1
|
Rimac V, Bojanić I, Škifić M, Dabelić S, Golubić Ćepulić B. Quality Assessment of Cryopreserved Peripheral Blood Stem Cell Products: Evaluation of Two Methods for Flow Cytometric Viability Testing. Int J Lab Hematol 2025; 47:93-100. [PMID: 39308095 DOI: 10.1111/ijlh.14374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 01/14/2025]
Abstract
INTRODUCTION The standard flow cytometry method for viability testing using 7-aminoactinomycin D (7-AAD) determines cells in necrosis and late apoptosis. The colony-forming unit (CFU) assay, which evaluates the proliferation ability of HSCs, is also used in graft quality assessment despite known deficiencies that make this assay impractical in routine clinical settings. The aim was to compare the effectiveness of the flow cytometry 7-AAD/annexin V method with the 7-AAD method in assessing the quality of HSCs in autologous and allogeneic peripheral blood stem cell (PBSC) products. METHODS Thirty autologous and 30 allogeneic fresh and thawed cryopreserved PBSC products were included in this study. The viability of HSCs was determined using the 7-AAD method and 7-AAD/annexin V method on a flow cytometer, while their clonogenic capacity was assessed by CFU assay. RESULTS There was an excellent correlation for CD34+ cell viability between the 7-AAD and the 7-AAD/annexin V method for fresh samples (Rs = 0.930, p < 0.001) and a good correlation for thawed PBSC samples (Rs = 0.739, p < 0.001). Excellent correlation was observed for post-thaw CD34+ cell recovery between the two methods for viability (Rs = 0.980, p < 0.001). Statistical analysis showed a weak correlation between CFU-GM recovery and CD34+ cell recovery, regardless of which viability testing method was used (7-AAD method p = 0.021, Rs = 0.298; 7-AAD/annexin V method p = 0.029, Rs = 0.282). CONCLUSIONS Results of this study showed that in the quality assessment of cryopreserved PBSC product viability, the 7-AAD/annexin V method had no added value compared to the 7-AAD method, which was suitable enough for routine quality control of cryopreserved autologous and allogeneic PBSC samples.
Collapse
Affiliation(s)
- Vladimira Rimac
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ines Bojanić
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Marijana Škifić
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Sanja Dabelić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Branka Golubić Ćepulić
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
2
|
Fang WH, Vangsness CT. Orthobiologic Products: Preservation Options for Orthopedic Research and Clinical Applications. J Clin Med 2024; 13:6577. [PMID: 39518716 PMCID: PMC11546119 DOI: 10.3390/jcm13216577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The biological products used in orthopedics include musculoskeletal allografts-such as bones, tendons, ligaments, and cartilage-as well as biological therapies. Musculoskeletal allografts support the body's healing process by utilizing preserved and sterilized donor tissue. These allografts are becoming increasingly common in surgical practice, allowing patients to avoid more invasive procedures and the risks associated with donor site morbidity. Bone grafting is one of the most frequently used procedures in orthopedics and traumatology. Biologic approaches aim to improve clinical outcomes by enhancing the body's natural healing capacity and reducing inflammation. They serve as an alternative to surgical interventions. While preliminary results from animal studies and small-scale clinical trials have been promising, the field of biologics still lacks robust clinical evidence supporting their efficacy. Biological therapies include PRP (platelet-rich plasma), mesenchymal stem cells (MSCs)/stromal cells/progenitor cells, bone marrow stem/stromal cells (BMSCs), adipose stem/stromal cells/progenitor cells (ASCs), cord blood (CB), and extracellular vesicles (EVs), including exosomes. The proper preservation and storage of these cellular therapies are essential for future use. Preservation techniques include cryopreservation, vitrification, lyophilization, and the use of cryoprotective agents (CPAs). The most commonly used CPA is DMSO (dimethyl sulfoxide). The highest success rates and post-thaw viability have been achieved by preserving PRP with a rate-controlled freezer using 6% DMSO and storing other cellular treatments using a rate-controlled freezer with 5% or 10% DMSO as the CPA. Extracellular vesicles (EVs) have shown the best results when lyophilized with 50 mM or 4% trehalose to prevent aggregation and stored at room temperature.
