1
|
Jacob S, Jacob SA, Thoppil J. Targeting sepsis through inflammation and oxidative metabolism. World J Crit Care Med 2025; 14:101499. [DOI: 10.5492/wjccm.v14.i1.101499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024] Open
Abstract
Infection is a public health problem and represents a spectrum of disease that can result in sepsis and septic shock. Sepsis is characterized by a dysregulated immune response to infection. Septic shock is the most severe form of sepsis which leads to distributive shock and high mortality rates. There have been significant advances in sepsis management mainly focusing on early identification and therapy. However, complicating matters is the lack of reliable diagnostic tools and the poor specificity and sensitivity of existing scoring tools i.e., systemic inflammatory response syndrome criteria, sequential organ failure assessment (SOFA), or quick SOFA. These limitations have underscored the modest progress in reducing sepsis-related mortality. This review will focus on novel therapeutics such as oxidative stress targets, cytokine modulation, endothelial cell modulation, etc., that are being conceptualized for the management of sepsis and septic shock.
Collapse
Affiliation(s)
- Salena Jacob
- Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328, United States
| | - Sanjana Ann Jacob
- Dell School of Medicine, University of Texas at Austin Medical School, Austin, TX 78712, United States
| | - Joby Thoppil
- Emergency Medicine, UT Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
2
|
AlOraibi S, Taurin S, Alshammary S. Advancements in Umbilical Cord Biobanking: A Comprehensive Review of Current Trends and Future Prospects. Stem Cells Cloning 2024; 17:41-58. [PMID: 39655226 PMCID: PMC11626973 DOI: 10.2147/sccaa.s481072] [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: 06/02/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024] Open
Abstract
Biobanking has emerged as a transformative concept in advancing the medical field, particularly with the exponential growth of umbilical cord (UC) biobanking in recent decades. UC blood and tissue provide a rich source of primitive hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) for clinical transplantation, offering distinct advantages over alternative adult stem cell sources. However, to fully realize the therapeutic potential of UC-derived stem cells and establish a comprehensive global UC-biobanking network, it is imperative to optimize and standardize UC processing, cryopreservation methods, quality control protocols, and regulatory frameworks, alongside developing effective consent provisions. This review aims to comprehensively explore recent advancements in UC biobanking, focusing on the establishment of rigorous safety and quality control procedures, the standardization of biobanking operations, and the optimization and automation of UC processing and cryopreservation techniques. Additionally, the review examines the expanded clinical applications of UC stem cells, addresses the challenges associated with umbilical cord biobanking and UC-derived stem cell therapies, and discusses the promising role of artificial intelligence (AI) in enhancing various operational aspects of biobanking, streamlining data processing, and improving data analysis accuracy while ensuring compliance with safety and quality standards. By addressing these critical areas, this review seeks to provide insights into the future direction of UC biobanking and its potential to significantly impact regenerative medicine.
Collapse
Affiliation(s)
- Sahar AlOraibi
- Molecular Medicine Department, Princess Al Jawhara Center for Molecular Medicine, Genetics, and Hereditary Diseases, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| | - Sebastien Taurin
- Molecular Medicine Department, Princess Al Jawhara Center for Molecular Medicine, Genetics, and Hereditary Diseases, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| | - Sfoug Alshammary
- Molecular Medicine Department, Princess Al Jawhara Center for Molecular Medicine, Genetics, and Hereditary Diseases, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
3
|
Li Z, Qi J, Fu S, Luan J, Wang Q. Effects of nanographene oxide on adipose-derived stem cell cryopreservation. Cell Tissue Bank 2024; 25:805-830. [PMID: 38844606 DOI: 10.1007/s10561-024-10140-5] [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: 12/12/2023] [Accepted: 05/17/2024] [Indexed: 09/06/2024]
Abstract
Cryoinjury mitigation is key in cell cryopreservation. Here, we aimed to assess the effectiveness of nanographene oxide (nano-GO) for improving cryoprotectant agents (CPAs) in human adipose stem cell (hADSC) cryopreservation. For in vitro experiments, nano-GO (5 μg/mL) was added to the CPAs in the control, and passage (P) 2 hADSCs were collected and cryopreserved for around two weeks. We compared cytotoxicity, cell viability, immunophenotypes, proliferation, cell apoptosis, and tri-lineage differentiation. In vivo, studies used lipoaspirate to create non-enriched or hADSC-enriched fat tissues by combining it with PBS or hADSCs cryopreserved with the aforementioned CPAs. Each nude mouse received a 0.3 mL subcutaneous injection of the graft. At 12 weeks, the grafts were harvested. Histology, adipocyte-associated genes and protein, vascular density and angiogenic cytokines, macrophage infiltration, and inflammatory cytokines were analyzed. Nano-GO CPA contributed to increased cell viability, improved cell recovery, and lowered levels of early apoptosis. Nano GO at concentrations of 0.01-100 μg/mL caused no cytotoxicity to hADSCs. The absence of nano GOs in the intracellular compartments of the cells was confirmed by transmission electron microscopy. The fat grafts from the CPA-GO group showed more viable adipocytes and significantly increased angiogenesis compared to the PBS and CPA-C groups. Adding hADSCs from the CPA-GO group to the graft reduced macrophage infiltration and MCP-1 expression. Nano-GO plays an anti-apoptotic role in the cryopreservation of hADSCs, which could improve the survival of transplanted fat tissues, possibly via improved angiogenesis and lower inflammatory response in the transplanted adipose tissue.
Collapse
Affiliation(s)
- Zifei Li
- Facial and Cervical Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan, Beijing, 100144, People's Republic of China
| | - Jun Qi
- Breast Plastic and Reconstructive Surgery Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China
| | - Su Fu
- Breast Plastic and Reconstructive Surgery Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China
| | - Jie Luan
- Breast Plastic and Reconstructive Surgery Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China.
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 33 Badachu Road, Shijingshan Dist., Beijing, 100144, People's Republic of China.
| |
Collapse
|
4
|
Safwan M, Bourgleh MS, Aldoush M, Haider KH. Tissue-source effect on mesenchymal stem cells as living biodrugs for heart failure: Systematic review and meta-analysis. World J Cardiol 2024; 16:469-483. [PMID: 39221190 PMCID: PMC11362808 DOI: 10.4330/wjc.v16.i8.469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/24/2024] [Accepted: 07/23/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), as living biodrugs, have entered advanced phases of clinical assessment for cardiac function restoration in patients with myocardial infarction and heart failure. While MSCs are available from diverse tissue sources, bone-marrow-derived MSCs (BM-MSCs) remain the most well-studied cell type, besides umbilical-cord-derived MSCs (UC-MSCs). The latter offers advantages, including noninvasive availability without ethical considerations.
AIM To compare the safety and efficacy of BM-MSCs and UC-MSCs in terms of left ventricular ejection fraction (LVEF), 6-min walking distance (6MWD), and major adverse cardiac events (MACEs).
METHODS Five databases were systematically searched to identify randomized controlled trials (RCTs). Thirteen RCTs (693 patients) were included using predefined eligibility criteria. Weighted mean differences and odds ratio (OR) for the changes in the estimated treatment effects.
RESULTS UC-MSCs significantly improved LVEF vs controls by 5.08% [95% confidence interval (CI): 2.20%-7.95%] at 6 mo and 2.78% (95%CI: 0.86%-4.70%) at 12 mo. However, no significant effect was observed for BM-MSCs vs controls. No significant changes were observed in the 6MWD with either of the two cell types. Also, no differences were observed for MACEs, except rehospitalization rates, which were lower only with BM-MSCs (odds ratio 0.48, 95%CI: 0.24-0.97) vs controls.
CONCLUSION UC-MSCs significantly improved LVEF compared with BM-MSCs. Their advantageous characteristics position them as a promising alternative to MSC-based therapy.
