS parameters and metrics is necessary to leverage muscle microtissues for advancing skeletal muscle therapies.NEW & NOTEWORTHY Electrical field stimulation (EFS) parameters remain to be standardized for engineered skeletal muscle. Herein, we report a protocol for defining EFS parameters that elicit the maximal contractile force of muscle microtissues cultivated in micropost devices and highlight the value of developing appropriate metrics. The dynamic oscillation of force and duration-at-peak force are introduced as novel metrics of contraction kinetics.
Collapse
Affiliation(s)
- Yekaterina Tiper
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Zhuoye Xie
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Arne Hofemeier
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Lower Saxony, Göttingen, Germany
| | - Heta Lad
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Mattias Luber
- Third Institute of Physics, University of Göttingen, Göttingen, Germany
| | - Roman Krawetz
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Timo Betz
- Third Institute of Physics, University of Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Lower Saxony, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- German Center for Child and Adolescent Health (DZKJ), Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
| | - Aaron B Morton
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, Texas, United States
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Martins F, Ribeiro MHL. Quality and Regulatory Requirements for the Manufacture of Master Cell Banks of Clinical Grade iPSCs: The EU and USA Perspectives. Stem Cell Rev Rep 2025; 21:645-679. [PMID: 39821060 DOI: 10.1007/s12015-024-10838-9] [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] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) and protocols for their differentiation into various cell types have revolutionized the field of tissue engineering and regenerative medicine. Developing manufacturing guidelines for safe and GMP-compliant final products has become essential. Allogeneic iPSCs-derived cell therapies are now the preferred manufacturing alternative. This option requires the establishment of clinical-grade master cell banks of iPSCs. This study aimed at reviewing the Quality and Regulatory requirements from the two main authorities in the world-Europe (EMA) and the United States (FDA)-regarding the manufacture of clinical grade master cell banks (iPSCs). The minimum requirements for iPSCs to be used in first-in-human clinical trials were also reviewed, as well as current best practices currently followed by iPSC bank manufacturers for final product characterisation. The methodology used for this work was a review of various sources of information ranging from scientific literature, published guidance documents available on the EMA and FDA websites, GMP and ICH guidelines, and applicable compendial monographs. Manufacturers of iPSCs cell banks looking to qualify them for clinical use are turning to the ICH guidelines and trying to adapt their requirements. Specifically with the impact of the field of iPSC cell banks, the following areas should be subject to guidance and harmonisation: i) expression vectors authorized for iPSC generation; ii) minimum identity testing; iii) minimum purity testing (including adventitious agent testing); and iv) stability testing. Current ICH guidelines for biotechnological/biological products should be extended to cover cell banks used for cell therapies.
Collapse
Affiliation(s)
- Fernando Martins
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
- Stemamatters S.A., 4805-017, Guimarães, Portugal
| | - Maria H L Ribeiro
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
- Research Institute for Medicines (i-Med.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
| |
Collapse
|
7
|
Estrada A, Furmanski O, Klarmann GJ, Scheidt N, Ho VB. Progress in Development of Functional Biological and Synthetic Blood Products to Augment Transfusable Blood Supply in Operational Medicine. Bioengineering (Basel) 2025; 12:256. [PMID: 40150720 PMCID: PMC11939705 DOI: 10.3390/bioengineering12030256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
A reliable, accessible, and high-quality blood supply is critical for the sustainment of any healthcare system. World events such as the COVID-19 pandemic have proven that maintaining the supply of blood presents a logistical challenge. The current blood supply is overseen by extensive donor programs around the world. In the United States, as in other countries, the need for blood has increased, with a decline in blood donations and increasing exclusions for blood donor qualification. While there is a need to improve blood donation participation, there is also need for new alternatives to traditional donation to ensure readiness to treat hemorrhagic shock common in the setting of trauma, as often occurs during a natural disaster or conflict. These operational medicine scenarios require significant blood availability which may tax the current blood supply chain. Aside from a walking blood bank (WBB) model for blood collection in suboptimal conditions, researchers have proposed alternatives for blood that include the manufacturing of blood from stem cell sources. Other alternatives include synthetic liquids that can carry oxygen such as Perfluoro-Chemicals (PFCs) and hemoglobin-based oxygen-carrying systems (HBCOs). Here, we review some of these alternatives to the traditional donor blood model. Researchers now have the technology that makes it feasible to develop blood alternatives that one day may supplement and help alleviate the limitations in blood supply.
Collapse
Affiliation(s)
- Armando Estrada
- 4D Bio Center for Biotechnology, Department of Radiology and Bioengineering, Uniformed Services University of the Health Sciences, Rockville, MD 20850, USA; (A.E.III)
- The Geneva Foundation, Bethesda, MD 20817, USA
| | - Orion Furmanski
- 4D Bio Center for Biotechnology, Department of Radiology and Bioengineering, Uniformed Services University of the Health Sciences, Rockville, MD 20850, USA; (A.E.III)
- The Geneva Foundation, Bethesda, MD 20817, USA
| | - George J. Klarmann
- 4D Bio Center for Biotechnology, Department of Radiology and Bioengineering, Uniformed Services University of the Health Sciences, Rockville, MD 20850, USA; (A.E.III)
- The Geneva Foundation, Bethesda, MD 20817, USA
| | - Nathan Scheidt
- The United States Air Force Academy, USAF Academy, CO 80840, USA
| | - Vincent B. Ho
- 4D Bio Center for Biotechnology, Department of Radiology and Bioengineering, Uniformed Services University of the Health Sciences, Rockville, MD 20850, USA; (A.E.III)
- The Geneva Foundation, Bethesda, MD 20817, USA
| |
Collapse
|
8
|
Jebran AF, Seidler T, Tiburcy M, Daskalaki M, Kutschka I, Fujita B, Ensminger S, Bremmer F, Moussavi A, Yang H, Qin X, Mißbach S, Drummer C, Baraki H, Boretius S, Hasenauer C, Nette T, Kowallick J, Ritter CO, Lotz J, Didié M, Mietsch M, Meyer T, Kensah G, Krüger D, Sakib MS, Kaurani L, Fischer A, Dressel R, Rodriguez-Polo I, Stauske M, Diecke S, Maetz-Rensing K, Gruber-Dujardin E, Bleyer M, Petersen B, Roos C, Zhang L, Walter L, Kaulfuß S, Yigit G, Wollnik B, Levent E, Roshani B, Stahl-Henning C, Ströbel P, Legler T, Riggert J, Hellenkamp K, Voigt JU, Hasenfuß G, Hinkel R, Wu JC, Behr R, Zimmermann WH. Engineered heart muscle allografts for heart repair in primates and humans. Nature 2025; 639:503-511. [PMID: 39880949 PMCID: PMC11903342 DOI: 10.1038/s41586-024-08463-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 11/27/2024] [Indexed: 01/31/2025]
Abstract
Cardiomyocytes can be implanted to remuscularize the failing heart1-7. Challenges include sufficient cardiomyocyte retention for a sustainable therapeutic impact without intolerable side effects, such as arrhythmia and tumour growth. We investigated the hypothesis that epicardial engineered heart muscle (EHM) allografts from induced pluripotent stem cell-derived cardiomyocytes and stromal cells structurally and functionally remuscularize the chronically failing heart without limiting side effects in rhesus macaques. After confirmation of in vitro and in vivo (nude rat model) equivalence of the newly developed rhesus macaque EHM model with a previously established Good Manufacturing Practice-compatible human EHM formulation8, long-term retention (up to 6 months) and dose-dependent enhancement of the target heart wall by EHM grafts constructed from 40 to 200 million cardiomyocytes/stromal cells were demonstrated in macaques with and without myocardial infarction-induced heart failure. In the heart failure model, evidence for EHM allograft-enhanced target heart wall contractility and ejection fraction, which are measures for local and global heart support, was obtained. Histopathological and gadolinium-based perfusion magnetic resonance imaging analyses confirmed cell retention and functional vascularization. Arrhythmia and tumour growth were not observed. The obtained feasibility, safety and efficacy data provided the pivotal underpinnings for the approval of a first-in-human clinical trial on tissue-engineered heart repair. Our clinical data confirmed remuscularization by EHM implantation in a patient with advanced heart failure.
Collapse
Affiliation(s)
- Ahmad-Fawad Jebran
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Tim Seidler
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Department of Cardiology, Campus Kerckhoff of the Justus-Liebig-Universität Gießen, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Malte Tiburcy
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Maria Daskalaki
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Ingo Kutschka
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Buntaro Fujita
- Clinic for Cardiac and Thoracic Vascular Surgery, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site North, Lübeck, Germany
| | - Stephan Ensminger
- Clinic for Cardiac and Thoracic Vascular Surgery, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site North, Lübeck, Germany
| | - Felix Bremmer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Amir Moussavi
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Functional Imaging Laboratory, German Primate Center, Göttingen, Germany
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xulei Qin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sophie Mißbach
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Charis Drummer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Hassina Baraki
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Susann Boretius
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Functional Imaging Laboratory, German Primate Center, Göttingen, Germany
| | - Christopher Hasenauer
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Nette
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Johannes Kowallick
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Christian O Ritter
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Joachim Lotz
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Didié
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Mathias Mietsch
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Tim Meyer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - George Kensah
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Dennis Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Md Sadman Sakib
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Andre Fischer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Ralf Dressel
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Ignacio Rodriguez-Polo
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Michael Stauske
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sebastian Diecke
- Pluripotent Stem Cells Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Kerstin Maetz-Rensing
- Pathology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Eva Gruber-Dujardin
- Pathology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Martina Bleyer
- Pathology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Beatrix Petersen
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Christian Roos
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Liye Zhang
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Lutz Walter
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Silke Kaulfuß
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Gökhan Yigit
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Bernd Wollnik
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Elif Levent
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Berit Roshani
- Unit of Infection Models, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Christiane Stahl-Henning
- Unit of Infection Models, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Legler
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Joachim Riggert
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Kristian Hellenkamp
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens-Uwe Voigt
- Department of Cardiovascular Sciences, Catholic University of Leuven and Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Gerd Hasenfuß
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Rabea Hinkel
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rüdiger Behr
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany.
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany.
| |
Collapse
|
9
|
Nakamura M, Tanaka Y, Hakoda K, Ohira M, Kobayashi T, Kurachi K, Tamura K, Ohdan H. Antitumor effects of natural killer cells derived from gene-engineered human-induced pluripotent stem cells on hepatocellular carcinoma. Cancer Immunol Immunother 2025; 74:99. [PMID: 39904787 PMCID: PMC11794780 DOI: 10.1007/s00262-025-03940-5] [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: 07/19/2024] [Accepted: 01/01/2025] [Indexed: 02/06/2025]
Abstract
Mortality and recurrence rates of hepatocellular carcinoma (HCC) remain high despite the use of various treatment methods. Recently, cell-based immunotherapy using natural killer (NK) cells has attracted considerable attention in cancer immunotherapy. NK cells generated from induced pluripotent stem cells (iPSCs) are a new option for use as an NK cell resource. The eNK cells (HLCN061, developed by HEALIOS K.K.) are human iPSC-derived NK cells differentiated from clinical-grade iPSCs in which IL-15, CCR2B, CCL19, CD16a, and NKG2D have been introduced. In this study, we aimed to evaluate the potential of eNK cell therapy for HCC treatment. The analysis of eNK cells for cell surface and intracellular molecules revealed that antitumor-related surface molecules (TRAIL, CD226, and CD16) and intracellular cytotoxic factors (perforin, granzyme B, TNFα, and IFNγ) were highly expressed. In addition, eNK cells exhibited high cytotoxicity against HCC cell lines (HepG2, HuH7, and SNU-423), which are sensitive to NKG2D, TRAIL, and CD226. The TRAIL and perforin/granzyme B pathways are largely involved in this cytotoxic mechanism, as indicated by the reduction in cytotoxicity induced by TRAIL inhibitory antibodies and concanamycin A, which inhibits perforin/granzyme B-mediated cytotoxicity. Our data suggest that eNK cells, whose functions have been enhanced by genetic engineering, have the potential to improve HCC treatment.