Collapse
Affiliation(s)
- William H. Fang
- Department of Orthopedic Surgery, Valley Health Systems, 620 Shadow Lane, Las Vegas, NV 89106, USA
| | - C. Thomas Vangsness
- Department of Orthopedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Cai Y, Prochazkova M, Kim YS, Jiang C, Ma J, Moses L, Martin K, Pham V, Zhang N, Highfill SL, Somerville RP, Stroncek DF, Jin P. Assessment and comparison of viability assays for cellular products. Cytotherapy 2024; 26:201-209. [PMID: 38085197 PMCID: PMC10872314 DOI: 10.1016/j.jcyt.2023.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND AIMS Accurate assessment of cell viability is crucial in cellular product manufacturing, yet selecting the appropriate viability assay presents challenges due to various factors. This study compares and evaluates different viability assays on fresh and cryopreserved cellular products, including peripheral blood stem cell (PBSC) and peripheral blood mononuclear cell (PBMC) apheresis products, purified PBMCs and cultured chimeric antigen receptor and T-cell receptor-engineered T-cell products. METHODS Viability assays, including manual Trypan Blue exclusion, flow cytometry-based assays using 7-aminoactinomycin D (7-AAD) or propidium iodide (PI) direct staining or cell surface marker staining in conjunction with 7-AAD, Cellometer (Nexcelom Bioscience LLC, Lawrence, MA, USA) Acridine Orange/PI staining and Vi-CELL BLU Cell Viability Analyzer (Beckman Coulter, Inc, Brea, CA, USA), were evaluated. A viability standard was established using live and dead cell mixtures to assess the accuracy of these assays. Furthermore, precision assessment was conducted to determine the reproducibility of the viability assays. Additionally, the viability of individual cell populations from cryopreserved PBSC and PBMC apheresis products was examined. RESULTS All methods provided accurate viability measurements and generated consistent and reproducible viability data. The assessed viability assays were demonstrated to be reliable alternatives when evaluating the viability of fresh cellular products. However, cryopreserved products exhibited variability among the tested assays. Additionally, analyzing the viability of each subset of the cryopreserved PBSC and PBMC apheresis products revealed that T cells and granulocytes were more susceptible to the freeze-thaw process, showing decreased viability. CONCLUSIONS The study demonstrates the importance of careful assay selection, validation and standardization, particularly for assessing the viability of cryopreserved products. Given the complexity of cellular products, choosing a fit-for-purpose viability assay is essential.
Collapse
Affiliation(s)
- Yihua Cai
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Michaela Prochazkova
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Yong-Soo Kim
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Chunjie Jiang
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Jinxia Ma
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Larry Moses
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Kathryn Martin
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Victoria Pham
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Nan Zhang
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Steven L Highfill
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Robert P Somerville
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - David F Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Ping Jin
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA.
| |
Collapse
|
4
|
Simard C, Fournier D, Trépanier P. Validation of a rapid potency assay for cord blood stem cells using phospho flow cytometry: The IL-3-pSTAT5 assay. Int J Lab Hematol 2023; 45:46-52. [PMID: 36196510 DOI: 10.1111/ijlh.13975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/14/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Public cord blood banks (CBBs) are required to measure cord blood units (CBUs) potency before their release, allowing for the identification of units that may be unsuitable for haematopoietic transplantation. We have developed a rapid flow cytometry assay based on the measurement of STAT-5 phosphorylation of CD34+ stem cells in response to IL-3 stimulation. METHOD To adapt the assay from a research setting to its implementation within our CBB regulated operations, we proceded with a full method validation and a correlation comparison of the IL-3-pSTAT5 assay results with the colony-forming unit assay (CFU) results. A total of 60 CBUs cryopreserved in vials were analysed by flow cytometry to determine the sensitivity, specificity, intra-assay precision, robustness, reproducibility, and inter-laboratory agreement of the assay. The CFU assay was also done on the same samples for comparison purposes. RESULTS The assay threshold was established at 50% CD34+CD45+pSTAT5+, which provides a 100% sensitivity and a 98.3% specificity. An average intra-assay CV of 7.3% was determined. All results met our qualitative results acceptance criteria regarding the inter-user and inter-laboratory agreements, IL-3 stimulation time, post-thaw incubation delay and staining time. The IL-3-pSTAT5 assay results correlated well with the total CFU determined using the CFU assay (r2 = 0.82, n = 56). CONCLUSION This study shows that our rapid flow cytometry assay can be successfully validated and that the potency data obtained display good sensitivity, specificity and robustness. These results demonstrate the feasibility of implementing this assay within CBB operations, as a validated potency assay.
Collapse
Affiliation(s)
- Carl Simard
- Héma-Québec, Medical Affairs and Innovation, Québec City, Québec, Canada
| | | | - Patrick Trépanier
- Héma-Québec, Medical Affairs and Innovation, Québec City, Québec, Canada
| |
Collapse
|
5
|
Burns JS. The Evolving Landscape of Potency Assays. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:165-189. [PMID: 37258790 DOI: 10.1007/978-3-031-30040-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
There is a "goldilocks" aspect to potency assays. On the one hand, a comprehensive evaluation of the cell product with detailed quantitative measurement of the critical quality attribute/s of the desired biological activity is required. On the other hand, the potency assay benefits from simplification and lean approaches that avoid unnecessary complication and enhance robustness, to provide a reproducible and scalable product. There is a need to balance insightful knowledge of complex biological healing processes with straightforward manufacture of an advanced therapeutic medicinal product (ATMP) that can be administered in a trustworthy cost-effective manner. While earlier chapters within this book have highlighted numerous challenges facing the potency assay conundrum, this chapter offers a forward-looking perspective regarding the many recent advances concerning acellular products, cryopreservation, induced MSC, cell priming, nanotechnology, 3D culture, regulatory guidelines and evolving institutional roles, that are likely to facilitate potency assay development in the future.