Collapse
Affiliation(s)
- Moaz Safwan
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| | - Mariam Safwan Bourgleh
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| | - Mohamed Aldoush
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| | - Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| |
Collapse
|
5
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2024:S2090-1232(24)00181-4. [PMID: 38729561 DOI: 10.1016/j.jare.2024.05.004] [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: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
6
|
Svoradová A, Vašíček J, Zmrhal V, Venusová E, Pavlík A, Bauer M, Olexiková L, Langraf V, Sláma P, Chrenek P. Mesenchymal stem cells of Oravka chicken breed: promising path to biodiversity conservation. Poult Sci 2023; 102:102807. [PMID: 37302325 PMCID: PMC10276279 DOI: 10.1016/j.psj.2023.102807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 06/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multilineage cells able to differentiate into other cell types. MSCs derived from bone marrow or compact bones are the most accessible stem cells used in tissue engineering. Therefore, the aim of this study was to isolate, characterize and cryopreserve MSCs of endangered Oravka chicken breed. MSCs were obtained from compact bones of the femur and tibiotarsus. MSCs were spindle-shaped and were able to differentiate into osteo-, adipo-, and chondrocytes under the specific differentiation conditions. Furthermore, MSCs were positive for surface markers such as CD29, CD44, CD73, CD90, CD105, CD146 and negative for CD34CD45 by flow cytometry. Moreover, MSCs demonstrated high positivity of "stemness" markers aldehyde dehydrogenase, alkaline phosphatase as well as for intracellular markers vimentin, desmin, α-SMA. Subsequently, MSCs were cryopreserved using 10% dimethyl sulfoxide in liquid nitrogen. Based on the results from the viability, phenotype, and ultrastructure assessment we can concluded that the MSCs were not negatively affected by the cryopreservation. Finally, MSCs of endangered Oravka chicken breed were successfully stored in animal gene bank, thus making them a valuable genetic resource.
Collapse
Affiliation(s)
- Andrea Svoradová
- Institute of Farm Animal Genetics and Reproduction, NPPC, Research Institute for Animal Production in Nitra, Nitra, Slovakia; Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Jaromír Vašíček
- Institute of Farm Animal Genetics and Reproduction, NPPC, Research Institute for Animal Production in Nitra, Nitra, Slovakia; Institute of Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Vladimír Zmrhal
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Eva Venusová
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Aleš Pavlík
- Laboratory of Animal Physiology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Miroslav Bauer
- Institute of Farm Animal Genetics and Reproduction, NPPC, Research Institute for Animal Production in Nitra, Nitra, Slovakia; Department of Botany and Genetics, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Lucia Olexiková
- Institute of Farm Animal Genetics and Reproduction, NPPC, Research Institute for Animal Production in Nitra, Nitra, Slovakia
| | - Vladimír Langraf
- Department of Zoology and Anthropology, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Petr Sláma
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Peter Chrenek
- Institute of Farm Animal Genetics and Reproduction, NPPC, Research Institute for Animal Production in Nitra, Nitra, Slovakia; Institute of Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Nitra, Slovakia.
| |
Collapse
|
7
|
Li CH, Zhao J, Zhang HY, Wang B. Banking of perinatal mesenchymal stem/stromal cells for stem cell-based personalized medicine over lifetime: Matters arising. World J Stem Cells 2023; 15:105-119. [PMID: 37181005 PMCID: PMC10173813 DOI: 10.4252/wjsc.v15.i4.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/07/2023] [Accepted: 03/22/2023] [Indexed: 04/26/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are currently applied in regenerative medicine and tissue engineering. Numerous clinical studies have indicated that MSCs from different tissue sources can provide therapeutic benefits for patients. MSCs derived from either human adult or perinatal tissues have their own unique advantages in their medical practices. Usually, clinical studies are conducted by using of cultured MSCs after thawing or short-term cryopreserved-then-thawed MSCs prior to administration for the treatment of a wide range of diseases and medical disorders. Currently, cryogenically banking perinatal MSCs for potential personalized medicine for later use in lifetime has raised growing interest in China as well as in many other countries. Meanwhile, this has led to questions regarding the availability, stability, consistency, multipotency, and therapeutic efficiency of the potential perinatal MSC-derived therapeutic products after long-term cryostorage. This opinion review does not minimize any therapeutic benefit of perinatal MSCs in many diseases after short-term cryopreservation. This article mainly describes what is known about banking perinatal MSCs in China and, importantly, it is to recognize the limitation and uncertainty of the perinatal MSCs stored in cryobanks for stem cell medical treatments in whole life. This article also provides several recommendations for banking of perinatal MSCs for potentially future personalized medicine, albeit it is impossible to anticipate whether the donor will benefit from banked MSCs during her/his lifetime.
Collapse
Affiliation(s)
- Cheng-Hai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Jing Zhao
- Department of Clinical Laboratory, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Hong-Yan Zhang
- Department of Pharmacy, Fuwai Central China Cardiovascular Hospital, Zhengzhou 450000, Henan Province, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China.
| |
Collapse
|
8
|
Yan S, Campos de Souza S, Xie Z, Bao Y. Research progress in clinical trials of stem cell therapy for stroke and neurodegenerative diseases. IBRAIN 2023; 9:214-230. [PMID: 37786546 PMCID: PMC10529019 DOI: 10.1002/ibra.12095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 10/04/2023]
Abstract
The incidence of stroke and neurodegenerative diseases is gradually increasing in modern society, but there is still no treatment that is effective enough. Stem cells are cells that can reproduce (self-renew) and differentiate into the body, which have shown significance in basic research, while doctors have also taken them into clinical trials to determine their efficacy and safety. Existing clinical trials mainly include middle-aged and elderly patients with stroke or Parkinson's disease (mostly 40-80 years old), mainly involving injection of mesenchymal stem cells and bone marrow mesenchymal stem cells through the veins and the putamen, with a dosage of mostly 106-108 cells. The neural and motor functions of the patients were restored after stem cell therapy, and the safety was found to be good during the follow-up period of 3 months to 5 years. Here, we review all clinical trials and the latest advances in stroke, Alzheimer's disease, and Parkinson's disease, with the hope that stem cell therapy will be used in the clinic in the future to achieve effective treatment rates and benefit patients.
Collapse
Affiliation(s)
- Shan‐Shan Yan
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Senio Campos de Souza
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Zhen‐Dong Xie
- Institute for Bioengineering of CataloniaUniversity of BarcelonaCarrer de Baldiri ReixacBarcelonaSpain
| | - Yong‐Xin Bao
- Qingdao Women and Children's HospitalQingdao UniversityQingdaoChina
| |
Collapse
|
9
|
Hypothermic Preservation of Adipose-Derived Mesenchymal Stromal Cells as a Viable Solution for the Storage and Distribution of Cell Therapy Products. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120805. [PMID: 36551011 PMCID: PMC9774331 DOI: 10.3390/bioengineering9120805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Cell and gene therapies (CGT) have reached new therapeutic targets but have noticeably high prices. Solutions to reduce production costs might be found in CGT storage and transportation since they typically involve cryopreservation, which is a heavily burdened process. Encapsulation at hypothermic temperatures (e.g., 2-8 °C) could be a feasible alternative. Adipose tissue-derived mesenchymal stromal cells (MSC(AT)) expanded using fetal bovine serum (FBS)- (MSC-FBS) or human platelet lysate (HPL)-supplemented mediums (MSC-HPL) were encapsulated in alginate beads for 30 min, 5 days, and 12 days. After bead release, cell recovery and viability were determined to assess encapsulation performance. MSC identity was verified by flow cytometry, and a set of assays was performed to evaluate functionality. MSC(AT) were able to survive encapsulated for a standard transportation period of 5 days, with recovery values of 56 ± 5% for MSC-FBS and 77 ± 6% for MSC-HPL (which is a negligible drop compared to earlier timepoints). Importantly, MSC function did not suffer from encapsulation, with recovered cells showing robust differentiation potential, expression of immunomodulatory molecules, and hematopoietic support capacity. MSC(AT) encapsulation was proven possible for a remarkable 12 day period. There is currently no solution to completely replace cryopreservation in CGT logistics and supply chain, although encapsulation has shown potential to act as a serious competitor.