Collapse
Affiliation(s)
- Mayuna Nakamura
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan.
| | - Keishi Hakoda
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | | | | | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
10
|
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
|
11
|
Madrid M, Lakshmipathy U, Zhang X, Bharti K, Wall DM, Sato Y, Muschler G, Ting A, Smith N, Deguchi S, Kawamata S, Moore JC, Makovoz B, Sullivan S, Falco V, Al-Riyami AZ. Considerations for the development of iPSC-derived cell therapies: a review of key challenges by the JSRM-ISCT iPSC Committee. Cytotherapy 2024; 26:1382-1399. [PMID: 38958627 PMCID: PMC11471376 DOI: 10.1016/j.jcyt.2024.05.022] [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: 11/19/2023] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024]
Abstract
Since their first production in 2007, human induced pluripotent stem cells (iPSCs) have provided a novel platform for the development of various cell therapies targeting a spectrum of diseases, ranging from rare genetic eye disorders to cancer treatment. However, several challenges must be tackled for iPSC-based cell therapy to enter the market and achieve broader global adoption. This white paper, authored by the Japanese Society for Regenerative Medicine (JSRM) - International Society for Cell Therapy (ISCT) iPSC Committee delves into the hurdles encountered in the pursuit of safe and economically viable iPSC-based therapies, particularly from the standpoint of the cell therapy industry. It discusses differences in global guidelines and regulatory frameworks, outlines a series of quality control tests required to ensure the safety of the cell therapy, and provides details and important considerations around cost of goods (COGs), including the impact of automated advanced manufacturing.
Collapse
Affiliation(s)
| | | | | | - Kapil Bharti
- National Eye Institute of the National Institutes of Health, Bethesda, USA
| | - Dominic M Wall
- Peter MacCallum Cancer Centre, Melbourne Australia; Cell Therapies Pty Ltd, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Yoji Sato
- National Institute of Health Sciences, Kawasaki, Japan
| | | | | | | | - Shuhei Deguchi
- CIRA Foundation, Facility for iPS Cell Therapy (FiT), Kyoto, Japan
| | - Shin Kawamata
- Cyto-Facto Inc., Kobe, Japan; Kobe University, Kobe, Japan.
| | | | | | | | | | - Arwa Z Al-Riyami
- Department of Hematology, Sultan Qaboos University Hospital, University Medical City, Muscat, Oman
| |
Collapse
|
12
|
Sun J, Yates C, Dingwall S, Ongtengco C, Power D, Gray P, Prowse A. Analysis of three characterization assays reveals ddPCR of LIN28A as the most sensitive for the detection of residual pluripotent stem cells in cellular therapy products. Cytotherapy 2024; 26:1374-1381. [PMID: 38934983 DOI: 10.1016/j.jcyt.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND AIMS With the continuous development and advancement of human pluripotent stem cell (PSC)-derived cell therapies, an ever-increasing number of clinical indications can benefit from their application. Due to the capacity for PSCs to form teratomas, safety testing is required to ensure the absence of residual PSCs in a cell product. To mitigate these limitations, in vitro analytical methods can be utilized as quality control after the production of a PSC-derived cell product. Sensitivity of these analytic methods is critical in accurately quantifying residual PSC in the final cell product. In this study, we compared the sensitivity of three in vitro assays: qPCR, ddPCR and RT-LAMP. METHODS The spike-in samples were produced from three independent experiments, each spiked with different PSC lines (PSC1, NH50191, and WA09 referred to as H9) into a background of primary fibroblasts (Hs68). These samples were then subjected to qPCR, ddPCR and RT-LAMP to determine their detection limit in measuring a commonly used PSC marker, LIN28A. RESULTS The results indicated that the three analytic methods all exhibited consistent results across different cell-line spiked samples, with ddPCR demonstrating the highest sensitivity of the three methods. The LIN28A ddPCR assay could confidently detect 10 residual PSCs in a million fibroblasts. DISCUSSION In our hand, ddPCR LIN28A assay demonstrated the highest sensitivity for detection of residual PSCs compared to the other two assays. Correlating such in vitro safety results with corresponding in vivo studies demonstrating the tumorigenicity profile of PSC-derived cell therapy could accelerate the safe clinical translation of cell therapy.
Collapse
Affiliation(s)
- Jinda Sun
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Clarissa Yates
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia; Ketim Therapeutics, Brisbane, Australia
| | - Steve Dingwall
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Cherica Ongtengco
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Dominique Power
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia; Viatris, Brisbane, Australia
| | - Peter Gray
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Andrew Prowse
- Australia Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
13
|
Poon F, Sambathkumar R, Korytnikov R, Aghazadeh Y, Oakie A, Misra PS, Sarangi F, Nostro MC. Tankyrase inhibition promotes endocrine commitment of hPSC-derived pancreatic progenitors. Nat Commun 2024; 15:8754. [PMID: 39384787 PMCID: PMC11464881 DOI: 10.1038/s41467-024-53068-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) have the potential to differentiate into various cell types, including pancreatic insulin-producing β cells, which are crucial for developing therapies for diabetes. However, current methods for directing hPSC differentiation towards pancreatic β-like cells are often inefficient and produce cells that do not fully resemble the native counterparts. Here, we report that highly selective tankyrase inhibitors, such as WIKI4, significantly enhances pancreatic differentiation from hPSCs. Our results show that WIKI4 promotes the formation of pancreatic progenitors that give rise to islet-like cells with improved β-like cell frequencies and glucose responsiveness compared to our standard cultures. These findings not only advance our understanding of pancreatic development, but also provide a promising new tool for generating pancreatic cells for research and potential therapeutic applications.
Collapse
Affiliation(s)
- Frankie Poon
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Sana Biotechnology, 300 Technology Square, Cambridge, MA, 02139, USA
| | - Rangarajan Sambathkumar
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Allarta Life Science Inc., 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Roman Korytnikov
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Yasaman Aghazadeh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Montreal Clinical Research Institute (IRCM), University of Montreal, Department of Medicine, Montreal, H2W 1R7, QC, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Paraish S Misra
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Farida Sarangi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
14
|
Tang F, Dong T, Zhou C, Deng L, Liu HB, Wang W, Liu G, Ying M, Li PP. Genetically engineered human induced pluripotent stem cells for the production of brain-targeting extracellular vesicles. Stem Cell Res Ther 2024; 15:345. [PMID: 39380039 PMCID: PMC11462716 DOI: 10.1186/s13287-024-03955-2] [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: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are cell-secreted membrane vesicles that have become a promising, natural nanoparticle system for delivering either naturally carried or exogenously loaded therapeutic molecules. Among reported cell sources for EV manufacture, human induced pluripotent stem cells (hiPSCs) offer numerous advantages. However, hiPSC-EVs only have a moderate ability for brain delivery. Herein, we sought to develop a stable hiPSC line for producing EVs with substantially enhanced brain targeting by genetic engineering to overexpress rabies viral glycoprotein (RVG) peptide fused to the N terminus of lysosomal associated membrane protein 2B (RVG-Lamp2B) which has been shown capable of boosting the brain delivery of EVs via the nicotinic acetylcholine receptor. METHODS An RVG-Lamp2B-HA expression cassette was knocked into the AAVS1 safe harbor locus of a control hiPSC line using the CRISPR/Cas9-assisted homologous recombination. Western blot was used to detect the expression of RVG-Lamp2B-HA in RVG-edited hiPSCs as well as EVs derived from RVG-edited hiPSCs. Uptake of EVs by SH-SY5Y cells in the presence of various endocytic inhibitors was analyzed using flow cytometry. Biodistribution and brain delivery of intravenously injected control and RVG-modified EVs in wild-type mice were examined using ex vivo fluorescent imaging. RESULTS Here we report that an RVG-Lamp2B-HA expression cassette was knocked into the AAVS1 safe harbor locus of a control hiPSC line using the CRISPR/Cas9-assisted homologous recombination. The RVG-edited iPSCs have normal karyotype, express pluripotency markers, and have differentiation potential. Expression of RVG-Lamp2B-HA was detected in total cell extracts as well as EVs derived from RVG-edited (vs. control) hiPSCs. The RVG-modified EVs enter neuronal cells via distinct endocytic pathways, compared with control EVs. The biodistribution study confirmed that EVs derived from RVG-edited hiPSCs possess higher brain delivery efficiency. CONCLUSION Taken together, we have established stable, genetically engineered hiPSCs for producing EVs with RVG expression, offering the improved ability for brain-targeted drug delivery.
Collapse
Affiliation(s)
- Fan Tang
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD, 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Dong
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD, 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chengqian Zhou
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD, 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leon Deng
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD, 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hans B Liu
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD, 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wenshen Wang
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Guanshu Liu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Mingyao Ying
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Pan P Li
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD, 21287, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Kim R, Nagel SH, Liaw NY, Zimmermann WH, Zelarayán LC, Schoger E. Human induced pluripotent stem cells for live cell cycle monitoring and endogenous gene activation. Stem Cell Res 2024; 80:103531. [PMID: 39128210 DOI: 10.1016/j.scr.2024.103531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024] Open
Abstract
The fluorescence ubiquitination cell cycle inhibitor (FUCCI) has been introduced to monitor cell cycle activity in living cells, including human induced pluripotent stem cells (hiPSC) and derived cell types. We have recently developed hiPSC with stable expression of dCas9VPR for endogenous gene activation and a Citrine-tagged ACTN2 cell line to monitor sarcomere development and function in muscle cells. Here, we present dual and triple transgenic hiPSC lines developed by genomic integration of FUCCI with and without dCas9VPR into the ROSA26 and AAVS1 loci, respectively, in the previously introduced ACTN2-Citrine line. Functionality of the transgenes was demonstrated in the novel hiPSC line, which we introduce as Myo-CCER and CraCCER.
Collapse
Affiliation(s)
- Rosa Kim
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany; DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Sites Lower Saxony and Heidelberg/Mannheim, Germany
| | - Sebastian H Nagel
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany; DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Sites Lower Saxony and Heidelberg/Mannheim, Germany
| | - Norman Y Liaw
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany; DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Sites Lower Saxony and Heidelberg/Mannheim, Germany
| | - Wolfram-H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany; DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Sites Lower Saxony and Heidelberg/Mannheim, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Georg-August-University Göttingen, 37075 Göttingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 37075 Göttingen, Germany; DZNE (German Center for Neurodegenerative Diseases), 37075 Göttingen, Germany
| | - Laura C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany; DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Sites Lower Saxony and Heidelberg/Mannheim, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Georg-August-University Göttingen, 37075 Göttingen, Germany; Medical Clinic I: Cardiology and Angiology, Experimental Cardiology, Justus-Liebig-University Gießen, 35392 Gießen, Germany
| | - Eric Schoger
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany; DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Sites Lower Saxony and Heidelberg/Mannheim, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Georg-August-University Göttingen, 37075 Göttingen, Germany; Internal Medicine VIII: Institute of Experimental Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
16
|
Douvaras P, Buenaventura DF, Sun B, Lepack A, Baker E, Simpson E, Ebel M, Lallos G, LoSchiavo D, Stitt N, Adams N, McAuliffe C, Forton-Juarez A, Kosmyna B, Pereira E, Burnett B, Dilworth D, Fisher S, Wang J, Tonge P, Tomishima M, Paladini C, Wilkinson D, Soh CL, Srinivas M, Patsch C, Irion S. Ready-to-use iPSC-derived microglia progenitors for the treatment of CNS disease in mouse models of neuropathic mucopolysaccharidoses. Nat Commun 2024; 15:8132. [PMID: 39284802 PMCID: PMC11405712 DOI: 10.1038/s41467-024-52400-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
Mucopolysaccharidoses are inherited metabolic disorders caused by the deficiency in lysosomal enzymes required to break down glycosaminoglycans. Accumulation of glycosaminoglycans leads to progressive, systemic degenerative disease. The central nervous system is particularly affected, resulting in developmental delays, neurological regression, and early mortality. Current treatments fail to adequately address neurological defects. Here we explore the potential of human induced pluripotent stem cell (hiPSC)-derived microglia progenitors as a one-time, allogeneic off-the-shelf cell therapy for several mucopolysaccharidoses (MPS). We show that hiPSC-derived microglia progenitors, possessing normal levels of lysosomal enzymes, can deliver functional enzymes into four subtypes of MPS knockout cell lines through mannose-6-phosphate receptor-mediated endocytosis in vitro. Additionally, our findings indicate that a single administration of hiPSC-derived microglia progenitors can reduce toxic glycosaminoglycan accumulation and prevent behavioral deficits in two different animal models of MPS. Durable efficacy is observed for eight months after transplantation. These results suggest a potential avenue for treating MPS with hiPSC-derived microglia progenitors.