Collapse
Affiliation(s)
- Jorge S Burns
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
6
|
Li M, Morse B, Kassim S. Development and clinical translation considerations for the next wave of gene modified hematopoietic stem and progenitor cells therapies. Expert Opin Biol Ther 2022; 22:1177-1191. [PMID: 35833356 DOI: 10.1080/14712598.2022.2101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Consistent and reliable manufacture of gene modified hematopoietic stem and progenitor cell (HPSC) therapies will be of the utmost importance as they become more mainstream and address larger populations. Robust development campaigns will be needed to ensure that these products will be delivered to patients with the highest quality standards. AREAS COVERED Through publicly available manuscripts, press releases, and news articles - this review touches on aspects related to HSPC therapy, development, and manufacturing. EXPERT OPINION Recent advances in genome modification technology coupled with the longstanding clinical success of HSPCs warrants great optimism for the next generation of engineered HSPC-based therapies. Treatments for some diseases that have thus far been intractable now appear within reach. Reproducible manufacturing will be of critical importance in delivering these therapies but will be challenging due to the need for bespoke materials and methods in combination with the lack of off-the-shelf solutions. Continued progress in the field will manifest in the form of industrialization which currently requires attention and resources directed toward the custom reagents, a focus on closed and automated processes, and safer and more precise genome modification technologies that will enable broader, faster, and safer access to these life-changing therapies.
Collapse
Affiliation(s)
| | - Brent Morse
- Dark Horse Consulting Group, Walnut Creek, CA, USA
| | | |
Collapse
|
7
|
Welschinger L, Milton C, Zaunders G, Ashraf A. Effect of nucleated cell count and cryopreservation on engraftment post autologous stem cell transplant. Transfus Apher Sci 2022; 61:103495. [DOI: 10.1016/j.transci.2022.103495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022]
|
8
|
Simard C, Fournier D, Pineault N, Trépanier P. Rapid potency assessment of autologous peripheral blood stem cells by intracellular flow cytometry: the PBSC-IL-3-pSTAT5 assay. Cytotherapy 2022; 24:879-883. [DOI: 10.1016/j.jcyt.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 11/15/2022]
|
9
|
Makhani SS, Oza SP, Reich-Slotky R, Munshi PN, Biran N, Donato ML, Siegel DS, Vesole DH, Naam S, Rowley SD. Sustained Hematopoietic Engraftment Potential after Prolonged Storage of Cryopreserved Hematopoietic Stem Cells (HSC) Used in Salvage Autologous Stem Cell Transplantation: Prolonged Storage of HSCs Sustains Hematological Reconstitution Potential. Transplant Cell Ther 2022; 28:306.e1-306.e7. [PMID: 35248777 DOI: 10.1016/j.jtct.2022.02.023] [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: 01/16/2022] [Revised: 02/17/2022] [Accepted: 02/27/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Salvage autologous hematopoietic stem cell transplantation (HSCT) is an effective treatment for patients with relapsed multiple myeloma (MM). Peripheral blood stem cells (PBSCs), a source of hematopoietic stem cells (HSCs), are collected prior to first transplant and adequate quantities of PBSCs can be collected and stored potentially for years to support at least two transplants for eligible patients. To ensure the safety of salvage HSCT used in the treatment of patients in subsequent relapse, PBSCs must retain the potential to engraft even after several years of cryopreservation. Although PBSC viability has been extensively studied using in vitro techniques, few publications describe the most rigorous functional potency measure: of patients receiving a myeloablative conditioning regimen. This study describes a large single-institution experience evaluating the engraftment kinetics of PBSCs used in salvage transplantation after multiple years of storage in comparison to first transplantation of the same patients in the treatment of MM. STUDY DESIGN AND METHODS A retrospective chart review of patients with MM undergoing HSCT from 2000 to 2021, identified 89 patients who received salvage autologous PBSC stored > 1 year after first HSCT. PBSC were cryopreserved and stored in vapor-phase liquid nitrogen refrigerators at a temperature of ≤ -150°C. All patients received a PBSC product for both transplants from the same collection cycle. Differences in CD34+ cell doses and days to engraftment between the first and salvage transplant were tested using a paired 2-tailed t-test and Wilcoxon signed-rank test. Univariate and multivariable linear regressions were used to determine association between storage time and days to engraftment, adjusting for CD34+ cell dose and conditioning regimen in the multivariable model. RESULTS The median storage time between day of initial collection and salvage transplant was 5.4 years (range, 1.0 - 19.7). Engraftment kinetics demonstrated a sustained neutrophil engraftment (absolute neutrophil count (ANC) > 0.5 × 109 cells/L) at a median of 11 days after both the first and salvage transplant (ranges, 8 - 15 and 8 - 19 respectively, p<0.05). The median time to sustained platelet engraftment (> 20 × 109 cells/L without transfusion support) was 13.5 days after first HSCT and 14 days after salvage HSCT (ranges, 9 - 27 and 10 - 56 respectively, p = 0.616). After adjusting for CD34+ cell doses and conditioning regimens, there was no association between the duration of cryopreservation and days to neutrophil (r = 0.178, p = 0.130) or platelet (r = 0.244, p = 0.100) engraftments. CONCLUSION Engraftment kinetics of the salvage HSCT are comparable to the first HSCT even when products are stored in vapor-phase nitrogen refrigerators for a median time of 5.4 years. There is no association between storage duration and time to engraftment when controlling for CD34+ dose and conditioning regimens. Prolonged storage of cryopreserved HSC products is a safe practice for MM patients undergoing salvage autologous HSCT.