Collapse
|
10
|
Linkova DD, Rubtsova YP, Egorikhina MN. Cryostorage of Mesenchymal Stem Cells and Biomedical Cell-Based Products. Cells 2022; 11:cells11172691. [PMID: 36078098 PMCID: PMC9454587 DOI: 10.3390/cells11172691] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) manifest vast opportunities for clinical use due both to their ability for self-renewal and for effecting paracrine therapeutic benefits. At the same time, difficulties with non-recurrent generation of large numbers of cells due to the necessity for long-term MSC expansion ex vivo, or the requirement for repeated sampling of biological material from a patient significantly limits the current use of MSCs in clinical practice. One solution to these problems entails the creation of a biobank using cell cryopreservation technology. This review is aimed at analyzing and classifying literature data related to the development of protocols for the cryopreservation of various types of MSCs and tissue-engineered structures. The materials in the review show that the existing techniques and protocols for MSC cryopreservation are very diverse, which significantly complicates standardization of the entire process. Here, the selection of cryoprotectors and of cryoprotective media shows the greatest variability. Currently, it is the cryopreservation of cell suspensions that has been studied most extensively, whereas there are very few studies in the literature on the freezing of intact tissues or of tissue-engineered structures. However, even now it is possible to develop general recommendations to optimize the cryopreservation process, making it less traumatic for cells.
Collapse
|
11
|
Li T, Zhou L, Fan M, Chen Z, Yan L, Lu H, Jia M, Wu H, Shan L. Human Umbilical Cord-Derived Mesenchymal Stem Cells Ameliorate Skin Aging of Nude Mice Through Autophagy-Mediated Anti-Senescent Mechanism. Stem Cell Rev Rep 2022; 18:2088-2103. [PMID: 35864432 DOI: 10.1007/s12015-022-10418-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
Skin aging is a currently irreversible process, affected by increased oxidative stress, activated cellular senescence, and lacked regeneration of the dermal layer. Mesenchymal stem cells (MSCs), such as human umbilical cord-derived MSCs (hucMSCs), have pro-regeneration and anti-aging potencies. To explore whether hucMSCs can be used to treat skin aging, this study employed skin-aging model of nude mice to conduct in vivo assays, including biochemical analysis of superoxide dismutase (SOD) and malondialdehyde (MDA), gross observation, histopathological observation, and immunohistochemical analysis. To clarify how hucMSCs work on skin aging, this study employed skin-aging model of human dermal fibroblasts (HDFs) to conduct in vitro assays by applying conditional medium of hucMSCs (CMM), including wound healing assay, senescence staining, flow cytometric oxidative detection, real time PCR, and western blot analysis. The in vivo data demonstrated that hucMSCs dose-dependently removed wrinkles, smoothed skin texture, and increased dermal thickness and collagen production of aged skin by reversing SOD and MDA levels and up-regulating Col-1 and VEGF expressions, indicating anti-oxidative and pro-regenerative effects against skin aging. The in vitro data revealed that hucMSCs significantly reversed the senescence of HDFs by promoting cell migration, inhibiting ROS production, and restoring the overexpressions of oxidative and senescent markers through paracrine mode of action, and the paracrine mechanism was mediated by the inhibition of autophagy. This study provided novel knowledge regarding the anti-aging efficacy and paracrine mechanism of hucMSCs on skin, making hucMSCs-based therapy a promising regime for skin aging treatment.
Collapse
Affiliation(s)
- Ting Li
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqiang Fan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Haishan Lu
- Department of Dermatology, PLA 903 Hospital, Hangzhou, China
| | - Ming Jia
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiling Wu
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China. .,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China. .,Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China.
| |
Collapse
|
12
|
Dave C, Mei SHJ, McRae A, Hum C, Sullivan KJ, Champagne J, Ramsay T, McIntyre L. Comparison of freshly cultured versus cryopreserved mesenchymal stem cells in animal models of inflammation: A pre-clinical systematic review. eLife 2022; 11:75053. [PMID: 35838024 PMCID: PMC9286731 DOI: 10.7554/elife.75053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/05/2022] [Indexed: 12/09/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) are multipotent cells that demonstrate therapeutic potential for the treatment of acute and chronic inflammatory-mediated conditions. Although controversial, some studies suggest that MSCs may lose their functionality with cryopreservation which could render them non-efficacious. Hence, we conducted a systematic review of comparative pre-clinical models of inflammation to determine if there are differences in in vivo measures of pre-clinical efficacy (primary outcomes) and in vitro potency (secondary outcomes) between freshly cultured and cryopreserved MSCs. Methods: A systematic search on OvidMEDLINE, EMBASE, BIOSIS, and Web of Science (until January 13, 2022) was conducted. The primary outcome included measures of in vivo pre-clinical efficacy; secondary outcomes included measures of in vitro MSC potency. Risk of bias was assessed by the SYRCLE ‘Risk of Bias’ assessment tool for pre-clinical studies. Results: Eighteen studies were included. A total of 257 in vivo pre-clinical efficacy experiments represented 101 distinct outcome measures. Of these outcomes, 2.3% (6/257) were significantly different at the 0.05 level or less; 2 favoured freshly cultured and 4 favoured cryopreserved MSCs. A total of 68 in vitro experiments represented 32 different potency measures; 13% (9/68) of the experiments were significantly different at the 0.05 level or less, with seven experiments favouring freshly cultured MSC and two favouring cryopreserved MSCs. Conclusions: The majority of preclinical primary in vivo efficacy and secondary in vitro potency outcomes were not significantly different (p<0.05) between freshly cultured and cryopreserved MSCs. Our systematic summary of the current evidence base may provide MSC basic and clinical research scientists additional rationale for considering a cryopreserved MSC product in their pre-clinical studies and clinical trials as well as help identify research gaps and guide future related research. Funding: Ontario Institute for Regenerative Medicine
Collapse
Affiliation(s)
- Chintan Dave
- Division of Critical Care Medicine, Department of Medicine, Western University, London, Canada
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Andrea McRae
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Christine Hum
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada.,University of Ottawa, Ottawa, Canada
| | - Katrina J Sullivan
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Josee Champagne
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Tim Ramsay
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Lauralyn McIntyre
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada.,Division of Critical Care, Department of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
13
|
Impact of Cryopreservation and Freeze-Thawing on Therapeutic Properties of Mesenchymal Stromal/Stem Cells and Other Common Cellular Therapeutics. CURRENT STEM CELL REPORTS 2022; 8:72-92. [PMID: 35502223 PMCID: PMC9045030 DOI: 10.1007/s40778-022-00212-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/19/2022]
Abstract
Purpose of Review Cryopreservation and its associated freezing and thawing procedures–short “freeze-thawing”–are among the final steps in economically viable manufacturing and clinical application of diverse cellular therapeutics. Translation from preclinical proof-of-concept studies to larger clinical trials has indicated that these processes may potentially present an Achilles heel to optimal cell product safety and particularly efficacy in clinical trials and routine use. Recent Findings We review the current state of the literature on how cryopreservation of cellular therapies has evolved and how the application of this technique to different cell types is interlinked with their ability to engraft and function upon transfer in vivo, in particular for hematopoietic stem and progenitor cells (HSPCs), their progeny, and therapeutic cell products derived thereof. We also discuss pros and cons how this may differ for non-hematopoietic mesenchymal stromal/stem cell (MSC) therapeutics. We present different avenues that may be crucial for cell therapy optimization, both, for hematopoietic (e.g., effector, regulatory, and chimeric antigen receptor (CAR)-modified T and NK cell based products) and for non-hematopoietic products, such as MSCs and induced pluripotent stem cells (iPSCs), to achieve optimal viability, recovery, effective cell dose, and functionality of the cryorecovered cells. Summary Targeted research into optimizing the cryopreservation and freeze-thawing routines and the adjunct manufacturing process design may provide crucial advantages to increase both the safety and efficacy of cellular therapeutics in clinical use and to enable effective market deployment strategies to become economically viable and sustainable medicines.