Collapse
Affiliation(s)
| | | | - Bruce Sun
- BlueRock Therapeutics, New York, NY, USA
| | | | | | | | - Mark Ebel
- BlueRock Therapeutics, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Jing Wang
- BlueRock Therapeutics, New York, NY, USA
| | | | | | | | | | | | | | - Christoph Patsch
- BlueRock Therapeutics, New York, NY, USA.
- Merck KGaA, Darmstadt, Germany.
| | | |
Collapse
|
17
|
Terheyden-Keighley D, Hühne M, Berger T, Hiller B, Martins S, Gamerschlag A, Sabour D, Meffert A, Kislat A, Slotta C, Hafezi F, Lichte J, Sudheer S, Tessmer K, Psathaki K, Ader M, Kogler G, Greber B. GMP-compliant iPS cell lines show widespread plasticity in a new set of differentiation workflows for cell replacement and cancer immunotherapy. Stem Cells Transl Med 2024; 13:898-911. [PMID: 39042522 PMCID: PMC11386223 DOI: 10.1093/stcltm/szae047] [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: 03/06/2024] [Accepted: 06/08/2024] [Indexed: 07/25/2024] Open
Abstract
Cell therapeutic applications based on induced pluripotent stem cells (iPSCs) appear highly promising and challenging at the same time. Good manufacturing practice (GMP) regulations impose necessary yet demanding requirements for quality and consistency when manufacturing iPSCs and their differentiated progeny. Given the scarcity of accessible GMP iPSC lines, we have established a corresponding production workflow to generate the first set of compliant cell banks. Hence, these lines met a comprehensive set of release specifications and, for instance, displayed a low overall mutation load reflecting their neonatal origin, cord blood. Based on these iPSC lines, we have furthermore developed a set of GMP-compatible workflows enabling improved gene targeting at strongly enhanced efficiencies and directed differentiation into critical cell types: A new protocol for the generation of retinal pigment epithelium (RPE) features a high degree of simplicity and efficiency. Mesenchymal stromal cells (MSCs) derived from iPSCs displayed outstanding expansion capacity. A fully optimized cardiomyocyte differentiation protocol was characterized by a particularly high batch-to-batch consistency at purities above 95%. Finally, we introduce a universal immune cell induction platform that converts iPSCs into multipotent precursor cells. These hematopoietic precursors could selectively be stimulated to become macrophages, T cells, or natural killer (NK) cells. A switch in culture conditions upon NK-cell differentiation induced a several thousand-fold expansion, which opens up perspectives for upscaling this key cell type in a feeder cell-independent approach. Taken together, these cell lines and improved manipulation platforms will have broad utility in cell therapy as well as in basic research.
Collapse
Affiliation(s)
| | | | | | - Björn Hiller
- Catalent Düsseldorf GmbH, 40764 Langenfeld, Germany
| | | | | | | | | | | | | | | | - Jens Lichte
- Catalent Düsseldorf GmbH, 40764 Langenfeld, Germany
| | | | - Karen Tessmer
- Center for Regenerative Therapies Dresden (CRTD) and Center for Molecular and Cellular Bioengineering, Dresden University of Technology, 01307 Dresden, Germany
| | - Katherina Psathaki
- Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, 49076 Osnabrück, Germany
| | - Marius Ader
- Center for Regenerative Therapies Dresden (CRTD) and Center for Molecular and Cellular Bioengineering, Dresden University of Technology, 01307 Dresden, Germany
| | - Gesine Kogler
- Institute of Transplantation Diagnostics and Cell Therapeutics and Jose Carreras Stem Cell Bank, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Boris Greber
- Catalent Düsseldorf GmbH, 40764 Langenfeld, Germany
| |
Collapse
|
18
|
Novoa J, Westra I, Steeneveld E, Neves NF, Daleman L, Asensio AB, Davis RP, Carlotti F, Freund C, Rabelink T, Meij P, Wieles B. Validating human induced pluripotent stem cell-specific quality control tests for the release of an intermediate drug product in a Good Manufacturing Practice quality system. Cytotherapy 2024; 26:1105-1117. [PMID: 38703154 DOI: 10.1016/j.jcyt.2024.04.004] [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: 01/25/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
One of the challenges in Good Manufacturing Practice (GMP)-compliant human induced pluripotent stem cell (hiPSC) production is the validation of quality control (QC) tests specific for hiPSCs, which are required for GMP batch release. This study presents a comprehensive description of the validation process for hiPSC-specific GMP-compliant QC assays; more specifically, the validation of assays to assess the potential presence of residual episomal vectors (REVs), the expression of markers of the undifferentiated state and the directed differentiation potential of hiPSCs. Critical aspects and specific acceptance criteria were formulated in a validation plan prior to assay validation. Assay specificity, sensitivity and reproducibility were tested, and the equipment used for each assay was subjected to performance qualification. A minimum input of 20 000 cells (120 ng of genomic DNA) was defined for accurate determination of the presence of REVs. Furthermore, since vector loss in hiPSC lines is a passage-dependent process, we advocate screening for REVs between passages eight and 10, as testing at earlier passages might lead to unnecessary rejection of hiPSC lines. The cutoff value for assessment of markers of the undifferentiated state was set to the expression of at least three individual markers on at least 75% of the cells. When multi-color flow cytometry panels are used, a fluorescence minus one control is advised to ensure the control for fluorescent spread. For the assay to assess the directed differentiation potential, the detection limit was set to two of three positive lineage-specific markers for each of the three individual germ layers. All of our assays proved to be reproducible and specific. Our data demonstrate that our implemented analytical procedures are suitable as QC assays for the batch release of GMP-compliant hiPSCs.
Collapse
Affiliation(s)
- Juan Novoa
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge Westra
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Esther Steeneveld
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Natascha Fonseca Neves
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Lizanne Daleman
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Albert Blanch Asensio
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christian Freund
- Leiden University Medical Center hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton Rabelink
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands.
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Brigitte Wieles
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
19
|
Gadomski SJ, Mui BW, Gorodetsky R, Paravastu SS, Featherall J, Li L, Haffey A, Kim JC, Kuznetsov SA, Futrega K, Lazmi-Hailu A, Merling RK, NIDCD/NIDCR Genomics and Computational Biology Core ,, Martin D, McCaskie AW, Robey PG. Time- and cell-specific activation of BMP signaling restrains chondrocyte hypertrophy. iScience 2024; 27:110537. [PMID: 39193188 PMCID: PMC11347861 DOI: 10.1016/j.isci.2024.110537] [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: 04/14/2023] [Revised: 02/29/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Stem cell therapies for degenerative cartilage disease are limited by an incomplete understanding of hyaline cartilage formation and maintenance. Human bone marrow stromal cells/skeletal stem cells (hBMSCs/SSCs) produce stable hyaline cartilage when attached to hyaluronic acid-coated fibrin microbeads (HyA-FMBs), yet the mechanism remains unclear. In vitro, hBMSC/SSC/HyA-FMB organoids exhibited reduced BMP signaling early in chondrogenic differentiation, followed by restoration of BMP signaling in chondrogenic IGFBP5 + /MGP + cells. Subsequently, human-induced pluripotent stem cell (hiPSC)-derived sclerotome cells were established (BMP inhibition) and then treated with transforming growth factor β (TGF-β) -/+ BMP2 and growth differentiation factor 5 (GDF5) (BMP signaling activation). TGF-β alone elicited a weak chondrogenic response, but TGF-β/BMP2/GDF5 led to delamination of SOX9 + aggregates (chondrospheroids) with high expression of COL2A1, ACAN, and PRG4 and minimal expression of COL10A1 and ALP in vitro. While transplanted hBMSCs/SSCs/HyA-FMBs did not heal articular cartilage defects in immunocompromised rodents, chondrospheroid-derived cells/HyA-FMBs formed non-hypertrophic cartilage that persisted until at least 5 months in vivo.
Collapse
Affiliation(s)
- Stephen J. Gadomski
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Oxford-Cambridge Scholars Program in Partnership with Medical University of South Carolina, Charleston, SC 29425, USA
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Byron W.H. Mui
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
- NIH Oxford-Cambridge Scholars Program in Partnership with Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Raphael Gorodetsky
- Lab of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sriram S. Paravastu
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph Featherall
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Li
- National Institute of Dental and Craniofacial Research Imaging Core, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abigail Haffey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- National Institute of Dental and Craniofacial Research Summer Internship Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jae-Chun Kim
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- National Institute of Dental and Craniofacial Research Summer Dental Student Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergei A. Kuznetsov
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Kathryn Futrega
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Astar Lazmi-Hailu
- Lab of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Randall K. Merling
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - NIDCD/NIDCR Genomics and Computational Biology Core,
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, 35A Convent Drive, Room 1F-103, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, 35A Convent Drive, Room 1F-103, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew W. McCaskie
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
- Department of Surgery, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
20
|
Gurwitz D, Steeg R. Enriching iPSC research diversity: Harnessing human biobank collections for improved ethnic representation. Drug Dev Res 2024; 85:e22227. [PMID: 38943497 DOI: 10.1002/ddr.22227] [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: 04/21/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
Biobanks of human biosamples and cell lines are indispensable for biomedical research on human health and disease and for drug development projects. Many human cell line biobanks worldwide hold collections of lymphoblastoid cell lines (LCLs), representing thousands of affected and control donors from diverse ethnic/ancestry groups. In recent years, induced human pluripotent stem cells (iPSCs) and differentiated human cells derived from these iPSCs have become indispensable for applied biomedical research. Establishing iPSCs remains a laborious and costly step towards generating differentiated human cells. To address this research need, several non-profit and commercial biobanks have established iPSC collections for distribution to researchers, thereby serving as a resource for generating differentiated human cells. The most common starting materials for generation of iPSCs are a skin biopsy for harvesting fibroblasts, or a blood sample for collection of peripheral blood mononuclear cells. However untapped resources include the large established collections of biobanked human LCLs which can be reprogrammed to iPSCs using a variety of published protocols including the use of non-integrating episomal vectors. Many biobanks curate LCLs from diverse ethnic/ancestry populations, an aspect largely absent in most established iPSC biobanks which tend to primarily reflect populations from developed countries. Here, we call upon researchers across the breadth of iPSC research to tap the unique resource of existing and diverse human LCL collections for establishing biobanked iPSC panels that better represent the varied human ethnic (and hence genomic) diversity, thereby benefiting precision medicine and drug development research on a global scale.