Collapse
Affiliation(s)
- Sarah S Makhani
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL
| | - Samir P Oza
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Ronit Reich-Slotky
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ.
| | - Pashna N Munshi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Noa Biran
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Michele L Donato
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - David S Siegel
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - David H Vesole
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Suzan Naam
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Scott D Rowley
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
10
|
Araújo AB, Salton GD, Angeli MH, Furlan JM, Schmalfuss T, Röhsig LM. Effects of cell concentration, time of fresh storage, and cryopreservation on peripheral blood stem cells: PBSC fresh storage and cryopreservation. Transfus Apher Sci 2021; 61:103298. [PMID: 34696981 DOI: 10.1016/j.transci.2021.103298] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/14/2021] [Accepted: 10/17/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Peripheral blood stem cells are widely used in autologous or allogeneic transplantation. The quality of the product directly impacts clinical outcomes, and the cell quality and/or functionality may be influenced by the storage conditions as time, temperature, total nucleated cells (TNC) concentration and cryopreservation requirement. OBJECTIVE To verify the effects of time, cell concentration, and cryopreservation/thawing in the viability and functionality of stem cells for transplantation. METHODS We evaluated TNC, CD45+ viable cells, CD34+ viable cells, and cell viability and functionality of 11 samples. Measurements were performed immediately and 24 h, 48 h, 72 h, and 96 h after sample collection at high and low TNC concentrations. The same parameters were also evaluated after cryopreservation and thawing of the samples. RESULT Duration of storage and TNC concentration exhibited a negative effect on cell quality (CD45+ viable cells, CD34+ viable cells and functionality). Moreover, the association of these parameters increased the negative effect on graft quality. Cryopreservation and thawing also negatively affected the collected sample regarding viable CD34+ cells (recovery 66.2 %), viable CD45+ cells (recovery 56.8 %), and 7-AAD viability. No significant losses in viable CD45+/CD34+ cells and functionality were observed in the first 24 h in both TNC conditions. CONCLUSION These results emphasize the importance to consider carefully the storage conditions until transplantation, measuring TNC/μL until 24 h after collection (diluting the product when TNC > 300 × 103/μL) and infusing fresh graft as soon as possible.
Collapse
Affiliation(s)
- Anelise B Araújo
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, RS, 90035-903, Brazil.
| | - Gabrielle D Salton
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, RS, 90035-903, Brazil.
| | - Melissa H Angeli
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, RS, 90035-903, Brazil.
| | - Juliana M Furlan
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, RS, 90035-903, Brazil.
| | - Tissiana Schmalfuss
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, RS, 90035-903, Brazil.
| | - Liane M Röhsig
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, RS, 90035-903, Brazil.
| |
Collapse
|
11
|
Měřička P, Janoušek L, Benda A, Lainková R, Sabó J, Dalecká M, Prokšová P, Salmay M, Špunda R, Pecha O, Jandová M, Gregor J, Štěrba L, Špaček M, Lindner J. Cell Viability Assessment Using Fluorescence Vital Dyes and Confocal Microscopy in Evaluating Freezing and Thawing Protocols Used in Cryopreservation of Allogeneic Venous Grafts. Int J Mol Sci 2021; 22:ijms221910653. [PMID: 34638994 PMCID: PMC8509073 DOI: 10.3390/ijms221910653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/29/2022] Open
Abstract
The authors present their contribution to the improvement of methods suitable for the detection of the freezing and thawing damage of cells of cryopreserved venous grafts used for lower limb revascularization procedures. They studied the post-thaw viability of cells of the wall of cryopreserved venous grafts (CVG) immediately after thawing and after 24 and 48 h culture at +37 °C in two groups of six CVG selected randomly for slow thawing in the refrigerator and rapid thawing in a water bath at +37 °C. The grafts were collected from multi-organ and tissue brain-dead donors, cryopreserved, and stored in a liquid nitrogen vapor phase for five years. The viability was assessed from tissue slices obtained by perpendicular and longitudinal cuts of the thawed graft samples using in situ staining with fluorescence vital dyes. The mean and median immediate post-thaw viability values above 70% were found in using both thawing protocols and both types of cutting. The statistically significant decline in viability after the 48-h culture was observed only when using the slow thawing protocol and perpendicular cutting. The possible explanation might be the “solution effect damage” during slow thawing, which caused a gentle reduction in the graft cellularity. The possible influence of this phenomenon on the immunogenicity of CVG should be the subject of further investigations.
Collapse
Affiliation(s)
- Pavel Měřička
- Tissue Bank, University Hospital, 500 05 Hradec Králové, Czech Republic; (P.M.); (M.J.); (J.G.); (L.Š.)
| | - Libor Janoušek
- Department of Transplantation Surgery, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic;
| | - Aleš Benda
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, 252 50 Prague, Czech Republic; (A.B.); (J.S.); (M.D.); (P.P.)
| | - Radka Lainková
- 2nd Department of Surgery–Department of Cardiovascular Surgery, 1st Medical Faculty, Charles University and General University Hospital, 128 08 Prague, Czech Republic; (R.L.); (M.S.); (R.Š.); (J.L.)
| | - Ján Sabó
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, 252 50 Prague, Czech Republic; (A.B.); (J.S.); (M.D.); (P.P.)
| | - Markéta Dalecká
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, 252 50 Prague, Czech Republic; (A.B.); (J.S.); (M.D.); (P.P.)