Collapse
|
14
|
Bačenková D, Trebuňová M, Čížková D, Hudák R, Dosedla E, Findrik-Balogová A, Živčák J. In Vitro Model of Human Trophoblast in Early Placentation. Biomedicines 2022; 10:biomedicines10040904. [PMID: 35453654 PMCID: PMC9029210 DOI: 10.3390/biomedicines10040904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 12/26/2022] Open
Abstract
The complex process of placental implantation and development affects trophoblast progenitors and uterine cells through the regulation of transcription factors, cytokines, adhesion receptors and their ligands. Differentiation of trophoblast precursors in the trophectoderm of early ontogenesis, caused by the transcription factors, such as CDX2, TEAD4, Eomes and GATA3, leads to the formation of cytotrophoblast and syncytiotrophoblast populations. The molecular mechanisms involved in placental formation inside the human body along with the specification and differentiation of trophoblast cell lines are, mostly due to the lack of suitable cell models, not sufficiently elucidated. This review is an evaluation of current technologies, which are used to study the behavior of human trophoblasts and other placental cells, as well as their ability to represent physiological conditions both in vivo and in vitro. An in vitro 3D model with a characteristic phenotype is of great benefit for the study of placental physiology. At the same time, it provides great support for future modeling of placental disease.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
- Correspondence: ; Tel.: +42-1055-602-2380
| | - Marianna Trebuňová
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| | - Daša Čížková
- Centre for Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy, 04181 Košice, Slovakia;
| | - Radovan Hudák
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| | - Erik Dosedla
- Department of Gynecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafarik Univerzity Hospital AGEL Košice-Šaca, Pavol Jozef Šafarik University in Košice, 04015 Košice-Šaca, Slovakia;
| | - Alena Findrik-Balogová
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| | - Jozef Živčák
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| |
Collapse
|
15
|
Nguyen MQ, Bui HTH, Tuyet ANT, Nhung TTH, Hoang DM, Liem NT, Hoang VT. Comparative Bioactivity Analysis for Off-the-Shelf and Culture-Rescued Umbilical Cord-Derived Mesenchymal Stem/Stromal Cells in a Xeno- and Serum-Free Culture System. Cell Transplant 2021; 30:9636897211039441. [PMID: 34538123 PMCID: PMC8718162 DOI: 10.1177/09636897211039441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We recently reported a standardized xeno- and serum-free culture platform to isolate and expand umbilical cord-derived mesenchymal stem/stromal cells (UC-MSCs). Comparing populations from the same passage, cells that were cryopreserved and culture-rescued exhibited characteristics similar to those of their fresh counterparts, continuously cultured cells without interim cryopreservation. The culture rescue after thawing allowed for the cells to be fully recovered. However, since it would be more cost-effective and timesaving if cryopreserved cells can be used as an off-the-shelf product, we set out to compare the bioactivity of freshly thawed UC-MSCs versus culture-rescued UC-MSCs of the same batch that were recultured for an additional passage under our xeno- and serum-free protocol. UC-MSCs showed high viability in both the freshly thawed and the re-cultured group. Both populations displayed a similar proliferation capacity which is indicated by a comparable population doubling time and colony-forming ability. Both freshly thawed and culture-rescued UC-MSCs expressed the characteristic immunophenotype and were capable of differentiating into osteocytes, chondrocytes, and adipocytes. On the other hand, culture-rescued cells appeared to be more potent in immunosuppression than freshly thawed cells. In conclusion, freshly thawed and culture-rescued cell products share comparable bioactivity in cell growth and proliferation, immunophenotype, and differentiation potential. However, the culture-rescued cells that were allowed to grow for an additional passage appear to display a more favorable immunomodulatory potential when compared to their freshly thawed parent cells.
Collapse
Affiliation(s)
- Minh Quang Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam.,VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Hue T H Bui
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam.,Vinmec Institute of Applied Science and Regenerative Medicine (VIASRM), Vinmec Healthcare System, Hanoi, Vietnam
| | - Anh Nguyen Thi Tuyet
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam.,Vinmec Institute of Applied Science and Regenerative Medicine (VIASRM), Vinmec Healthcare System, Hanoi, Vietnam
| | - Trinh Thi Hong Nhung
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam.,Vinmec Institute of Applied Science and Regenerative Medicine (VIASRM), Vinmec Healthcare System, Hanoi, Vietnam
| | - Duc M Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam
| | - Nguyen Thanh Liem
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam
| | - Van T Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Healthcare System, Hanoi, Vietnam
| |
Collapse
|
16
|
Raposo L, Lourenço AP, Nascimento DS, Cerqueira R, Cardim N, Leite-Moreira A. Human umbilical cord tissue-derived mesenchymal stromal cells as adjuvant therapy for myocardial infarction: a review of current evidence focusing on pre-clinical large animal models and early human trials. Cytotherapy 2021; 23:974-979. [PMID: 34112613 DOI: 10.1016/j.jcyt.2021.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022]
Abstract
Although biologically appealing, the concept of tissue regeneration underlying first- and second-generation cell therapies has failed to translate into consistent results in clinical trials. Several types of cells from different origins have been tested in pre-clinical models and in patients with acute myocardial infarction (AMI). Mesenchymal stromal cells (MSCs) have gained attention because of their potential for immune modulation and ability to promote endogenous tissue repair, mainly through their secretome. MSCs can be easily obtained from several human tissues, the umbilical cord being the most abundant source, and further expanded in culture, making them attractive as an allogeneic "of-the-shelf" cell product, suitable for the AMI setting. The available evidence concerning umbilical cord-derived MSCs in AMI is reviewed, focusing on large animal pre-clinical studies and early human trials. Molecular and cellular mechanisms as well as current limitations and possible translational solutions are also discussed.
Collapse
Affiliation(s)
- Luís Raposo
- Cardiology Department, Santa Cruz Hospital, West Lisbon Hospital Center, Lisbon, Portugal; Hospital da Luz Lisboa, Luz Saúde, Lisbon, Portugal; Nova Medical School, Lisbon, Portugal.
| | - André P Lourenço
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diana S Nascimento
- Institute for Research and Innovation in Health, University of Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Portugal; Instituto Nacional de Engenharia Biomédica, University of Porto, Portugal
| | - Rui Cerqueira
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Nuno Cardim
- Hospital da Luz Lisboa, Luz Saúde, Lisbon, Portugal; Nova Medical School, Lisbon, Portugal
| | - Adelino Leite-Moreira
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
17
|
Barros I, Marcelo A, Silva TP, Barata J, Rufino-Ramos D, Pereira de Almeida L, Miranda CO. Mesenchymal Stromal Cells' Therapy for Polyglutamine Disorders: Where Do We Stand and Where Should We Go? Front Cell Neurosci 2020; 14:584277. [PMID: 33132851 PMCID: PMC7573388 DOI: 10.3389/fncel.2020.584277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022] Open
Abstract
Polyglutamine (polyQ) diseases are a group of inherited neurodegenerative disorders caused by the expansion of the cytosine-adenine-guanine (CAG) repeat. This mutation encodes extended glutamine (Q) tract in the disease protein, resulting in the alteration of its conformation/physiological role and in the formation of toxic fragments/aggregates of the protein. This group of heterogeneous disorders shares common molecular mechanisms, which opens the possibility to develop a pan therapeutic approach. Vast efforts have been made to develop strategies to alleviate disease symptoms. Nonetheless, there is still no therapy that can cure or effectively delay disease progression of any of these disorders. Mesenchymal stromal cells (MSC) are promising tools for the treatment of polyQ disorders, promoting protection, tissue regeneration, and/or modulation of the immune system in animal models. Accordingly, data collected from clinical trials have so far demonstrated that transplantation of MSC is safe and delays the progression of some polyQ disorders for some time. However, to achieve sustained phenotypic amelioration in clinics, several treatments may be necessary. Therefore, efforts to develop new strategies to improve MSC's therapeutic outcomes have been emerging. In this review article, we discuss the current treatments and strategies used to reduce polyQ symptoms and major pre-clinical and clinical achievements obtained with MSC transplantation as well as remaining flaws that need to be overcome. The requirement to cross the blood-brain-barrier (BBB), together with a short rate of cell engraftment in the lesioned area and low survival of MSC in a pathophysiological context upon transplantation may contribute to the transient therapeutic effects. We also review methods like pre-conditioning or genetic engineering of MSC that can be used to increase MSC survival in vivo, cellular-free approaches-i.e., MSC-conditioned medium (CM) or MSC-derived extracellular vesicles (EVs) as a way of possibly replacing the use of MSC and methods required to standardize the potential of MSC/MSC-derived products. These are fundamental questions that need to be addressed to obtain maximum MSC performance in polyQ diseases and therefore increase clinical benefits.