Collapse
Affiliation(s)
- David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Rachel Steeg
- European Bank for Induced Pluripotent Stem Cells, Fraunhofer UK Research Ltd, Glasgow, UK
| |
Collapse
|
21
|
Tsuneyoshi N, Hosoya T, Takeno Y, Saitoh K, Murai H, Amimoto N, Tatsumi R, Watanabe S, Hasegawa Y, Kikkawa E, Goto K, Nishigaki F, Tamura K, Kimura H. Hypoimmunogenic human iPSCs expressing HLA-G, PD-L1, and PD-L2 evade innate and adaptive immunity. Stem Cell Res Ther 2024; 15:193. [PMID: 38956724 PMCID: PMC11218117 DOI: 10.1186/s13287-024-03810-4] [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: 04/18/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND The human induced pluripotent stem cells (hiPSCs) can generate all the cells composing the human body, theoretically. Therefore, hiPSCs are thought to be a candidate source of stem cells for regenerative medicine. The major challenge of allogeneic hiPSC-derived cell products is their immunogenicity. The hypoimmunogenic cell strategy is allogenic cell therapy without using immune suppressants. Advances in gene engineering technology now permit the generation of hypoimmunogenic cells to avoid allogeneic immune rejection. In this study, we generated a hypoimmunogenic hiPSC (HyPSC) clone that had diminished expression of human leukocyte antigen (HLA) class Ia and class II and expressed immune checkpoint molecules and a safety switch. METHODS First, we generated HLA class Ia and class II double knockout (HLA class Ia/II DKO) hiPSCs. Then, a HyPSC clone was generated by introducing exogenous β-2-microglobulin (B2M), HLA-G, PD-L1, and PD-L2 genes, and the Rapamycin-activated Caspase 9 (RapaCasp9)-based suicide gene as a safety switch into the HLA class Ia/II DKO hiPSCs. The characteristics and immunogenicity of the HyPSCs and their derivatives were analyzed. RESULTS We found that the expression of HLA-G on the cell surface can be enhanced by introducing the exogenous HLA-G gene along with B2M gene into HLA class Ia/II DKO hiPSCs. The HyPSCs retained a normal karyotype and had the characteristics of pluripotent stem cells. Moreover, the HyPSCs could differentiate into cells of all three germ layer lineages including CD45+ hematopoietic progenitor cells (HPCs), functional endothelial cells, and hepatocytes. The HyPSCs-derived HPCs exhibited the ability to evade innate and adaptive immunity. Further, we demonstrated that RapaCasp9 could be used as a safety switch in vitro and in vivo. CONCLUSION The HLA class Ia/II DKO hiPSCs armed with HLA-G, PD-L1, PD-L2, and RapaCasp9 molecules are a potential source of stem cells for allogeneic transplantation.
Collapse
Affiliation(s)
- Norihiro Tsuneyoshi
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Tomonori Hosoya
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Yuriko Takeno
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Kodai Saitoh
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Hidetaka Murai
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Naoki Amimoto
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Rie Tatsumi
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Sono Watanabe
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Yudai Hasegawa
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Eri Kikkawa
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Kumiko Goto
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Fusako Nishigaki
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Kouichi Tamura
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan.
| | - Hironobu Kimura
- HEALIOS K.K. Kobe Research Institute, Kobe KIMEC Center Bldg. 3F, 1-5-2 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
22
|
Jain S, Voulgaris D, Thongkorn S, Hesen R, Hägg A, Moslem M, Falk A, Herland A. On-Chip Neural Induction Boosts Neural Stem Cell Commitment: Toward a Pipeline for iPSC-Based Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401859. [PMID: 38655836 PMCID: PMC11220685 DOI: 10.1002/advs.202401859] [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: 02/21/2024] [Indexed: 04/26/2024]
Abstract
The clinical translation of induced pluripotent stem cells (iPSCs) holds great potential for personalized therapeutics. However, one of the main obstacles is that the current workflow to generate iPSCs is expensive, time-consuming, and requires standardization. A simplified and cost-effective microfluidic approach is presented for reprogramming fibroblasts into iPSCs and their subsequent differentiation into neural stem cells (NSCs). This method exploits microphysiological technology, providing a 100-fold reduction in reagents for reprogramming and a ninefold reduction in number of input cells. The iPSCs generated from microfluidic reprogramming of fibroblasts show upregulation of pluripotency markers and downregulation of fibroblast markers, on par with those reprogrammed in standard well-conditions. The NSCs differentiated in microfluidic chips show upregulation of neuroectodermal markers (ZIC1, PAX6, SOX1), highlighting their propensity for nervous system development. Cells obtained on conventional well plates and microfluidic chips are compared for reprogramming and neural induction by bulk RNA sequencing. Pathway enrichment analysis of NSCs from chip showed neural stem cell development enrichment and boosted commitment to neural stem cell lineage in initial phases of neural induction, attributed to a confined environment in a microfluidic chip. This method provides a cost-effective pipeline to reprogram and differentiate iPSCs for therapeutics compliant with current good manufacturing practices.
Collapse
Affiliation(s)
- Saumey Jain
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
| | - Dimitrios Voulgaris
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
- AIMESCenter for Integrated Medical and Engineering ScienceDepartment of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| | - Surangrat Thongkorn
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
- Chulalongkorn Autism Research and Innovation Center of Excellence (Chula ACE)Department of Clinical ChemistryFaculty of Allied Health SciencesChulalongkorn UniversityBangkok10330Thailand
| | - Rick Hesen
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
| | - Alice Hägg
- Neural Stem CellsDepartment of Experimental Medical ScienceLund Stem Cell CenterLund UniversityLund221 84Sweden
| | - Mohsen Moslem
- Department of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| | - Anna Falk
- Neural Stem CellsDepartment of Experimental Medical ScienceLund Stem Cell CenterLund UniversityLund221 84Sweden
- Department of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| | - Anna Herland
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
- AIMESCenter for Integrated Medical and Engineering ScienceDepartment of NeuroscienceKarolinska InstitutetSolna171 65Sweden
- Department of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| |
Collapse
|
23
|
Novoa JJ, Westra IM, Steeneveld E, Fonseca Neves N, Arendzen CH, Rajaei B, Grundeken E, Yildiz M, van der Valk W, Salvador A, Carlotti F, Dijkers PF, Locher H, van den Berg CW, Raymond KI, Kirkeby A, Mummery CL, Rabelink TJ, Freund C, Meij P, Wieles B. Good Manufacturing Practice-compliant human induced pluripotent stem cells: from bench to putative clinical products. Cytotherapy 2024; 26:556-566. [PMID: 38483359 DOI: 10.1016/j.jcyt.2024.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND AIMS Few human induced pluripotent stem cell (hiPSC) lines are Good Manufacturing Practice (GMP)-compliant, limiting the clinical use of hiPSC-derived products. Here, we addressed this by establishing and validating an in-house platform to produce GMP-compliant hiPSCs that would be appropriate for producing both allogeneic and autologous hiPSC-derived products. METHODS Our standard research protocol for hiPSCs production was adapted and translated into a GMP-compliant platform. In addition to the generation of GMP-compliant hiPSC, the platform entails the methodology for donor recruitment, consent and screening, donor material procurement, hiPSCs manufacture, in-process control, specific QC test validation, QC testing, product release, hiPSCs storage and stability testing. For platform validation, one test run and three production runs were performed. Highest-quality lines were selected to establish master cell banks (MCBs). RESULTS Two MCBs were successfully released under GMP conditions. They demonstrated safety (sterility, negative mycoplasma, endotoxins <5.0 EU/mL and negative adventitious agents), cell identity (>75% of cells expressing markers of undifferentiated state, identical STR profile, normal karyotype in >20 metaphases), purity (negative residual vectors and no plasmid integration in the genome) and potency (expression of at least two of the three markers for each of the three germ layers). In addition, directed differentiation to somitoids (skeletal muscle precursors) and six potential clinical products from all three germ layers was achieved: pancreatic islets (endoderm), kidney organoids and cardiomyocytes (mesoderm), and keratinocytes, GABAergic interneurons and inner-ear organoids (ectoderm). CONCLUSIONS We successfully developed and validated a platform for generating GMP-compliant hiPSC lines. The two MCBs released were shown to differentiate into clinical products relevant for our own and other regenerative medicine interests.
Collapse
Affiliation(s)
- Juan J Novoa
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge M Westra
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Esther Steeneveld
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Natascha Fonseca Neves
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, The Netherlands
| | - Christiaan H Arendzen
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bahareh Rajaei
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Esmée Grundeken
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Mehmet Yildiz
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter van der Valk
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Alison Salvador
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Pascale F Dijkers
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Heiko Locher
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Cathelijne W van den Berg
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Karine I Raymond
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Agnete Kirkeby
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands.
| | - Christian Freund
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Brigitte Wieles
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Hayashi Y, Uno Y, Kino-Oka M, Sugiyama H. Computer-aided exploration of multiobjective optimal temperature profiles in slow freezing for human induced pluripotent stem cells. Cryobiology 2024; 115:104885. [PMID: 38513997 DOI: 10.1016/j.cryobiol.2024.104885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/05/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Human induced pluripotent stem (hiPS) cells have demonstrated promising potential in regenerative medical therapeutics. After successful clinical trials, the demand for hiPS cells has steadily increased. Therefore, the optimization of hiPS cell freezing processes for storage and transportation is essential. Here, we presented a computer-aided exploration of multiobjective optimal temperature profiles in slow freezing for hiPS cells. This study was based on a model that calculates cell survival rates after thawing, and the model was extended to evaluate cell potentials until 24 h after seeding. To estimate parameter values for this extension, freezing experiments were performed using constant cooling rates. Using quality and productivity indicators, we evaluated 16,206 temperature profiles using our model, and a promising profile was obtained. Finally, an experimental investigation of the profile was undertaken, and the contribution of the temperature profile to both quality and productivity was confirmed.
Collapse
Affiliation(s)
- Yusuke Hayashi
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-8656, Tokyo, Japan
| | - Yuki Uno
- Department of Biotechnology, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Hirokazu Sugiyama
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-8656, Tokyo, Japan.
| |
Collapse
|
25
|
Agrawal T, Maddileti S, Mariappan I. Generation and characterization of three CRISPR/Cas9 edited RB1 null hiPSC lines for retinoblastoma disease modelling. Stem Cell Res 2024; 76:103373. [PMID: 38452707 DOI: 10.1016/j.scr.2024.103373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
Complete loss of RB1 causes retinoblastoma. Here, we report the generation of three RB1-/- iPSC lines using CRISPR/Cas9 based editing at exon 18 of RB1 in a healthy control hiPSC line. The edited cells were clonally expanded, genotyped and characterized to establish the mutant lines. Two of the mutant lines are compound heterozygous, with different in-del mutations in each of their alleles, while the third mutant line is homozygous, with identical edits in both alleles. All lines maintained their stemness, pluripotency, formed embryoid bodies with cell types of all three lineages, displayed a normal karyotype and lost RB1 expression.
Collapse
Affiliation(s)
- Trupti Agrawal
- Centre for Ocular Regeneration, Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L.V. Prasad Eye Institute, Hyderabad, Telangana, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Savitri Maddileti
- Centre for Ocular Regeneration, Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L.V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Indumathi Mariappan
- Centre for Ocular Regeneration, Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L.V. Prasad Eye Institute, Hyderabad, Telangana, India.
| |
Collapse
|
26
|
Sun C, Serra C, Kalicharan BH, Harding J, Rao M. Challenges and Considerations of Preclinical Development for iPSC-Based Myogenic Cell Therapy. Cells 2024; 13:596. [PMID: 38607035 PMCID: PMC11011706 DOI: 10.3390/cells13070596] [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: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Cell therapies derived from induced pluripotent stem cells (iPSCs) offer a promising avenue in the field of regenerative medicine due to iPSCs' expandability, immune compatibility, and pluripotent potential. An increasing number of preclinical and clinical trials have been carried out, exploring the application of iPSC-based therapies for challenging diseases, such as muscular dystrophies. The unique syncytial nature of skeletal muscle allows stem/progenitor cells to integrate, forming new myonuclei and restoring the expression of genes affected by myopathies. This characteristic makes genome-editing techniques especially attractive in these therapies. With genetic modification and iPSC lineage specification methodologies, immune-compatible healthy iPSC-derived muscle cells can be manufactured to reverse the progression of muscle diseases or facilitate tissue regeneration. Despite this exciting advancement, much of the development of iPSC-based therapies for muscle diseases and tissue regeneration is limited to academic settings, with no successful clinical translation reported. The unknown differentiation process in vivo, potential tumorigenicity, and epigenetic abnormality of transplanted cells are preventing their clinical application. In this review, we give an overview on preclinical development of iPSC-derived myogenic cell transplantation therapies including processes related to iPSC-derived myogenic cells such as differentiation, scaling-up, delivery, and cGMP compliance. And we discuss the potential challenges of each step of clinical translation. Additionally, preclinical model systems for testing myogenic cells intended for clinical applications are described.