- Department of Cell Biology, Charles University, Viničná 7, 128 00 Prague, Czech Republic
| | - Petra Prokšová
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, 252 50 Prague, Czech Republic; (A.B.); (J.S.); (M.D.); (P.P.)
| | - Myroslav Salmay
- 2nd Department of Surgery–Department of Cardiovascular Surgery, 1st Medical Faculty, Charles University and General University Hospital, 128 08 Prague, Czech Republic; (R.L.); (M.S.); (R.Š.); (J.L.)
| | - Rudolf Špunda
- 2nd Department of Surgery–Department of Cardiovascular Surgery, 1st Medical Faculty, Charles University and General University Hospital, 128 08 Prague, Czech Republic; (R.L.); (M.S.); (R.Š.); (J.L.)
| | - Ondřej Pecha
- Technology Centre of the Czech Academy of Sciences, 160 00 Prague, Czech Republic;
| | - Miroslava Jandová
- Tissue Bank, University Hospital, 500 05 Hradec Králové, Czech Republic; (P.M.); (M.J.); (J.G.); (L.Š.)
- Department of Anatomy, Histology and Embryology Medical Faculty in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Jiří Gregor
- Tissue Bank, University Hospital, 500 05 Hradec Králové, Czech Republic; (P.M.); (M.J.); (J.G.); (L.Š.)
| | - Lubomír Štěrba
- Tissue Bank, University Hospital, 500 05 Hradec Králové, Czech Republic; (P.M.); (M.J.); (J.G.); (L.Š.)
| | - Miroslav Špaček
- 2nd Department of Surgery–Department of Cardiovascular Surgery, 1st Medical Faculty, Charles University and General University Hospital, 128 08 Prague, Czech Republic; (R.L.); (M.S.); (R.Š.); (J.L.)
- Correspondence:
| | - Jaroslav Lindner
- 2nd Department of Surgery–Department of Cardiovascular Surgery, 1st Medical Faculty, Charles University and General University Hospital, 128 08 Prague, Czech Republic; (R.L.); (M.S.); (R.Š.); (J.L.)
| |
Collapse
|
12
|
Jung SE, Ahn JS, Kim YH, Oh HJ, Kim BJ, Kim SU, Ryu BY. Autophagy modulation alleviates cryoinjury in murine spermatogonial stem cell cryopreservation. Andrology 2021; 10:340-353. [PMID: 34499811 DOI: 10.1111/andr.13105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 08/11/2021] [Accepted: 08/28/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Cryopreservation can expand the usefulness of spermatogonial stem cells (SSCs) in various fields. However, previous investigations that have attempted to modulate cryoinjury-induced mechanisms to increase cryoprotective efficiency have mainly focused on apoptosis and necrosis. OBJECTIVES This study aimed to establish an effective molecular-based cryoprotectant for SSC cryopreservation via autophagy modulation. MATERIALS AND METHODS To determine the efficacy of autophagy modulation, we assessed the recovery rate and relative proliferation rate and performed western blotting for the determination of autophagy flux, immunocytochemistry and real-time quantitative polymerase chain reaction (RT-qPCR) for SSC characterization, and spermatogonial transplantation for in vivo SSC functional activity. RESULTS The results showed that a basal level of autophagy caused a higher relative proliferation rate (pifithrin-μ 0.01 μM, 184.2 ± 11.2%; 3-methyladenine 0.01 μM, 175.3 ± 10.3%; pifithrin-μ 0.01 μM + 3-methyladenine 0.01 μM, P3, 224.6 ± 22.3%) than the DMSO control (100 ± 6.2%). All treatment groups exhibited normal characteristics, suggesting that these modulators could be used as effective cryoprotectants without changing the properties of the undifferentiated germ cells. According to the results of the in vivo spermatogonial transplantation assay, the colonies per total number of cultured SSCs was significantly higher in the pifithrin-μ 0.01 μM (1596.7 ± 172.5 colonies), 3-methyladenine 0.01 μM (1522.1 ± 179.2 colonies), and P3 (1727.5 ± 196.5 colonies) treatment groups than in the DMSO control (842.8 ± 110.08 colonies), which was comparable to that of the fresh control (1882.1 ± 132.1 colonies). DISCUSSION A basal level of autophagy is more essential for resilience in frozen SSCs after thawing, rather than the excessive activation or inhibition of autophagy. CONCLUSION A basal level of autophagy plays a critical role in the pro-survival response of frozen SSCs after thawing; herein, a new approach by which SSC cryoprotective efficiency can be improved was identified.
Collapse
Affiliation(s)
- Sang-Eun Jung
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Jin Seop Ahn
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Yong-Hee Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Hui-Jo Oh
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Bang-Jin Kim
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sun-Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk-do, Republic of Korea.,Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| |
Collapse
|
13
|
Lysak D, Brychtová M, Leba M, Čedíková M, Georgiev D, Jindra P, Vlas T, Holubova M. Long-Term Cryopreservation Does Not Affect Quality of Peripheral Blood Stem Cell Grafts: A Comparative Study of Native, Short-Term and Long-Term Cryopreserved Haematopoietic Stem Cells. Cell Transplant 2021; 30:9636897211036004. [PMID: 34328022 PMCID: PMC8327011 DOI: 10.1177/09636897211036004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cryopreserved haematopoietic progenitor cells are used to restore autologous haematopoiesis after high dose chemotherapy. Although the cells are routinely stored for a long period, concerns remain about the maximum storage time and the possible negative effect of storage on their potency. We evaluated the effect of cryopreservation on the quality of peripheral stem cell grafts stored for a short (3 months) and a long (10 years) period and we compared it to native products.The viability of CD34+ cells remained unaffected during storage, the apoptotic cells were represented up to 10% and did not differ between groups. The clonogenic activity measured by ATP production has decreased with the length of storage (ATP/cell 1.28 nM in native vs. 0.63 in long term stored products, P < 0.05). Only borderline changes without statistical significance were detected when examining mitochondrial and aldehyde dehydrogenase metabolic activity and intracellular pH, showing their good preservation during cell storage. Our experience demonstrates that cryostorage has no major negative effect on stem cell quality and potency, and therefore autologous stem cells can be stored safely for an extended period of at least 10 years. On the other hand, long term storage for 10 years and longer may lead to mild reduction of clonogenic capacity. When a sufficient dose of stem cells is infused, these changes will not have a clinical impact. However, in products stored beyond 10 years, especially when a low number of CD34+ cells is available, the quality of stem cell graft should be verified before infusion using the appropriate potency assays.