Collapse
Affiliation(s)
- Inês Barros
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,III-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Adriana Marcelo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Teresa P Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - João Barata
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - David Rufino-Ramos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Viravector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal
| | - Catarina O Miranda
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,III-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
18
|
Chinnadurai R, Forsberg MH, Kink JA, Hematti P, Capitini CM. Use of MSCs and MSC-educated macrophages to mitigate hematopoietic acute radiation syndrome. CURRENT STEM CELL REPORTS 2020; 6:77-85. [PMID: 32944493 DOI: 10.1007/s40778-020-00176-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review Innovative and minimally toxic treatment approaches are sorely needed for the prevention and treatment of hematopoietic acute radiation syndrome (H-ARS). Cell therapies have been increasingly studied for their potential use as countermeasures for accidental and intentional ionizing radiation exposures which can lead to fatal ARS. Mesenchymal stem/stromal cells (MSCs) are a cell therapy that have shown promising results in preclinical studies of ARS, and are being developed in clinical trials specifically for H-ARS. MSCs, MSC-educated macrophages (MEMs) and MSC-exosome educated macrophages (EEMs) all have the potential to be used as adoptive cell therapies for H-ARS. Here we review how MSCs have been reported to mitigate inflammation from radiation injury while also stimulating hematopoiesis during ARS. Recent findings We discuss emerging work with immune cell subsets educated by MSCs, including MEMs and EEMs, in promoting hematopoiesis in xenogeneic models of ARS. We also discuss the first placental-derived MSC product to enter phase I trials, PLX-R18, and the challenges faced by bringing MSC and other cell therapies into the clinic for treating ARS. Summary Although MSCs, MEMs and EEMs are potential cell therapy candidates in promoting hematopoietic HRS, challenges persist in translational clinical development of these products to the clinic. Whether any of these cellular therapies will be sufficient as stand-alone therapies to mitigate H-ARS or if they will be a bridging therapy that insures survival until a curative allogeneic hematopoietic stem cell transplant can be performed are the key questions that will have to be answered.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA
| | - Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - John A Kink
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
19
|
Allen A, Vaninov N, Li M, Nguyen S, Singh M, Igo P, Tilles AW, O'Rourke B, Miller BLK, Parekkadan B, Barcia RN. Mesenchymal Stromal Cell Bioreactor for Ex Vivo Reprogramming of Human Immune Cells. Sci Rep 2020; 10:10142. [PMID: 32576889 PMCID: PMC7311545 DOI: 10.1038/s41598-020-67039-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/22/2020] [Indexed: 11/20/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (MSCs) have been studied for decades as potent immunomodulators. Clinically, they have shown some promise but with limited success. Here, we report the ability of a scalable hollow fiber bioreactor to effectively maintain ideal MSC function as a single population while also being able to impart an immunoregulatory effect when cultured in tandem with an inflamed lymphocyte population. MSCs were seeded on the extraluminal side of hollow fibers within a bioreactor where they indirectly interact with immune cells flowing within the lumen of the fibers. MSCs showed a stable and predictable metabolite and secreted factor profile during several days of perfusion culture. Exposure of bioreactor-seeded MSCs to inflammatory stimuli reproducibly switched MSC secreted factor profiles and altered microvesicle composition. Furthermore, circulating, activated human peripheral blood mononuclear cells (PBMCs) were suppressed by MSC bioreactor culture confirmed by a durable change in their immunophenotype and function. This platform was useful to study a model of immobilized MSCs and circulating immune cells and showed that monocytes play an important role in MSC driven immunomodulation. This coculture technology can have broad implications for use in studying MSC-immune interactions under flow conditions as well as in the generation of ex vivo derived immune cellular therapeutics.
Collapse
Affiliation(s)
- Ashley Allen
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
| | | | - Matthew Li
- Department of Surgery, Center for Surgery, Innovation, and Bioengineering, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, 02114, USA
| | - Sunny Nguyen
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
| | - Maneet Singh
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
| | - Peter Igo
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
| | - Arno W Tilles
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
| | - Brian O'Rourke
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
| | | | - Biju Parekkadan
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA
- Department of Surgery, Center for Surgery, Innovation, and Bioengineering, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, 02138, USA
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, 08854, USA
| | - Rita N Barcia
- Sentien Biotechnologies, Inc., Lexington, MA, 02421, USA.
| |
Collapse
|
20
|
Laroye C, Boufenzer A, Jolly L, Cunat L, Alauzet C, Merlin JL, Yguel C, Bensoussan D, Reppel L, Gibot S. Bone marrow vs Wharton's jelly mesenchymal stem cells in experimental sepsis: a comparative study. Stem Cell Res Ther 2019; 10:192. [PMID: 31248453 PMCID: PMC6598309 DOI: 10.1186/s13287-019-1295-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/23/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The use of mesenchymal stem cells (MSCs) is being extensively studied in clinical trials in the setting of various diseases including diabetes, stroke, and progressive multiple sclerosis. The unique immunomodulatory properties of MSCs also point them as a possible therapeutic tool during sepsis and septic shock, a devastating syndrome associated with 30-35% mortality. However, MSCs are not equal regarding their activity, depending on their tissue origin. Here, we aimed at comparing the in vivo properties of MSCs according to their tissue source (bone marrow (BM) versus Wharton's jelly (WJ)) in a murine cecal ligation and puncture (CLP) model of sepsis that mimics a human peritonitis. We hypothesized that MSC properties may vary depending on their tissue source in the setting of sepsis. METHODS CLP, adult, male, C57BL/6 mice were randomized in 3 groups receiving respectively 0.25 × 106 BM-MSCs, 0.25 × 106 WJ-MSCs, or 150 μL phosphate-buffered saline (PBS) intravenously 24 h after the CLP procedure. RESULTS We observed that both types of MSCs regulated leukocyte trafficking and reduced organ dysfunction, while only WJ-MSCs were able to improve bacterial clearance and survival. CONCLUSION This study highlights the importance to determine the most appropriate source of MSCs for a given therapeutic indication and suggests a better profile for WJ-MSCs during sepsis.