Collapse
Affiliation(s)
- Congshan Sun
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| | - Carlo Serra
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Mahendra Rao
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| |
Collapse
|
27
|
Shahriyari M, Rinn M, Hofemeier AD, Babych A, Zimmermann WH, Tiburcy M. Protocol to develop force-generating human skeletal muscle organoids. STAR Protoc 2024; 5:102794. [PMID: 38133957 PMCID: PMC10776636 DOI: 10.1016/j.xpro.2023.102794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/24/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Force generation is an essential property of skeletal muscle models in vitro. We describe a versatile 1-step procedure to direct undifferentiated human pluripotent stem cells (PSCs) into contractile skeletal muscle organoids (SMOs). Our protocol provides detailed steps for 3D casting of PSCs using either collagen-I/Matrigel- or fibrin/Geltrex-based hydrogels, SMO differentiation, and application of different culture platforms for mechanical loading and contractility analysis. The SMO model may be particularly useful to study human muscle development and developmental skeletal muscle disorders in vitro. For complete details on the use and execution of this protocol, please refer to Shahriyari et al.1.
Collapse
Affiliation(s)
- Mina Shahriyari
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| | - Malte Rinn
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Arne D Hofemeier
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Artem Babych
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
28
|
Mizuno M, Maeda Y, Sanami S, Matsuzaki T, Yoshikawa HY, Ozeki N, Koga H, Sekiya I. Noninvasive total counting of cultured cells using a home-use scanner with a pattern sheet. iScience 2024; 27:109170. [PMID: 38405610 PMCID: PMC10884908 DOI: 10.1016/j.isci.2024.109170] [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: 02/20/2023] [Revised: 11/07/2023] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
The inherent variability in cell culture techniques hinders their reproducibility. To address this issue, we introduce a comprehensive cell observation device. This new approach enhances the features of existing home-use scanners by implementing a pattern sheet. Compared with fluorescent staining, our method over- or underestimated the cell count by a mere 5%. The proposed technique showcased a strong correlation with conventional methodologies, displaying R2 values of 0.91 and 0.99 compared with the standard chamber and fluorescence methods, respectively. Simulations of microscopic observations indicated the potential to estimate accurately the total cell count using just 20 fields of view. Our proposed cell-counting device offers a straightforward, noninvasive means of measuring the number of cultured cells. By harnessing the power of deep learning, this device ensures data integrity, thereby making it an attractive option for future cell culture research.
Collapse
Affiliation(s)
- Mitsuru Mizuno
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Yushima, Tokyo 113-8519, Japan
| | - Yoshitaka Maeda
- Medical & Healthcare Division, Dai Nippon Printing Co., Ltd., Tokyo, Japan
| | - Sho Sanami
- Medical & Healthcare Division, Dai Nippon Printing Co., Ltd., Tokyo, Japan
| | - Takahisa Matsuzaki
- Department of Applied Physics, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Hiroshi Y. Yoshikawa
- Department of Applied Physics, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Nobutake Ozeki
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Yushima, Tokyo 113-8519, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Yushima, Tokyo 113-8519, Japan
| |
Collapse
|
29
|
Bose D, Ortolan D, Farnoodian M, Sharma R, Bharti K. Considerations for Developing an Autologous Induced Pluripotent Stem Cell (iPSC)-Derived Retinal Pigment Epithelium (RPE) Replacement Therapy. Cold Spring Harb Perspect Med 2024; 14:a041295. [PMID: 37487631 PMCID: PMC10910357 DOI: 10.1101/cshperspect.a041295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Cell-replacement therapies are a new class of treatments, which include induced pluripotent stem cell (iPSC)-derived tissues that aim to replace degenerated cells. iPSCs can potentially be used to generate any cell type of the body, making them a powerful tool for treating degenerative diseases. Cell replacement for retinal degenerative diseases is at the forefront of cell therapies, given the accessibility of the eye for surgical procedures and a huge unmet medical need for retinal degenerative diseases with no current treatment options. Clinical trials are ongoing in different parts of the world using stem cell-derived retinal pigment epithelium (RPE). This review focuses on scientific and regulatory considerations when developing an iPSC-derived RPE cell therapy from the development of a robust and efficient differentiation protocol to critical quality control assays for cell validation, the choice of an appropriate animal model for preclinical testing, and the regulatory aspects that dictate the final approval for proceeding to a first-in-human clinical trial.
Collapse
Affiliation(s)
- Devika Bose
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
30
|
Gregor BW, Coston ME, Adams EM, Arakaki J, Borensztejn A, Do TP, Fuqua MA, Haupt A, Hendershott MC, Leung W, Mueller IA, Nath A, Nelson AM, Rafelski SM, Sanchez EE, Swain-Bowden MJ, Tang WJ, Thirstrup DJ, Wiegraebe W, Whitney BP, Yan C, Gunawardane RN, Gaudreault N. Automated human induced pluripotent stem cell culture and sample preparation for 3D live-cell microscopy. Nat Protoc 2024; 19:565-594. [PMID: 38087082 DOI: 10.1038/s41596-023-00912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/08/2023] [Indexed: 02/12/2024]
Abstract
To produce abundant cell culture samples to generate large, standardized image datasets of human induced pluripotent stem (hiPS) cells, we developed an automated workflow on a Hamilton STAR liquid handler system. This was developed specifically for culturing hiPS cell lines expressing fluorescently tagged proteins, which we have used to study the principles by which cells establish and maintain robust dynamic localization of cellular structures. This protocol includes all details for the maintenance, passage and seeding of cells, as well as Matrigel coating of 6-well plastic plates and 96-well optical-grade, glass plates. We also developed an automated image-based hiPS cell colony segmentation and feature extraction pipeline to streamline the process of predicting cell count and selecting wells with consistent morphology for high-resolution three-dimensional (3D) microscopy. The imaging samples produced with this protocol have been used to study the integrated intracellular organization and cell-to-cell variability of hiPS cells to train and develop deep learning-based label-free predictions from transmitted-light microscopy images and to develop deep learning-based generative models of single-cell organization. This protocol requires some experience with robotic equipment. However, we provide details and source code to facilitate implementation by biologists less experienced with robotics. The protocol is completed in less than 10 h with minimal human interaction. Overall, automation of our cell culture procedures increased our imaging samples' standardization, reproducibility, scalability and consistency. It also reduced the need for stringent culturist training and eliminated culturist-to-culturist variability, both of which were previous pain points of our original manual pipeline workflow.
Collapse
Affiliation(s)
| | | | | | - Joy Arakaki
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | - Thao P Do
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | - Amanda Haupt
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | - Winnie Leung
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | - Aditya Nath
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | | | | | | | - W Joyce Tang
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | | | | | - Calysta Yan
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | | |
Collapse
|
31
|
Netsrithong R, Garcia-Perez L, Themeli M. Engineered T cells from induced pluripotent stem cells: from research towards clinical implementation. Front Immunol 2024; 14:1325209. [PMID: 38283344 PMCID: PMC10811463 DOI: 10.3389/fimmu.2023.1325209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived T (iT) cells represent a groundbreaking frontier in adoptive cell therapies with engineered T cells, poised to overcome pivotal limitations associated with conventional manufacturing methods. iPSCs offer an off-the-shelf source of therapeutic T cells with the potential for infinite expansion and straightforward genetic manipulation to ensure hypo-immunogenicity and introduce specific therapeutic functions, such as antigen specificity through a chimeric antigen receptor (CAR). Importantly, genetic engineering of iPSC offers the benefit of generating fully modified clonal lines that are amenable to rigorous safety assessments. Critical to harnessing the potential of iT cells is the development of a robust and clinically compatible production process. Current protocols for genetic engineering as well as differentiation protocols designed to mirror human hematopoiesis and T cell development, vary in efficiency and often contain non-compliant components, thereby rendering them unsuitable for clinical implementation. This comprehensive review centers on the remarkable progress made over the last decade in generating functional engineered T cells from iPSCs. Emphasis is placed on alignment with good manufacturing practice (GMP) standards, scalability, safety measures and quality controls, which constitute the fundamental prerequisites for clinical application. In conclusion, the focus on iPSC as a source promises standardized, scalable, clinically relevant, and potentially safer production of engineered T cells. This groundbreaking approach holds the potential to extend hope to a broader spectrum of patients and diseases, leading in a new era in adoptive T cell therapy.
Collapse
Affiliation(s)
- Ratchapong Netsrithong
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Laura Garcia-Perez
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maria Themeli
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
32
|
Muok L, Sun L, Esmonde C, Worden H, Vied C, Duke L, Ma S, Zeng O, Driscoll T, Jung S, Li Y. Extracellular vesicle biogenesis of three-dimensional human pluripotent stem cells in a novel Vertical-Wheel bioreactor. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e133. [PMID: 38938678 PMCID: PMC11080838 DOI: 10.1002/jex2.133] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) secreted by human-induced pluripotent stem cells (hiPSCs) have great potential as cell-free therapies in various diseases, including prevention of blood-brain barrier senescence and stroke. However, there are still challenges in pre-clinical and clinical use of hiPSC-EVs due to the need for large-scale production of a large quantity. Vertical-Wheel bioreactors (VWBRs) have design features that allow the biomanufacturing of hiPSC-EVs using a scalable aggregate or microcarrier-based culture system under low shear stress. EV secretion by undifferentiated hiPSCs expanded as 3-D aggregates and on Synthemax II microcarriers in VWBRs were investigated. Additionally, two types of EV collection media, mTeSR and HBM, were compared. The hiPSCs were characterized by metabolite and transcriptome analysis as well as EV biogenesis markers. Protein and microRNA cargo were analysed by proteomics and microRNA-seq, respectively. The in vitro functional assays of microglia stimulation and proliferation were conducted. HiPSCs expanded as 3-D aggregates and on microcarriers had comparable cell number, while microcarrier culture had higher glucose consumption, higher glycolysis and lower autophagy gene expression based on mRNA-seq. The microcarrier cultures had at least 17-23 fold higher EV secretion, and EV collection in mTeSR had 2.7-3.7 fold higher yield than HBM medium. Microcarrier culture with mTeSR EV collection had a smaller EV size than other groups, and the cargo was enriched with proteins (proteomics) and miRNAs (microRNA-seq) reducing apoptosis and promoting cell proliferation (e.g. Wnt-related pathways). hiPSC-EVs demonstrated the ability of stimulating proliferation and M2 polarization of microglia in vitro. HiPSC expansion on microcarriers produces much higher yields of EVs than hiPSC aggregates in VWBRs. EV collection in mTeSR increases yield compared to HBM. The biomanufactured EVs from microcarrier culture in mTeSR have exosomal characteristics and are functional in microglia stimulation, which paves the ways for future in vivo anti-aging study.
Collapse
Affiliation(s)
- Laureana Muok
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- Department of Biomedical Sciences, College of MedicineFlorida State UniversityTallahasseeFloridaUSA
| | - Colin Esmonde
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | | | - Cynthia Vied
- Department of Biomedical Sciences, College of MedicineFlorida State UniversityTallahasseeFloridaUSA
| | - Leanne Duke
- Department of Biomedical Sciences, College of MedicineFlorida State UniversityTallahasseeFloridaUSA
| | - Shaoyang Ma
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Olivia Zeng
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Tristan Driscoll
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | | | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| |
Collapse
|
33
|
Selvarajah K, Tan JJ, Shaharuddin B. Corneal Epithelial Development and the Role of Induced Pluripotent Stem Cells for Regeneration. Curr Stem Cell Res Ther 2024; 19:292-306. [PMID: 36915985 DOI: 10.2174/1574888x18666230313094121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/02/2022] [Accepted: 01/02/2023] [Indexed: 03/16/2023]
Abstract
Severe corneal disorders due to infective aetiologies, trauma, chemical injuries, and chronic cicatricial inflammations, are among vision-threatening pathologies leading to permanent corneal scarring. The whole cornea or lamellar corneal transplantation is often used as a last resort to restore vision. However, limited autologous tissue sources and potential adverse post-allotransplantation sequalae urge the need for more robust and strategic alternatives. Contemporary management using cultivated corneal epithelial transplantation has paved the way for utilizing stem cells as a regenerative potential. Humaninduced pluripotent stem cells (hiPSCs) can generate ectodermal progenitors and potentially be used for ocular surface regeneration. This review summarizes the process of corneal morphogenesis and the signaling pathways underlying the development of corneal epithelium, which is key to translating the maturation and differentiation process of hiPSCs in vitro. The current state of knowledge and methodology for driving efficient corneal epithelial cell differentiation from pluripotent stem cells are highlighted.