Collapse
Affiliation(s)
- Daniel Lysak
- Department of Haematology and Oncology, University Hospital Pilsen, Pilsen, Czech Republic
| | - Michaela Brychtová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Martin Leba
- New Technologies for the Information Society European Centre of Excellence, Faculty of Applied Sciences, University of West Bohemia, Pilsen, Czech Republic
| | - Miroslava Čedíková
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Daniel Georgiev
- New Technologies for the Information Society European Centre of Excellence, Faculty of Applied Sciences, University of West Bohemia, Pilsen, Czech Republic
| | - Pavel Jindra
- Department of Haematology and Oncology, University Hospital Pilsen, Pilsen, Czech Republic
| | - Tomáš Vlas
- Institute of Immunology and Allergology, University Hospital Pilsen, Pilsen, Czech Republic
| | - Monika Holubova
- Department of Haematology and Oncology, University Hospital Pilsen, Pilsen, Czech Republic.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| |
Collapse
|
14
|
Jung SE, Ahn JS, Kim YH, Kim SM, Um TG, Kim BJ, Ryu BY. Inhibition of Caspase-8 Activity Improves Freezing Efficiency of Male Germline Stem Cells in Mice. Biopreserv Biobank 2021; 19:493-502. [PMID: 33926212 DOI: 10.1089/bio.2021.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cryopreservation of male germline stem cells (GSCs) is an essential technique for their long-term preservation and utilization in various fields. However, the specific apoptosis pathways involved in cryoinjury during freezing remain unclear. Therefore, our study sought to identify the pathways involved in cryoinjury-induced apoptosis and thereby to improve freezing efficiency during GSC cryopreservation through the creation of a specific molecular-based cryoprotectant. The activities of caspase-8, caspase-9, caspase-3, and caspase-7 were assessed by Western blot analyses to determine the role of specific apoptosis pathways in GSC cryoinjury. Specifically, the role of a specific caspase was identified by recovery rate, relative proliferation rate, Annexin V/propidium iodide co-staining, and caspase activity using its inhibitor and activator. Moreover, the safety of the cryoprotectant was assessed by immunofluorescence and quantitative real-time polymerase chain reaction (qRT-PCR). Furthermore, the efficacy of the molecular-based cryoprotectant was assessed using frozen cells in the presence of dimethyl sulfoxide (DMSO) (control), trehalose, a caspase-8 inhibitor Z-IETD-FMK [ZIF], or a mixture of the aforementioned compounds, after which the changes in Src signaling were measured. Our results demonstrated that caspase-8 plays a major role in cryoinjury-induced apoptosis and therefore its inhibition improves freezing efficiency. Specifically, a significantly higher relative proliferation rate was observed in the Z-IETD-FMK 0.01 μM-treated cells than in the DMSO control (100% ± 6.2% vs. 189.8% ± 9.5%), with decreases in both early apoptosis (16.6% ± 2.2% vs. 7.5% ± 1.0%) and caspase-8 activity (1.0-fold vs. 0.4-fold). The relative proliferation rate was significantly higher in the cryoprotectant mixture (246.0% ± 12.2%) than other individual treatment groups (trehalose 200 mM, 189.8% ± 9.5%; Z-IETD-FMK 0.01 μM, 189.7% ± 2.2%) with no significant differences in Src signaling. Therefore, our findings provide novel insights into the development of freezing protocols to enhance GSC freezing efficiency, thereby facilitating the wider adoption of GSCs in the livestock industry and/or clinical trials.
Collapse
Affiliation(s)
- Sang-Eun Jung
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Jin Seop Ahn
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Yong-Hee Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Seok-Man Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Tea Gun Um
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Bang-Jin Kim
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| |
Collapse
|
15
|
Automated dry thawing of cryopreserved haematopoietic cells is not adversely influenced by cryostorage time, patient age or gender. PLoS One 2020; 15:e0240310. [PMID: 33104704 PMCID: PMC7588046 DOI: 10.1371/journal.pone.0240310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/23/2020] [Indexed: 01/01/2023] Open
Abstract
Cell therapies are becoming increasingly widely used, and their production and cryopreservation should take place under tightly controlled GMP conditions, with minimal batch-to-batch variation. One potential source of variation is in the thawing of cryopreserved samples, typically carried out in water baths. This study looks at an alternative, dry thawing, to minimise variability in the thawing of a cryopreserved cell therapy, and compares the cellular outcome on thaw. Factors such as storage time, patient age, and gender are considered in terms of cryopreservation and thawing outcomes. Cryopreserved leukapheresis samples from 41 donors, frozen by the same protocol and stored for up to 17 years, have been thawed using automated, water-free equipment and by conventional wet thawing using a water bath. Post-thaw viability, assessed by both trypan blue and flow cytometry, showed no significant differences between the techniques. Similarly, there was no negative effect of the duration of frozen storage, donor age at sample collection or donor gender on post-thaw viability using either thawing method. The implication of these results is that the cryopreservation protocol chosen initially remains robust and appropriate for use with a wide range of donors. The positive response of the samples to water-free thawing offers potential benefits for clinical situations by removing the subjective element inherent in water bath thawing and eliminating possible contamination issues.