Collapse
Affiliation(s)
- Caroline Laroye
- CHRU de Nancy, Unité de Thérapie Cellulaire et banque de Tissus, Allée du Morvan, 54500 Vandoeuvre-lès-Nancy, France
- INSERM UMRS-1116, Vandoeuvre-lès-Nancy, France
- CNRS, UMR 7365, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
| | | | - Lucie Jolly
- INSERM UMRS-1116, Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
- INOTREM, 54500 Vandoeuvre-lès-Nancy, France
| | - Lisiane Cunat
- Université de Lorraine, 54000 Nancy, France
- EA 7300 Stress Immunité Pathogènes, 54500 vandoeuvre-lès-Nancy, France
| | - Corentine Alauzet
- Université de Lorraine, 54000 Nancy, France
- EA 7300 Stress Immunité Pathogènes, 54500 vandoeuvre-lès-Nancy, France
| | - Jean-Louis Merlin
- Université de Lorraine, 54000 Nancy, France
- Service de Biopathologie - Unité de Biologie des Tumeurs, Institut de Cancérologie de Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Clémence Yguel
- CHRU de Nancy, laboratoire anatomie et cytologie pathologiques, 54500 Vandoeuvre-lès-Nancy, France
| | - Danièle Bensoussan
- CHRU de Nancy, Unité de Thérapie Cellulaire et banque de Tissus, Allée du Morvan, 54500 Vandoeuvre-lès-Nancy, France
- CNRS, UMR 7365, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
| | - Loïc Reppel
- CHRU de Nancy, Unité de Thérapie Cellulaire et banque de Tissus, Allée du Morvan, 54500 Vandoeuvre-lès-Nancy, France
- CNRS, UMR 7365, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
| | - Sébastien Gibot
- INSERM UMRS-1116, Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
- CHRU de Nancy, Service de Réanimation Médicale, Hôpital Central, 54000 Nancy, France
| |
Collapse
|
21
|
Bremm M, Pfeffermann LM, Cappel C, Katzki V, Erben S, Betz S, Quaiser A, Merker M, Bonig H, Schmidt M, Klingebiel T, Bader P, Huenecke S, Rettinger E. Improving Clinical Manufacturing of IL-15 Activated Cytokine-Induced Killer (CIK) Cells. Front Immunol 2019; 10:1218. [PMID: 31214182 PMCID: PMC6554420 DOI: 10.3389/fimmu.2019.01218] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are an immunotherapeutic approach to combat relapse following allogeneic hematopoietic stem cell transplantation (HSCT) in acute leukemia or myelodysplastic syndrome (MDS) patients. Prompt and sequential administration of escalating cell doses improves the efficacy of CIK cell therapy without exacerbating graft vs. host disease (GVHD). This study addresses manufacturing-related issues and aimed to develop a time-, personal- and cost-saving good manufacturing process (GMP)-compliant protocol for the generation of ready-for-use therapeutic CIK cell doses starting from one unstimulated donor-derived peripheral blood (PB) or leukocytapheresis (LP) products. Culture medium with or without the addition of either AB serum, fresh frozen plasma (FFP) or platelet lysate (PL) was used for culture. Fresh and cryopreserved CIK cells were compared regarding expansion rate, viability, phenotype, and ability to inhibit leukemia growth. Cell numbers increased by a median factor of 10-fold in the presence of FFP, PL, or AB serum, whereas cultivation in FFP/PL-free or AB serum-free medium failed to promote adequate CIK cell proliferation (p < 0.01) needed to provide clinical doses of 1 × 106 T cells/kG, 5 × 106 T cells/kG, 1 × 107 T cells/kG, and 1 × 108 T cells/kG recipient body weight. CIK cells consisting of T cells, T- natural killer (T-NK) cells and a minor fraction of NK cells were not significantly modified by different medium supplements. Moreover, neither cytotoxic potential against leukemic THP-1 cells nor cell activation shown by CD25 expression were significantly influenced. Moreover, overnight and long-term cryopreservation had no significant effect on the composition of CIK cells, their phenotype or cytotoxic potential. A viability of almost 93% (range: 89–96) and 89.3% (range: 84–94) was obtained after freeze-thawing procedure and long-term storage, respectively, whereas viability was 96% (range: 90-97) in fresh CIK cells. Altogether, GMP-complaint CIK cell generation from an unstimulated donor-derived PB or LP products was feasible. Introducing FFP, which is easily accessible, into CIK cell cultures was time- and cost-saving without loss of viability and potency in a 10-12 day batch culture. The feasibility of cryopreservation enabled storage and delivery of sequential highly effective ready-for-use CIK cell doses and therefore reduced the number of manufacturing cycles.
Collapse
Affiliation(s)
- Melanie Bremm
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | | | - Claudia Cappel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Verena Katzki
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Stephanie Erben
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Sibille Betz
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Andrea Quaiser
- Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Michael Merker
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Halvard Bonig
- Division for Translational Development of Cellular Therapeutics, Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Michael Schmidt
- Division for Translational Development of Cellular Therapeutics, Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Thomas Klingebiel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Peter Bader
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Sabine Huenecke
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Eva Rettinger
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| |
Collapse
|
22
|
Crain SK, Robinson SR, Thane KE, Davis AM, Meola DM, Barton BA, Yang VK, Hoffman AM. Extracellular Vesicles from Wharton's Jelly Mesenchymal Stem Cells Suppress CD4 Expressing T Cells Through Transforming Growth Factor Beta and Adenosine Signaling in a Canine Model. Stem Cells Dev 2019; 28:212-226. [PMID: 30412034 DOI: 10.1089/scd.2018.0097] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are widely investigated as potential therapeutic agents due to their potent immunomodulatory capacity. Although specific mechanisms by which MSC acts on immune cells are emerging, many questions remain, including the potential of extracellular vesicles (EVs) to mediate biological activities. Canine MSCs are of interest for both veterinary and comparative models of disease and have been shown to suppress CD4pos T cell proliferation. The aim of this study was to determine whether EV isolated from canine Wharton's jelly-derived MSC (WJ-MSC EV) suppresses CD4pos T cell proliferation using biochemical mechanisms previously ascribed to soluble mediators [transforming growth factor beta (TGF-β) and adenosine]. WJ-MSC EV exhibited mode of 125 nm diameter, low buoyant density (1.1 g/mL), and expression of EV proteins Alix and TSG101. Functionally, EVs inhibited CD4pos T cell proliferation in a dose-dependent manner, which was absent in EV-depleted samples and EVs from non-MSC fibroblasts. EV suppression of CD4pos T cell proliferation was inhibited by a TGF-βRI antagonist, neutralizing antibodies to TGF-β, or A2A adenosine receptor blockade. TGF-β was present on EVs as latent complexes most likely tethered to EV membrane by betaglycan. These data demonstrate that canine WJ-MSC EV utilizes TGF-β and adenosine signaling to suppress proliferation of CD4pos T cell and will enable further investigation into mechanisms of immune cell modulation, as well as refinement of WJ-MSC and their EVs for therapeutic application.
Collapse
Affiliation(s)
- Sarah K Crain
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Sally R Robinson
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Kristen E Thane
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Airiel M Davis
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Dawn M Meola
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Bruce A Barton
- 2 Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Vicky K Yang
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Andrew M Hoffman
- 1 Regenerative Medicine Laboratory, Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| |
Collapse
|
23
|
Chaudhury S, Saqibuddin J, Birkett R, Falcon-Girard K, Kraus M, Ernst LM, Grobman W, Mestan KK. Variations in Umbilical Cord Hematopoietic and Mesenchymal Stem Cells With Bronchopulmonary Dysplasia. Front Pediatr 2019; 7:475. [PMID: 31799226 PMCID: PMC6867971 DOI: 10.3389/fped.2019.00475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022] Open
Abstract
Objective: To test the hypothesis that umbilical cord blood-derived CD34+ hematopoietic stem cells (HPSC), cord tissue-derived CD90+ and CD105+ mesenchymal stem cells (MSC) vary with bronchopulmonary dysplasia (BPD). Methods: We conducted a prospective longitudinal study at a large birth center (Prentice Women's Hospital in Chicago, IL). Premature infants (N = 200) were enrolled in 2:1:1 ratio based on gestational age (GA): mildly preterm (31-32 weeks), moderately preterm (29-30 weeks), and extremely preterm (23-28 weeks). Cord blood (CB) and cord tissues (CT) were collected at birth using commercial banking kits, and analyzed for collection blood volume, tissue mass, CD34+, CD90+, CD105+ counts, and concentrations. Multiplex immunoassay was used to measure 12 cytokines and growth factors in CB plasma of 74 patients. BPD severity was defined according to NIH consensus definitions. Univariate and multivariate regression models were used to identify perinatal covariates and assess associations between stem cell concentrations, cytokines, and BPD outcomes. Results: Of 200 patients enrolled (mean GA = 30 ± 2 weeks), 30 developed mild, 24 moderate, and 19 severe BPD. Concentrations of HPSC and MSC, as measured by %CD34+, %CD90+, and %CD105+ of total cells, increased with degree of prematurity. Collection parameters varied with GA, birth weight (BW), gender, prolonged rupture of membranes, mode of delivery, chorioamnionitis, and multiple gestation. Moderate-severe BPD or death was increased with lower GA, BW, Apgar scores, and documented delayed cord clamping. %CD34+ and %CD90+ were increased with BPD and directly correlated with BPD severity. Severe BPD was positively associated with %CD34+ (beta-coefficient = 0.9; 95% CI = 0.4-1.5; P < 0.01) and %CD90+ (beta-coefficient = 0.4; 95% CI = 0.2-0.6; P < 0.001) after adjustment for covariates. CB plasma granulocyte-colony stimulating factor (G-CSF) was inversely associated with %CD90+, and decreased with BPD. Below median G-CSF combined with elevated %CD90+ predicted BPD (positive predictive value = 100%). Conclusions: CB and CT collections yielded high concentrations of HPSCs and MSCs in BPD infants, accompanied by low circulating G-CSF. These variations suggest possible mechanisms by which stem cell differentiation and function predict BPD.