Collapse
Affiliation(s)
- Komathi Selvarajah
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
- Department of Microbiology, Faculty of Medicine, Asian Institute of Medical Sciences and Technology (AIMST) University, Kedah, Malaysia
| | - Jun Jie Tan
- Department of Microbiology, Faculty of Medicine, Asian Institute of Medical Sciences and Technology (AIMST) University, Kedah, Malaysia
| | - Bakiah Shaharuddin
- Department of Microbiology, Faculty of Medicine, Asian Institute of Medical Sciences and Technology (AIMST) University, Kedah, Malaysia
| |
Collapse
|
34
|
Seibertz F, Rubio T, Springer R, Popp F, Ritter M, Liutkute A, Bartelt L, Stelzer L, Haghighi F, Pietras J, Windel H, Pedrosa NDI, Rapedius M, Doering Y, Solano R, Hindmarsh R, Shi R, Tiburcy M, Bruegmann T, Kutschka I, Streckfuss-Bömeke K, Kensah G, Cyganek L, Zimmermann WH, Voigt N. Atrial fibrillation-associated electrical remodelling in human induced pluripotent stem cell-derived atrial cardiomyocytes: a novel pathway for antiarrhythmic therapy development. Cardiovasc Res 2023; 119:2623-2637. [PMID: 37677054 PMCID: PMC10730244 DOI: 10.1093/cvr/cvad143] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is associated with tachycardia-induced cellular electrophysiology alterations which promote AF chronification and treatment resistance. Development of novel antiarrhythmic therapies is hampered by the absence of scalable experimental human models that reflect AF-associated electrical remodelling. Therefore, we aimed to assess if AF-associated remodelling of cellular electrophysiology can be simulated in human atrial-like cardiomyocytes derived from induced pluripotent stem cells in the presence of retinoic acid (iPSC-aCM), and atrial-engineered human myocardium (aEHM) under short term (24 h) and chronic (7 days) tachypacing (TP). METHODS AND RESULTS First, 24-h electrical pacing at 3 Hz was used to investigate whether AF-associated remodelling in iPSC-aCM and aEHM would ensue. Compared to controls (24 h, 1 Hz pacing) TP-stimulated iPSC-aCM presented classical hallmarks of AF-associated remodelling: (i) decreased L-type Ca2+ current (ICa,L) and (ii) impaired activation of acetylcholine-activated inward-rectifier K+ current (IK,ACh). This resulted in action potential shortening and an absent response to the M-receptor agonist carbachol in both iPSC-aCM and aEHM subjected to TP. Accordingly, mRNA expression of the channel-subunit Kir3.4 was reduced. Selective IK,ACh blockade with tertiapin reduced basal inward-rectifier K+ current only in iPSC-aCM subjected to TP, thereby unmasking an agonist-independent constitutively active IK,ACh. To allow for long-term TP, we developed iPSC-aCM and aEHM expressing the light-gated ion-channel f-Chrimson. The same hallmarks of AF-associated remodelling were observed after optical-TP. In addition, continuous TP (7 days) led to (i) increased amplitude of inward-rectifier K+ current (IK1), (ii) hyperpolarization of the resting membrane potential, (iii) increased action potential-amplitude and upstroke velocity as well as (iv) reversibly impaired contractile function in aEHM. CONCLUSIONS Classical hallmarks of AF-associated remodelling were mimicked through TP of iPSC-aCM and aEHM. The use of the ultrafast f-Chrimson depolarizing ion channel allowed us to model the time-dependence of AF-associated remodelling in vitro for the first time. The observation of electrical remodelling with associated reversible contractile dysfunction offers a novel platform for human-centric discovery of antiarrhythmic therapies.
Collapse
Affiliation(s)
- Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| | - Tony Rubio
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Robin Springer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Fiona Popp
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Melanie Ritter
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Aiste Liutkute
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Lena Bartelt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Lea Stelzer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Fereshteh Haghighi
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Jan Pietras
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Hendrik Windel
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Núria Díaz i Pedrosa
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | | | - Yannic Doering
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Richard Solano
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Robin Hindmarsh
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
| | - Runzhu Shi
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Tobias Bruegmann
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Ingo Kutschka
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - George Kensah
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany
- Campus-Institute Data Science (CIDAS), University of Göttingen, Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
35
|
Kuebler B, Alvarez-Palomo B, Aran B, Castaño J, Rodriguez L, Raya A, Querol Giner S, Veiga A. Generation of a bank of clinical-grade, HLA-homozygous iPSC lines with high coverage of the Spanish population. Stem Cell Res Ther 2023; 14:366. [PMID: 38093328 PMCID: PMC10720139 DOI: 10.1186/s13287-023-03576-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Induced pluripotent stem cell (iPSC)-derived cell therapies are an interesting new area in the field of regenerative medicine. One of the approaches to decrease the costs of iPSC-derived therapies is the use of allogenic homozygous human leukocyte antigen (HLA)-matched donors to generate iPSC lines and to build a clinical-grade iPSC bank covering a high percentage of the Spanish population. METHODS The Spanish Stem Cell Transplantation Registry was screened for cord blood units (CBUs) homozygous for the most common HLA-A, HLA-B and HLA-DRB1 haplotypes. Seven donors were selected with haplotypes covering 21.37% of the haplotypes of the Spanish population. CD34-positive hematopoietic progenitors were isolated from the mononuclear cell fraction of frozen cord blood units from each donor by density gradient centrifugation and further by immune magnetic labeling and separation using purification columns. Purified CD34 + cells were reprogrammed to iPSCs by transduction with the CTS CytoTune-iPS 2.1 Sendai Reprogramming Kit. RESULTS The iPSCs generated from the 7 donors were expanded, characterized, banked and registered. Master cell banks (MCBs) and working cell banks (WCBs) from the iPSCs of each donor were produced under GMP conditions in qualified clean rooms. CONCLUSIONS Here, we present the first clinical-grade, iPSC haplobank in Spain made from CD34 + cells from seven cord blood units homozygous for the most common HLA-A, HLA-B and HLA-DRB1 haplotypes within the Spanish population. We describe their generation by transduction with Sendai viral vectors and their GMP-compliant expansion and banking. These haplolines will constitute starting materials for advanced therapy medicinal product development (ATMP).
Collapse
Affiliation(s)
- B Kuebler
- Pluripotent Stem Cell Group, Regenerative Medicine Program, Institut d'Investigació Biomédica de Bellvitge (IDIBELL), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
- Program for Translation of Regenerative Medicine in Catalonia (P-[CMRC]), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - B Alvarez-Palomo
- Advanced and Cell Therapy Service, Banc de Sang I Teixits, Edifici Dr. Frederic Duran I Jordà, Passeig de Taulat, 106-116, 08005, Barcelona, Spain
| | - B Aran
- Pluripotent Stem Cell Group, Regenerative Medicine Program, Institut d'Investigació Biomédica de Bellvitge (IDIBELL), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
- Program for Translation of Regenerative Medicine in Catalonia (P-[CMRC]), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - J Castaño
- Advanced and Cell Therapy Service, Banc de Sang I Teixits, Edifici Dr. Frederic Duran I Jordà, Passeig de Taulat, 106-116, 08005, Barcelona, Spain
- Advanced Therapy Platform, Hospital Sant Joan de Déu de Barcelona, Pg. de Sant Joan de Déu, 2, Espluges de Llobregat, 08950, Barcelona, Spain
| | - L Rodriguez
- Advanced and Cell Therapy Service, Banc de Sang I Teixits, Edifici Dr. Frederic Duran I Jordà, Passeig de Taulat, 106-116, 08005, Barcelona, Spain
| | - A Raya
- Program for Translation of Regenerative Medicine in Catalonia (P-[CMRC]), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain.
- Stem Cell Potency Group, Regenerative Medicine Program, Institut d´Investigació Biomédica de Bellvitge (IDIBELL), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain.
- Centre for Networked Biomedical Research On Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain.
| | - S Querol Giner
- Advanced and Cell Therapy Service, Banc de Sang I Teixits, Edifici Dr. Frederic Duran I Jordà, Passeig de Taulat, 106-116, 08005, Barcelona, Spain.
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Autonomous University of Barcelona (UAB), Barcelona, Spain.
| | - A Veiga
- Pluripotent Stem Cell Group, Regenerative Medicine Program, Institut d'Investigació Biomédica de Bellvitge (IDIBELL), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain.
- Program for Translation of Regenerative Medicine in Catalonia (P-[CMRC]), Hospital Duran I Reynals, Gran Via de L'Hospitalet, 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain.
| |
Collapse
|
36
|
Nath SC, Menendez L, Friedrich Ben-Nun I. Overcoming the Variability of iPSCs in the Manufacturing of Cell-Based Therapies. Int J Mol Sci 2023; 24:16929. [PMID: 38069252 PMCID: PMC10706975 DOI: 10.3390/ijms242316929] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Various factors are known to contribute to the diversity of human induced pluripotent stem cells (hiPSCs). Among these are the donor's genetic background and family history, the somatic cell source, the iPSC reprogramming method, and the culture system of choice. Moreover, variability is seen even in iPSC clones, generated in a single reprogramming event, where the donor, somatic cell type, and reprogramming platform are the same. The diversity seen in iPSC lines often translates to epigenetic differences, as well as to differences in the expansion rate, iPSC line culture robustness, and their ability to differentiate into specific cell types. As such, the diversity of iPSCs presents a hurdle to standardizing iPSC-based cell therapy manufacturing. In this review, we will expand on the various factors that impact iPSC diversity and the strategies and tools that could be taken by the industry to overcome the differences amongst various iPSC lines, therefore enabling robust and reproducible iPSC-based cell therapy manufacturing processes.
Collapse
Affiliation(s)
- Suman C. Nath
- Cell Therapy Process Department, Lonza Inc., Houston, TX 77047, USA; (S.C.N.); (L.M.)
| | - Laura Menendez
- Cell Therapy Process Department, Lonza Inc., Houston, TX 77047, USA; (S.C.N.); (L.M.)
| | | |
Collapse
|
37
|
Chehelgerdi M, Behdarvand Dehkordi F, Chehelgerdi M, Kabiri H, Salehian-Dehkordi H, Abdolvand M, Salmanizadeh S, Rashidi M, Niazmand A, Ahmadi S, Feizbakhshan S, Kabiri S, Vatandoost N, Ranjbarnejad T. Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy. Mol Cancer 2023; 22:189. [PMID: 38017433 PMCID: PMC10683363 DOI: 10.1186/s12943-023-01873-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023] Open
Abstract
The advent of iPSCs has brought about a significant transformation in stem cell research, opening up promising avenues for advancing cancer treatment. The formation of cancer is a multifaceted process influenced by genetic, epigenetic, and environmental factors. iPSCs offer a distinctive platform for investigating the origin of cancer, paving the way for novel approaches to cancer treatment, drug testing, and tailored medical interventions. This review article will provide an overview of the science behind iPSCs, the current limitations and challenges in iPSC-based cancer therapy, the ethical and social implications, and the comparative analysis with other stem cell types for cancer treatment. The article will also discuss the applications of iPSCs in tumorigenesis, the future of iPSCs in tumorigenesis research, and highlight successful case studies utilizing iPSCs in tumorigenesis research. The conclusion will summarize the advancements made in iPSC-based tumorigenesis research and the importance of continued investment in iPSC research to unlock the full potential of these cells.