Collapse
|
16
|
Trummer T, Fox R, Koç JR, de Lima M, Otegbeye F. Cryopreservation of hematopoietic cells using a pre-constituted, protein-free cryopreservative solution with 5% dimethyl sulfoxide. Cytotherapy 2020; 22:613-616. [PMID: 32600975 DOI: 10.1016/j.jcyt.2020.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND AIMS Adequate cryopreservation techniques are critical to ensure optimal recovery of functional progenitor cells in hematopoietic cell (HC) transplantation, minimize risk of contamination and prevent infusion-related adverse events (irAEs). In this article, we provide graft function and infusion safety results observed by decreasing the concentration of dimethyl sulfoxide (DMSO) in cryopreservative media and by minimizing processor-dependent formulation. METHODS Ten HC products, collected after standard mobilization of multiple myeloma patients, were cryopreserved with PRIME-XV FreezIS (FreezIS) and compared with products previously cryopreserved with media formulated in-house to achieve a final DMSO concentration of 10% (Std10) and 5% (Std5). At infusion, HCs were analyzed for recovery of CD34+ cells and viability; irAEs and time to engraftment of neutrophils and platelets were also monitored. RESULTS Median CD34+ cell recovery for HC cryopreserved with Std10, Std5 and FreezIS was 38%, 78% and 68%, respectively (P = 0.0002). There were less frequent irAEs with Std5 and FreezIS (10%) compared with Std10 (80%) (P ≤ 0.0001). Median time to neutrophil engraftment was comparable (11 days) for all three groups, while platelet engraftment occurred at a median of 20, 19 and 17 days, respectively (p-values not significant). CONCLUSIONS FreezIS, a Good Manufacturing Practice-grade, pre-constituted cryopreservative with low DMSO content, maintains functional viability of the HC product while reducing the incidence of irAEs compared with 10% DMSO solutions. The pre-constituted nature of this agent also decreases processor-dependent handling, hence decreasing the risk of variability and infection.
Collapse
Affiliation(s)
- Tabatha Trummer
- University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Robert Fox
- University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Jane Reese Koç
- University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Marcos de Lima
- University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | | |
Collapse
|
17
|
Marinelli Busilacchi E, Costantini A, Mancini G, Bencivenga R, Olivieri J, Battaglini G, Velletri L, Viola N, Butini L, Capelli D, Poloni A, Olivieri A. A novel method to evaluate prethawing viability of cryopreserved CD34+ hematopoietic stem cells for autologous transplantation. Transfusion 2020; 60:1529-1535. [PMID: 32488938 DOI: 10.1111/trf.15825] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cryopreservation of CD34+ hematopoietic stem cells (HSCs) is associated with variable loss of viability. Although postfreezing CD34+ cell viability can be assessed on the sampling tube (bag tail) directly connected to the main bag (mother bag), results often underestimate the actual viability observed when the mother bag is thawed and reinfused. We assessed a novel method to measure postfreezing CD34+ cell viability, based on small bag (minibag) samples; results were compared with those obtained on the corresponding mother bags and bag tails. STUDY DESIGN AND METHODS Sixty-one apheresis procedures of 42 patients undergoing autologous HSC transplant were analyzed. Viable CD34+ cells were quantified with flow cytometry before controlled rate freezing (ICE-CUBE14M system, SY-LAB- IceCube, SIAD), after 10 days of storage (mini-bag and bag tail), and before reinfusion (aliquot from a thawed mother bag). Results were compared using Student's t test and Spearman's rho correlation test. RESULTS The mean CD34+ cell viability before cryopreservation was 99.3% (confidence interval [CI], 98.94-99.65%); the mean amount of CD34+ cells, white blood cells and neutrophils in the mother bag was 0.8 ± 1.1 × 109 /L, 63.4 ± 23.5 × 109 /L, and 25.7 ± 15.5 × 109 /L, respectively. Mother bags postthawing CD34+ cell viability was 72.3% (CI, 67.74-76.85%; p < 0.01 compared to prefreezing); no difference was observed with respect to minibags (73.7%; CI, 69.80-77.59%; p = NS), whereas significantly lower values were found for bag tails (58.6%; CI, 54.19-63.00%; p < 0.01 vs. both mini- and mother bags). CONCLUSION Compared to bag tails, minibags represent a more accurate tool to measure the CD34+ cell viability of the apheresis mother bag prior to reinfusion; in addition, minibags may could be of help for case-by-case calculation of the amount of apheresis to be infused to patients undergoing autologous HSC transplant.