Collapse
Affiliation(s)
- Sonali Chaudhury
- Division of Hematology/Stem Cell Transplant, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Juanita Saqibuddin
- Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Robert Birkett
- Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | | | - Morey Kraus
- ViaCord LLC, A Perkin Elmer Company, Cambridge, MA, United States
| | - Linda M Ernst
- Department of Pathology, NorthShore University, Evanston, IL, United States
| | - William Grobman
- Department of Obstetrics & Gynecology and Maternal Fetal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Karen K Mestan
- Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
24
|
Laroye C, Lemarié J, Boufenzer A, Labroca P, Cunat L, Alauzet C, Groubatch F, Cailac C, Jolly L, Bensoussan D, Reppel L, Gibot S. Clinical-grade mesenchymal stem cells derived from umbilical cord improve septic shock in pigs. Intensive Care Med Exp 2018; 6:24. [PMID: 30091119 PMCID: PMC6082751 DOI: 10.1186/s40635-018-0194-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/30/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Septic shock is the leading cause of death in intensive care units. The pathophysiological complexity of this syndrome contributes to an absence of specific treatment. Several preclinical studies in murine models of septic shock have shown improvements to organ injury and survival after administration of mesenchymal stem cells (MSCs). To better mimic a clinical approach in humans, we investigated the impact of randomized controlled double-blind administration of clinical-grade umbilical cord-derived MSCs to a relevant pig model of septic shock. METHODS Septic shock was induced by fecal peritonitis in 12 male domestic pigs. Animals were resuscitated by an experienced intensivist including fluid administration and vasopressors. Four hours after the induction of peritonitis, pigs were randomized to receive intravenous injection of thawed umbilical cord-derived MSCs (UCMSC) (1 × 106 UCMSCs/kg diluted in 75 mL hydroxyethyl starch (HES), (n = 6) or placebo (HES alone, n = 6). Researchers were double-blinded to the treatment administered. Hemodynamic parameters were continuously recorded. Gas exchange, acid-base status, organ function, and plasma cytokine concentrations were assessed at regular intervals until 24 h after the onset of peritonitis when animals were sacrificed under anesthesia. RESULTS Peritonitis induced profound hypotension, hyperlactatemia, and multiple organ failure. These disorders were significantly attenuated when animals were treated with UCMSCs. In particular, cardiovascular failure was attenuated, as attested by a better mean arterial pressure and reduced lactatemia, despite lower norepinephrine requirements. As such, UCMSCs improved survival in this very severe model (60% survival vs. 0% at 24 h). CONCLUSION UCMSCs administration is beneficial in this pig model of polymicrobial septic shock.
Collapse
Affiliation(s)
- Caroline Laroye
- CHRU de Nancy, Unité de Thérapie Cellulaire et banque de tissus, 54500 Vandoeuvre-lès-Nancy, France
- INSERM, U1116, 54500 Vandoeuvre-lès-Nancy, France
- CNRS, UMR 7365, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
| | - Jérémie Lemarié
- INSERM, U1116, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
- CHRU de Nancy, Service de Réanimation Médicale, Hôpital Central, 54000 Nancy, France
| | | | - Pierre Labroca
- CHRU de Nancy, Service de Réanimation Médicale, Hôpital Central, 54000 Nancy, France
| | - Lisiane Cunat
- Université de Lorraine, 54000 Nancy, France
- EA 7300 Stress Immunité Pathogènes, 54500 Vandoeuvre-lès-Nancy, France
| | - Corentine Alauzet
- Université de Lorraine, 54000 Nancy, France
- EA 7300 Stress Immunité Pathogènes, 54500 Vandoeuvre-lès-Nancy, France
| | - Frédérique Groubatch
- Université de Lorraine, 54000 Nancy, France
- Ecole de chirurgie, 54500 Vandoeuvre-lès-Nancy, France
| | - Clémence Cailac
- CHRU de Nancy, laboratoire anatomie et cytologie pathologiques, 54000 Nancy, France
| | - Lucie Jolly
- INSERM, U1116, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
- INOTREM, 54500 Vandoeuvre-lès-Nancy, France
| | - Danièle Bensoussan
- CHRU de Nancy, Unité de Thérapie Cellulaire et banque de tissus, 54500 Vandoeuvre-lès-Nancy, France
- CNRS, UMR 7365, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
| | - Loïc Reppel
- CHRU de Nancy, Unité de Thérapie Cellulaire et banque de tissus, 54500 Vandoeuvre-lès-Nancy, France
- CNRS, UMR 7365, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
| | - Sébastien Gibot
- INSERM, U1116, 54500 Vandoeuvre-lès-Nancy, France
- Université de Lorraine, 54000 Nancy, France
- CHRU de Nancy, Service de Réanimation Médicale, Hôpital Central, 54000 Nancy, France
| |
Collapse
|
25
|
Amărandi RM, Becheru DF, Vlăsceanu GM, Ioniță M, Burns JS. Advantages of Graphene Biosensors for Human Stem Cell Therapy Potency Assays. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1676851. [PMID: 30003089 PMCID: PMC5996421 DOI: 10.1155/2018/1676851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/22/2018] [Indexed: 12/11/2022]
Abstract
Regenerative medicine is challenged by the need to conform to rigorous guidelines for establishing safe and effective development and translation of stem cell-based therapies. Counteracting widespread concerns regarding unproven cell therapies, stringent cell-based assays seek not only to avoid harm but also to enhance quality and efficacy. Potency indicates that the cells are functionally fit for purpose before they are administered to the patient. It is a paramount quantitative critical quality attribute serving as a decisive release criterion. Given a broad range of stem cell types and therapeutic contexts the potency assay often comprises one of the most demanding hurdles for release of a cell therapy medicinal product. With need for improved biomarker assessment and expedited measurement, recent advances in graphene-based biosensors suggest that they are poised to be valuable platforms for accelerating potency assay development. Among several potential advantages, they offer versatility for sensitive measurement of a broad range of potential biomarker types, cell biocompatibility for direct measurement, and small sample sufficiency, plus ease of use and point-of-care applicability.
Collapse
Affiliation(s)
- Roxana-Maria Amărandi
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - Diana F. Becheru
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - George M. Vlăsceanu
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - Mariana Ioniță
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
- Advanced Polymer Materials Group, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - Jorge S. Burns
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
- Department of Medical and Surgical Sciences of Children and Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
26
|
The Effect of Pulsatile Flow on bMSC-Derived Endothelial-Like Cells in a Small-Sized Artificial Vessel Made by 3-Dimensional Bioprinting. Stem Cells Int 2018; 2018:7823830. [PMID: 29765422 PMCID: PMC5932426 DOI: 10.1155/2018/7823830] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/25/2017] [Accepted: 01/15/2018] [Indexed: 01/14/2023] Open
Abstract
Replacement of small-sized vessels is still challenging. This study is aimed at investigating the possibility of small-sized artificial vessels made by 3-dimensional bioprinting and the effect of pulsatile flow on bMSC-derived endothelial-like cells. Cells were harvested from rabbit bone marrow and primary cultured with or without growth factors. Endothelial differentiation was confirmed by the Matrigel tube formation assay, Western blot, and qRT-PCR. In addition, embedment of endothelial-like cells in an artificial vessel was made by 3-dimensional bioprinting, and the pulsatile flow was performed. For pumped and nonpumped groups, qRT-PCR was performed on CD31 and VE-cadherin gene expression. Endothelial-like cells showed increased gene expression of CD31 and VE-cadherin, and tube formation is observed at each week. Endothelial-like cells grow well in a small-sized artificial vessel made by 3-dimensional bioprinting and even express higher endothelial cell markers when they undergo pulsatile flow condition. Moreover, the pulsatile flow condition gives a positive effect for cell observation not only on the sodium alginate hydrogel layer but also on the luminal surface of the artificial vessel wall. We have developed an artificial vessel, which is a mixture of cells and carriers using a 3-dimensional bioprinting method, and applied pulsatile flow using a peristaltic pump, and we also demonstrated cell growth and differentiation into endothelial cells. This study suggests guidelines regarding a small-sized artificial vessel in the field of tissue engineering.