Collapse
Affiliation(s)
- Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fereshteh Behdarvand Dehkordi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Hamidreza Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Mohammad Abdolvand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Sharareh Salmanizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saba Ahmadi
- Department of Molecular and Medical Genetics, Tbilisi State Medical University, Tbilisi, Georgia
| | - Sara Feizbakhshan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saber Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Nasimeh Vatandoost
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tayebeh Ranjbarnejad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
38
|
Seet WT, Mat Afandi MA, Ishak MF, Hassan MNF, Ahmat N, Ng MH, Maarof M. Quality management overview for the production of a tissue-engineered human skin substitute in Malaysia. Stem Cell Res Ther 2023; 14:298. [PMID: 37858277 PMCID: PMC10588160 DOI: 10.1186/s13287-023-03536-9] [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: 06/01/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Treatments for skin injuries have recently advanced tremendously. Such treatments include allogeneic and xenogeneic transplants and skin substitutes such as tissue-engineered skin, cultured cells, and stem cells. The aim of this paper is to discuss the general overview of the quality assurance and quality control implemented in the manufacturing of cell and tissue product, with emphasis on our experience in the manufacturing of MyDerm®, an autologous bilayered human skin substitute. Manufacturing MyDerm® requires multiple high-risk open manipulation steps, such as tissue processing, cell culture expansion, and skin construct formation. To ensure the safety and efficacy of this product, the good manufacturing practice (GMP) facility should establish a well-designed quality assurance and quality control (QA/QC) programme. Standard operating procedures (SOP) should be implemented to ensure that the manufacturing process is consistent and performed in a controlled manner. All starting materials, including tissue samples, culture media, reagents, and consumables must be verified and tested to confirm their safety, potency, and sterility. The final products should also undergo a QC testing series to guarantee product safety, efficacy, and overall quality. The aseptic techniques of cleanroom operators and the environmental conditions of the facility are also important, as they directly influence the manufacturing of good-quality products. Hence, personnel training and environmental monitoring are necessary to maintain GMP compliance. Furthermore, risk management implementation is another important aspect of QA/QC, as it is used to identify and determine the risk level and to perform risk assessments when necessary. Moreover, procedures for non-conformance reporting should be established to identify, investigate, and correct deviations that occur during manufacturing. This paper provides insight and an overview of the QA/QC aspect during MyDerm® manufacturing in a GMP-compliant facility in the Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia.
Collapse
Affiliation(s)
- Wan Tai Seet
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia
| | - Mohd Asyraf Mat Afandi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia
| | - Muhammad Najib Fathi Hassan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia
| | - Nazeha Ahmat
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
39
|
Lin X, Sun Y, Dong X, Liu Z, Sugimura R, Xie G. IPSC-derived CAR-NK cells for cancer immunotherapy. Biomed Pharmacother 2023; 165:115123. [PMID: 37406511 DOI: 10.1016/j.biopha.2023.115123] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023] Open
Abstract
Adoptive cell therapies (ACT) based on chimeric antigen receptor (CAR)-modified immune cells have made great progress with six CAR-T cell products approved by the U.S. FDA for hematological malignancies. Compared with CAR-T cells, CAR-NK cells have attracted increasing attention owing to their multiple killing mechanisms, higher safety profile, and broad sources. Induced pluripotent stem cell (iPSC)-derived NK (iPSC-NK) cells possess a mature phenotype and potent cytolytic activity, and can provide a homogeneous population of CAR-NK cells that can be expanded to clinical scale. Thus, iPSC-derived CAR-NK (CAR-iNK) cells could be used as a standardized and "off-the-shelf" product for cancer immunotherapy. In this review, we summarize the current status of the manufacturing techniques, genetic modification strategies, preclinical and clinical evidence of CAR-iNK cells, and discuss the challenges and future prospects of CAR-iNK cell therapy as a novel cellular immunotherapy in cancer.
Collapse
Affiliation(s)
- Xiaotong Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yao Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zishen Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ryohichi Sugimura
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China.
| | - Guozhu Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
40
|
Zhu F, Nie G, Liu C. Engineered biomaterials in stem cell-based regenerative medicine. LIFE MEDICINE 2023; 2:lnad027. [PMID: 39872549 PMCID: PMC11749850 DOI: 10.1093/lifemedi/lnad027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/17/2023] [Indexed: 01/30/2025]
Abstract
Stem cell-based regenerative therapies, which harness the self-renewal and differentiation properties of stem cells, have been in the spotlight due to their widespread applications in treating degenerative, aging, and other, generally intractable diseases. Therapeutically effective hematopoietic stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells have been used in numerous basic and translational studies with exciting results. However, pre-/post-transplantation issues of poor cell survival and retention, uncontrolled differentiation, and insufficient numbers of cells engrafted into host tissues are the major challenges in stem cell-based regenerative therapies. Engineered biomaterials have adjustable biochemical and biophysical properties that significantly affect cell behaviors, such as cell engraftment, survival, migration, and differentiation outcomes, thereby enhancing the engraftment of implanted stem cells and guiding tissue regeneration. Therefore, the combination of stem cell biology with bioengineered materials is a promising strategy to improve the therapeutic outcomes of stem cell-based regenerative therapy. In this review, we summarize the advances in the modulation of behaviors of stem cells via engineered biomaterials. We then present different approaches to harnessing bioengineered materials to enhance the transplantation of stem cells. Finally, we will provide future directions in regenerative therapy using stem cells.
Collapse
Affiliation(s)
- Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
41
|
Tran LT, Li JY. The role of eye banking with cell-based therapies. Curr Opin Ophthalmol 2023; 34:334-338. [PMID: 37097197 DOI: 10.1097/icu.0000000000000962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
PURPOSE OF REVIEW Cell-based therapies are an exciting new frontier in managing corneal diseases. The introduction of these novel therapies may provide new alternatives to corneal transplantation and decrease the dependence on donor corneal tissue. These changes have the potential to significantly impact eye banking in the future. RECENT FINDINGS The current article reviews current research involving cell-based therapy for treating corneal disorders, including cultivated limbal stem cell transplantation, limbal mesenchymal stem cells for stromal regeneration, and the use of human-cultivated endothelial cells. We will look at barriers to the development and implementation of these therapies. SUMMARY As corneal surgery expands to include cell-based therapies; eye banks will need to redefine their role to support the everchanging landscape of corneal surgery and the decreased demand for corneal donor tissue.
Collapse
Affiliation(s)
- Lillian T Tran
- Department of Ophthalmology & Vision Science, University of California Davis Health, Sacramento, California, USA
| | | |
Collapse
|
42
|
Barrachina L, Arshaghi TE, O'Brien A, Ivanovska A, Barry F. Induced pluripotent stem cells in companion animals: how can we move the field forward? Front Vet Sci 2023; 10:1176772. [PMID: 37180067 PMCID: PMC10168294 DOI: 10.3389/fvets.2023.1176772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
Following a one medicine approach, the development of regenerative therapies for human patients leads to innovative treatments for animals, while pre-clinical studies on animals provide knowledge to advance human medicine. Among many different biological products under investigation, stem cells are among the most prominent. Mesenchymal stromal cells (MSCs) are extensively investigated, but they present challenges such as senescence and limited differentiation ability. Embryonic stem cells (ESCs) are pluripotent cells with a virtually unlimited capacity for self-renewal and differentiation, but the use of embryos carries ethical concerns. Induced pluripotent stem cells (iPSCs) can overcome all of these limitations, as they closely resemble ESCs but are derived from adult cells by reprogramming in the laboratory using pluripotency-associated transcription factors. iPSCs hold great potential for applications in therapy, disease modeling, drug screening, and even species preservation strategies. However, iPSC technology is less developed in veterinary species compared to human. This review attempts to address the specific challenges associated with generating and applying iPSCs from companion animals. Firstly, we discuss strategies for the preparation of iPSCs in veterinary species and secondly, we address the potential for different applications of iPSCs in companion animals. Our aim is to provide an overview on the state of the art of iPSCs in companion animals, focusing on equine, canine, and feline species, as well as to identify which aspects need further optimization and, where possible, to provide guidance on future advancements. Following a "step-by-step" approach, we cover the generation of iPSCs in companion animals from the selection of somatic cells and the reprogramming strategies, to the expansion and characterization of iPSCs. Subsequently, we revise the current applications of iPSCs in companion animals, identify the main hurdles, and propose future paths to move the field forward. Transferring the knowledge gained from human iPSCs can increase our understanding in the biology of pluripotent cells in animals, but it is critical to further investigate the differences among species to develop specific approaches for animal iPSCs. This is key for significantly advancing iPSC application in veterinary medicine, which at the same time will also allow gaining pre-clinical knowledge transferable to human medicine.
Collapse
Affiliation(s)
| | | | | | | | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, University of Galway, Galway, Ireland
| |
Collapse
|
43
|
Suresh Babu S, Duvvuru H, Baker J, Switalski S, Shafa M, Panchalingam KM, Dadgar S, Beller J, Ahmadian Baghbaderani B. Characterization of human induced pluripotent stems cells: Current approaches, challenges, and future solutions. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2023; 37:e00784. [PMID: 36818379 PMCID: PMC9929203 DOI: 10.1016/j.btre.2023.e00784] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/09/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
Human induced pluripotent stem cells (iPSC) have demonstrated massive potentials for use in regenerative and personalized medicine due to their ability to expand in culture and differentiate into specialized cells with therapeutic benefits. However, in order to industrialize iPSC-derived therapies, it is necessary to address the existing challenges surrounding the analytics implemented in the manufacturing process to evaluate and monitor cell expansion, differentiation, and quality of the final products. Here, we review some of the key analytical methods used as part of identity, potency, or safety for in-process or final product release testing and highlighted the challenges and potential solutions for consideration in the Chemistry, Manufacturing and Controls (CMC) strategy for iPSC-based therapies. Some of the challenges associated with characterization and testing of iPSC-based products are related to the choice of analytical technology (to ensure fit-for-purpose), assay reliability and robustness. Automation of analytical methods may be required to reduce hands on time, and improve reliability of the methods through reducing assay variability. Indeed, we have shown that automation of analytical methods is feasible (evaluated using an ELISA based assay) and would result in more precise measurements (demonstrated by lower co-efficient of Variation and standard deviation), less hands-on time, and swift compared to a manually run assay. Therefore, in order to support commercialization of iPSC-based therapies we suggest a well-designed testing strategy to be established in the development phase while incorporating robust, reproducible, reliable, and potentially automated analytics in the manufacturing process.
Collapse
|
44
|
Schoger E, Bleckwedel F, Germena G, Rocha C, Tucholla P, Sobitov I, Möbius W, Sitte M, Lenz C, Samak M, Hinkel R, Varga ZV, Giricz Z, Salinas G, Gross JC, Zelarayán LC. Single-cell transcriptomics reveal extracellular vesicles secretion with a cardiomyocyte proteostasis signature during pathological remodeling. Commun Biol 2023; 6:79. [PMID: 36681760 PMCID: PMC9867722 DOI: 10.1038/s42003-022-04402-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/23/2022] [Indexed: 01/22/2023] Open
Abstract
Aberrant Wnt activation has been reported in failing cardiomyocytes. Here we present single cell transcriptome profiling of hearts with inducible cardiomyocyte-specific Wnt activation (β-catΔex3) as well as with compensatory and failing hypertrophic remodeling. We show that functional enrichment analysis points to an involvement of extracellular vesicles (EVs) related processes in hearts of β-catΔex3 mice. A proteomic analysis of in vivo cardiac derived EVs from β-catΔex3 hearts has identified differentially enriched proteins involving 20 S proteasome constitutes, protein quality control (PQC), chaperones and associated cardiac proteins including α-Crystallin B (CRYAB) and sarcomeric components. The hypertrophic model confirms that cardiomyocytes reacted with an acute early transcriptional upregulation of exosome biogenesis processes and chaperones transcripts including CRYAB, which is ameliorated in advanced remodeling. Finally, human induced pluripotent stem cells (iPSC)-derived cardiomyocytes subjected to pharmacological Wnt activation recapitulated the increased expression of exosomal markers, CRYAB accumulation and increased PQC signaling. These findings reveal that secretion of EVs with a proteostasis signature contributes to early patho-physiological adaptation of cardiomyocytes, which may serve as a read-out of disease progression and can be used for monitoring cellular remodeling in vivo with a possible diagnostic and prognostic role in the future.
Collapse
Affiliation(s)
- Eric Schoger
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075, Göttingen, Germany
| | - Federico Bleckwedel
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
| | - Giulia Germena
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, 37075, Göttingen, Germany
| | - Cheila Rocha
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, 37075, Göttingen, Germany
| | - Petra Tucholla
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
| | - Izzatullo Sobitov
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
| | - Wiebke Möbius
- Max-Planck-Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Maren Sitte
- NGS Integrative Genomics Core Unit (NIG), University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
| | - Christof Lenz
- Department of Clinical Chemistry, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Mostafa Samak
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, 37075, Göttingen, Germany
| | - Rabea Hinkel
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, 37075, Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour (ITTN), Stiftung Tierärztliche Hochschule Hannover, University of Veterinary Medicine, 30173, Hannover, Germany
| | - Zoltán V Varga
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085, Budapest, Hungary
- Pharmahungary Group, H-1085, Budapest, Hungary
| | - Zoltán Giricz
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085, Budapest, Hungary
- Pharmahungary Group, H-1085, Budapest, Hungary
| | - Gabriela Salinas
- NGS Integrative Genomics Core Unit (NIG), University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
| | - Julia C Gross
- Health and Medical University, D-14471, Potsdam, Germany
| | - Laura C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK) partner site Göttingen, 37075, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075, Göttingen, Germany.
| |
Collapse
|
45
|
Torrents S, Grau-Vorster M, Vives J. Illustrative Potency Assay Examples from Approved Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:139-149. [PMID: 37258788 DOI: 10.1007/978-3-031-30040-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Advanced therapy medicinal products (ATMP) encompass a new type of drugs resulting from the manipulation of genes, cells, and tissues to generate innovative medicinal entities with tailored pharmaceutical activity. Definition of suitable potency tests for product release are challenging in this context, in which the active ingredient is composed of living cells and the mechanism of action often is poorly understood. In this chapter, we present and discuss actual potency assays used for the release of representative commercial ATMP from each category of products (namely, KYMRIAH® (tisagenlecleucel), Holoclar® (limbal epithelial stem cells), and PROCHYMAL®/RYONCIL™ (remestemcel-L)). We also examine concerns related to the biological relevance of selected potency assays and challenges ahead for harmonization and broader implementation in compliance with current quality standards and regulatory guidelines.
Collapse
Affiliation(s)
- Sílvia Torrents
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Transfusion Medicine group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Grau-Vorster
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Transfusion Medicine group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquim Vives
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain.
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
46
|
An improved protocol for generation and characterization of human-induced pluripotent stem cell-derived retinal pigment epithelium cells. STAR Protoc 2022; 3:101803. [DOI: 10.1016/j.xpro.2022.101803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
47
|
Schmidt J, Dreha-Kulaczewski S, Zafeiriou MP, Schreiber MK, Wilken B, Funke R, Neuhofer CM, Altmüller J, Thiele H, Nürnberg P, Biskup S, Li Y, Zimmermann WH, Kaulfuß S, Yigit G, Wollnik B. Somatic mosaicism in STAG2-associated cohesinopathies: Expansion of the genotypic and phenotypic spectrum. Front Cell Dev Biol 2022; 10:1025332. [PMID: 36467423 PMCID: PMC9710855 DOI: 10.3389/fcell.2022.1025332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/26/2022] [Indexed: 08/25/2024] Open
Abstract
STAG2 is a component of the large, evolutionarily highly conserved cohesin complex, which has been linked to various cellular processes like genome organization, DNA replication, gene expression, heterochromatin formation, sister chromatid cohesion, and DNA repair. A wide spectrum of germline variants in genes encoding subunits or regulators of the cohesin complex have previously been identified to cause distinct but phenotypically overlapping multisystem developmental disorders belonging to the group of cohesinopathies. Pathogenic variants in STAG2 have rarely been implicated in an X-linked cohesinopathy associated with undergrowth, developmental delay, and dysmorphic features. Here, we describe for the first time a mosaic STAG2 variant in an individual with developmental delay, microcephaly, and hemihypotrophy of the right side. We characterized the grade of mosaicism by deep sequencing analysis on DNA extracted from EDTA blood, urine and buccal swabs. Furthermore, we report an additional female with a novel de novo splice variant in STAG2. Interestingly, both individuals show supernumerary nipples, a feature that has not been reported associated to STAG2 before. Remarkably, additional analysis of STAG2 transcripts in both individuals showed only wildtype transcripts, even after blockage of nonsense-mediated decay using puromycin in blood lymphocytes. As the phenotype of STAG2-associated cohesinopathies is dominated by global developmental delay, severe microcephaly, and brain abnormalities, we investigated the expression of STAG2 and other related components of the cohesin complex during Bioengineered Neuronal Organoids (BENOs) generation by RNA sequencing. Interestingly, we observed a prominent expression of STAG2, especially between culture days 0 and 15, indicating an essential function of STAG2 in early brain development. In summary, we expand the genotypic and phenotypic spectrum of STAG2-associated cohesinopathies and show that BENOs represent a promising model to gain further insights into the critical role of STAG2 in the complex process of nervous system development.
Collapse
Affiliation(s)
- Julia Schmidt
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Steffi Dreha-Kulaczewski
- Department of Pediatics and Adolescent Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Maria-Patapia Zafeiriou
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Marie-Kristin Schreiber
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Bernd Wilken
- Department of Pediatric Neurology, Klinikum Kassel, Kassel, Germany
| | - Rudolf Funke
- Department of Pediatric Neurology, Klinikum Kassel, Kassel, Germany
| | - Christiane M Neuhofer
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- Institute of Neurogenomics, Helmholtz Zentrum Munich, Munich, Germany
- Department of Neurology, Friedrich-Baur-Institute, LMU Hospital, Ludwig Maximilians University, Munich, Germany
| | - Janine Altmüller
- Cologne Center for Genomics (CCG), University of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Berlin Institute of Health at Charité, Core Facility Genomics, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG), University of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Saskia Biskup
- CeGaT GmbH, Center for Genomics and Transcriptomics, Tübingen, Germany
| | - Yun Li
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfram Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Silke Kaulfuß
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
48
|
iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine. J Pers Med 2022; 12:jpm12091485. [PMID: 36143270 PMCID: PMC9500601 DOI: 10.3390/jpm12091485] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022] Open
Abstract
In the past, several animal disease models were developed to study the molecular mechanism of neurological diseases and discover new therapies, but the lack of equivalent animal models has minimized the success rate. A number of critical issues remain unresolved, such as high costs for developing animal models, ethical issues, and lack of resemblance with human disease. Due to poor initial screening and assessment of the molecules, more than 90% of drugs fail during the final step of the human clinical trial. To overcome these limitations, a new approach has been developed based on induced pluripotent stem cells (iPSCs). The discovery of iPSCs has provided a new roadmap for clinical translation research and regeneration therapy. In this article, we discuss the potential role of patient-derived iPSCs in neurological diseases and their contribution to scientific and clinical research for developing disease models and for developing a roadmap for future medicine. The contribution of humaniPSCs in the most common neurodegenerative diseases (e.g., Parkinson’s disease and Alzheimer’s disease, diabetic neuropathy, stroke, and spinal cord injury) were examined and ranked as per their published literature on PUBMED. We have observed that Parkinson’s disease scored highest, followed by Alzheimer’s disease. Furthermore, we also explored recent advancements in the field of personalized medicine, such as the patient-on-a-chip concept, where iPSCs can be grown on 3D matrices inside microfluidic devices to create an in vitro disease model for personalized medicine.
Collapse
|
49
|
Abberton KM, McDonald TL, Diviney M, Holdsworth R, Leslie S, Delatycki MB, Liu L, Klamer G, Johnson P, Elwood NJ. Identification and Re-consent of Existing Cord Blood Donors for Creation of Induced Pluripotent Stem Cell Lines for Potential Clinical Applications. Stem Cells Transl Med 2022; 11:1052-1060. [PMID: 36073721 PMCID: PMC9585951 DOI: 10.1093/stcltm/szac060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
We aim to create a bank of clinical grade cord blood-derived induced pluripotent stem cell lines in order to facilitate clinical research leading to the development of new cellular therapies. Here we present a clear pathway toward the creation of such a resource, within a strong quality framework, and with the appropriate regulatory, government and ethics approvals, along with a dynamic follow-up and re-consent process of cord blood donors from the public BMDI Cord Blood Bank. Interrogation of the cord blood bank inventory and next generation sequencing was used to identify and confirm 18 donors with suitable HLA homozygous haplotypes. Regulatory challenges that may affect global acceptance of the cell lines, along with the quality standards required to operate as part of a global network, are being met by working in collaboration with bodies such as the International Stem Cell Banking Initiative (ISCBI) and the Global Alliance for iPSC Therapies (GAiT). Ethics approval was granted by an Institutional Human Research Ethics Committee, and government approval has been obtained to use banked cord blood for this purpose. New issues of whole-genome sequencing and the relevant donor safeguards and protections were considered with input from clinical genetics services, including the rights and information flow to donors, and commercialization aspects. The success of these processes has confirmed feasibility and utility of using banked cord blood to produce clinical-grade iPSC lines for potential cellular therapies.
Collapse
Affiliation(s)
- Keren M Abberton
- BMDI Cord Blood Bank, Melbourne, Australia.,Murdoch Children's Research Institute, Melbourne, Australia.,Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Tricia L McDonald
- BMDI Cord Blood Bank, Melbourne, Australia.,Murdoch Children's Research Institute, Melbourne, Australia
| | - Mary Diviney
- VTIS at Australian Red Cross Lifeblood, Melbourne, Australia
| | | | - Stephen Leslie
- Schools of Mathematics and Statistics, and BioSciences, Melbourne Integrative Genomics, University of Melbourne, Melbourne, Australia
| | - Martin B Delatycki
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Pediatrics, University of Melbourne, Melbourne, Australia.,Victorian Clinical Genetics Services, Melbourne, Australia
| | - Lin Liu
- BMDI Cord Blood Bank, Melbourne, Australia.,Murdoch Children's Research Institute, Melbourne, Australia
| | - Guy Klamer
- Sydney Cord Blood Bank, Sydney Children's Hospitals Network, Sydney, Australia
| | - Phillip Johnson
- Queensland Cord Blood Bank At The Mater, Brisbane, Australia
| | - Ngaire J Elwood
- BMDI Cord Blood Bank, Melbourne, Australia.,Murdoch Children's Research Institute, Melbourne, Australia.,Department of Pediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
50
|
Nakashima Y, Yoshida S, Tsukahara M. Semi-3D cultures using Laminin 221 as a coating material for human induced pluripotent stem cells. Regen Biomater 2022; 9:rbac060. [PMID: 36176714 PMCID: PMC9514851 DOI: 10.1093/rb/rbac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/09/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022] Open
Abstract
It was previously believed that human induced pluripotent stem cells (hiPSCs) did not show adhesion to the coating material Laminin 221, which is known to have specific affinity for cardiomyocytes. In this study, we report that human mononuclear cell-derived hiPSCs, established with Sendai virus vector, form peninsular-like colonies rather than embryonic stem cell-like colonies; these peninsular-like colonies can be passaged more than 10 times after establishment. Additionally, initialization-deficient cells with residual Sendai virus vector adhered to the coating material Laminin 511 but not to Laminin 221. Therefore, the expression of undifferentiated markers tended to be higher in hiPSCs established on Laminin 221 than on Laminin 511. On Laminin 221, hiPSCs15M66 showed a semi-floating colony morphology. The expression of various markers of cell polarity was significantly lower in hiPSCs cultured on Laminin 221 than in hiPSCs cultured on Laminin 511. Furthermore, 201B7 and 15M66 hiPSCs showed 3D cardiomyocyte differentiation on Laminin 221. Thus, the coating material Laminin 221 provides semi-floating culture conditions for the establishment, culture and induced differentiation of hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Shinsuke Yoshida
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| |
Collapse
|