Collapse
|
27
|
Chinnadurai R, Rajan D, Qayed M, Arafat D, Garcia M, Liu Y, Kugathasan S, Anderson LJ, Gibson G, Galipeau J. Potency Analysis of Mesenchymal Stromal Cells Using a Combinatorial Assay Matrix Approach. Cell Rep 2018; 22:2504-2517. [PMID: 29490284 PMCID: PMC5855117 DOI: 10.1016/j.celrep.2018.02.013] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 09/11/2017] [Accepted: 02/02/2018] [Indexed: 02/07/2023] Open
Abstract
Assays that can characterize MSC immune potency need to be identified for use in advanced clinical trials. MSCs possess a number of putative regenerative and immunomodulatory properties, and an assay matrix approach may best capture involved effector pathways. We have tested two assay systems to measure the potency of MSCs derived from human subjects: MSC secretome analysis and a quantitative RNA-based array for genes specific to immunomodulatory and homing properties of MSCs. Secretome analysis identified a unique cytokine signature that is upregulated by MSCs or downregulated in responder PBMCs and correlated with T cell suppression. Use of interferon-γ as a surrogate for the action of activated PBMCs on MSCs served as an alternative for the use of human PBMCs as responder cells in a potency assay. Our approach and results define and simplify the multifunctional or matrix responses of MSCs and may serve as a platform for robust potency analysis.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Devi Rajan
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Muna Qayed
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Dalia Arafat
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | - Yifei Liu
- Department of Statistics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Subra Kugathasan
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Larry J Anderson
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Greg Gibson
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin - Madison, Madison, WI 53705, USA.
| |
Collapse
|
28
|
Laroye C, Gibot S, Reppel L, Bensoussan D. Concise Review: Mesenchymal Stromal/Stem Cells: A New Treatment for Sepsis and Septic Shock? Stem Cells 2017; 35:2331-2339. [PMID: 28856759 DOI: 10.1002/stem.2695] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/17/2022]
Abstract
Sepsis and septic shock are the leading cause of admission and mortality in non-coronary intensive care units. Currently, however, no specific treatments are available for this syndrome. Due to the failure of conventional treatments in recent years, research is focusing on innovative therapeutic agents, including cell therapy. One particular type of cell, mesenchymal stromal/stem cells (MSCs), has raised hopes for the treatment of sepsis. Indeed, their immunomodulatory properties, antimicrobial activity and capacity of protection against organ failure confer MSCs with a major advantage to treat the immune and inflammatory dysfunctions associated with sepsis and septic shock. After a brief description of the pathophysiology of sepsis and septic shock, the latest advances in the use of MSCs to treat sepsis will be presented. Stem Cells 2017;35:2331-2339.
Collapse
Affiliation(s)
- Caroline Laroye
- Unité de Thérapie Cellulaire et banque de Tissus, CHRU de Nancy, Vandœuvre-lès-Nancy, France.,INSERM, Vandœuvre-lès-Nancy, France.,UMR 7365 CNRS, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| | - Sébastien Gibot
- INSERM, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France.,CHRU Nancy, Service de Réanimation Médicale, Hôpital Central, Nancy, France
| | - Loïc Reppel
- Unité de Thérapie Cellulaire et banque de Tissus, CHRU de Nancy, Vandœuvre-lès-Nancy, France.,UMR 7365 CNRS, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| | - Danièle Bensoussan
- Unité de Thérapie Cellulaire et banque de Tissus, CHRU de Nancy, Vandœuvre-lès-Nancy, France.,UMR 7365 CNRS, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| |
Collapse
|
29
|
Avercenc-Léger L, Guerci P, Virion JM, Cauchois G, Hupont S, Rahouadj R, Magdalou J, Stoltz JF, Bensoussan D, Huselstein C, Reppel L. Umbilical cord-derived mesenchymal stromal cells: predictive obstetric factors for cell proliferation and chondrogenic differentiation. Stem Cell Res Ther 2017; 8:161. [PMID: 28676126 PMCID: PMC5497358 DOI: 10.1186/s13287-017-0609-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/09/2017] [Accepted: 06/14/2017] [Indexed: 01/20/2023] Open
Abstract
Background The umbilical cord is becoming a notable alternative to bone marrow (BM) as a source of mesenchymal stromal cells (MSC). Although age-dependent variations in BM-MSC are well described, less data are available for MSC isolated from Wharton’s jelly (WJ-MSC). We initiated a study to identify whether obstetric factors influenced MSC properties. We aimed to evaluate the correlation between a large number of obstetric factors collected during pregnancy and until peripartum (related to the mother, the labor and delivery, and the newborn) with WJ-MSC proliferation and chondrogenic differentiation parameters. Methods Correlations were made between 27 obstetric factors and 8 biological indicators including doubling time at passage (P)1 and P2, the percentage of proteoglycans and collagens, and the relative transcriptional expression of Sox-9, aggrecans, and total type 2 collagen (Coll2T). Results Amongst the obstetric factors considered, birth weight, the number of amenorrhea weeks, placental weight, normal pregnancy, and the absence of preeclampsia were identified as relevant factors for cell expansion, using multivariate linear regression analysis. Since all the above parameters are related to term, we concluded that WJ-MSC from healthy, full-term infants exhibit greater proliferation capacity. As for chondrogenesis, we also observed that obstetric factors influencing proliferation seemed beneficial, with no negative impact on MSC differentiation. Conclusions Awareness of obstetric factors influencing the proliferation and/or differentiation of WJ-MSC will make it possible to define criteria for collecting optimal umbilical cords with the aim of decreasing the variability of WJ-MSC batches produced for clinical use in cell and tissue engineering. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0609-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Léonore Avercenc-Léger
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France.,Université de Lorraine, 54000, Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Philippe Guerci
- CHRU de Nancy, Maternité Régionale Universitaire, Département d'Anesthésie-Réanimation, 54000, Nancy, France
| | - Jean-Marc Virion
- CHRU de Nancy, Epidémiologie et Evaluation Cliniques, 54500, Vandœuvre-lès-Nancy, France
| | - Ghislaine Cauchois
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France.,Université de Lorraine, 54000, Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Sébastien Hupont
- FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Rachid Rahouadj
- Université de Lorraine, 54000, Nancy, France.,UMR 7563 CNRS-Université de Lorraine, LEMTA, 54500, Vandœuvre-lès-Nancy, France
| | - Jacques Magdalou
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France.,Université de Lorraine, 54000, Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Jean-François Stoltz
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France.,CHRU de Nancy, Unité de Thérapie Cellulaire¸ Banque de Tissus, 54500, Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000, Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Danièle Bensoussan
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France.,CHRU de Nancy, Unité de Thérapie Cellulaire¸ Banque de Tissus, 54500, Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000, Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Céline Huselstein
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France.,Université de Lorraine, 54000, Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France
| | - Loïc Reppel
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus biologie-santé, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54500, Vandoeuvre-Les-nancy, France. .,CHRU de Nancy, Unité de Thérapie Cellulaire¸ Banque de Tissus, 54500, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, 54500, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
30
|
de Wolf C, van de Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human mesenchymal stromal cells used in immunotherapy. Cytotherapy 2017; 19:784-797. [PMID: 28457740 DOI: 10.1016/j.jcyt.2017.03.076] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/10/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells derived from various tissues that can differentiate into several cell types. MSCs are able to modulate the response of immune cells of the innate and adaptive immune system. Because of these multimodal properties, the potential use of MSCs for immunotherapies is currently explored in various clinical indications. Due to the diversity of potential MSC medicinal products at the level of cell source, manufacturing process and indication, distinct functionality tests may be needed to ensure the quality for each of the different products. In this review, we focus on in vitro potency assays proposed for characterization and release of different MSC medicinal products. We discuss the most used functional assays, as presented in scientific advices and literature, highlighting specific advantages and limitations of the various assays. Currently, the most proposed and accepted potency assay for release is based on in vitro inhibition of T cell proliferation or other functionalities. However, for some products, assays based on other MSC or responder cell properties may be more appropriate. In all cases, the biological relevance of the proposed assay for the intended clinical activity should be substantiated with appropriate product-specific (non-)clinical data. In case practical considerations prevent the use of the ideal potency assay at release, use of a surrogate marker or test could be considered if correlation with functionality has been demonstrated. Nevertheless, as the field of MSC immunology is evolving, improvements can be expected in relevant assays and consequently in guidance related to potency testing.
Collapse
|