Collapse
Affiliation(s)
- Elena Marinelli Busilacchi
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy.,Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Andrea Costantini
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy.,Servizio di Immunologia Clinica, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Giorgia Mancini
- Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Rossella Bencivenga
- Servizio di Medicina Trasfusionale, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Jacopo Olivieri
- Clinica Ematologica, Centro Trapianti e Terapie Cellulari "Carlo Melzi", Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Giovanna Battaglini
- Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Laura Velletri
- Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Nadia Viola
- Servizio di Immunologia Clinica, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Luca Butini
- Servizio di Immunologia Clinica, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Debora Capelli
- Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Antonella Poloni
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy.,Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Attilio Olivieri
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy.,Clinica di Ematologia, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| |
Collapse
|
18
|
Jeyaraman P, Borah P, Dayal N, Pathak S, Naithani R. Adequate Engraftment With Lower Hematopoietic Stem Cell Dose. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:260-263. [PMID: 32019732 DOI: 10.1016/j.clml.2019.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/29/2019] [Accepted: 12/31/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Adequate hematopoietic stem cell dose is required to proceed with autologous stem cell transplantation (ASCT). PATIENTS AND METHODS We conducted a retrospective analysis of 108 patients with multiple myeloma and lymphoma who underwent ASCT with noncryopreserved stem cells at our center. Data were compared for patients who received stem cell dose < 2 × 106/kg with those who received a higher dose. RESULTS The median CD34 dose collected in the lesser dose group was 1.76 × 106/kg (1.22 to 1.97 × 106/kg). Mean CD34 dose of the whole group was 4.96 ± 4.2 × 106/kg. Neutrophil engraftment was similar in both groups (12 vs. 11 days) (P = .065). Similarly, platelet engraftment occurred in 12 versus 11 days in both groups (P = .017). Length of hospital stay was similar in both groups. There was no significant difference in the incidence of proven bacterial infections between the 2 groups. There was no transplant-related mortality in lower dose group. CONCLUSION ASCT can be safely performed with lower hematopoietic stem cell dose in noncryopreserved setting.
Collapse
Affiliation(s)
- Preethi Jeyaraman
- Division of Hematology and Bone Marrow Transplantation, Max Super-specialty Hospital, Saket, New Delhi, India
| | - Pronamee Borah
- Division of Hematology and Bone Marrow Transplantation, Max Super-specialty Hospital, Saket, New Delhi, India
| | - Nitin Dayal
- Department of Lab Medicine, Max Super-specialty Hospital, Saket, New Delhi, India
| | - Sangeeta Pathak
- Department of Transfusion Medicine, Max Super-specialty Hospital, Saket, New Delhi, India
| | - Rahul Naithani
- Division of Hematology and Bone Marrow Transplantation, Max Super-specialty Hospital, Saket, New Delhi, India.
| |
Collapse
|
19
|
Kumar A, Xu Y, Du Y. Stem Cells from Human Trabecular Meshwork Hold the Potential to Develop into Ocular and Non-Ocular Lineages After Long-Term Storage. Stem Cells Dev 2020; 29:49-61. [PMID: 31680626 PMCID: PMC6931915 DOI: 10.1089/scd.2019.0169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/01/2019] [Indexed: 01/10/2023] Open
Abstract
Stem cells from the eye hold a great potential for vision restoration and can also be used for regeneration in other tissues. In this study, we characterized the stem cell properties of Trabecular meshwork stem cells (TMSCs) after long-term cryopreservation (∼8 years). TMSCs derived from four donors were examined for their viability and proliferation, as well as stem cell marker expression. Spheroid formation, colony formation, and multipotency were investigated. We observed that TMSCs were fully viable with variable proliferation ability. They expressed the stem cell markers CD90, CD166, CD105, CD73, OCT4, SSEA4, Notch1, KLF4, ABCG2, Nestin, and HNK1 detected by flow cytometry, quantitative polymerase chain reaction, or immunofluorescent staining. They could form spheroids and colonies after thawing. All TMSCs were able to differentiate into osteocytes, neural cells, and trabecular meshwork (TM) cells, but not adipocytes. Differentiated TM cells responded to dexamethasone treatment with increased expression of myocilin and angiopoietin-like 7 (ANGPTL7). In a nutshell, our study demonstrated that TMSCs retain their stem cell properties after long-term cryopreservation and hence can be an effective cell therapy source for various clinical applications.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yi Xu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Intracellular delivery of mRNA to human primary T cells with microfluidic vortex shedding. Sci Rep 2019; 9:3214. [PMID: 30824814 PMCID: PMC6397276 DOI: 10.1038/s41598-019-40147-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/07/2019] [Indexed: 02/06/2023] Open
Abstract
Intracellular delivery of functional macromolecules, such as DNA and RNA, across the cell membrane and into the cytosol, is a critical process in both biology and medicine. Herein, we develop and use microfluidic chips containing post arrays to induce microfluidic vortex shedding, or μVS, for cell membrane poration that permits delivery of mRNA into primary human T lymphocytes. We demonstrate transfection with μVS by delivery of a 996-nucleotide mRNA construct encoding enhanced green fluorescent protein (EGFP) and assessed transfection efficiencies by quantifying levels of EGFP protein expression. We achieved high transfection efficiency (63.6 ± 3.44% EGFP + viable cells) with high cell viability (77.3 ± 0.58%) and recovery (88.7 ± 3.21%) in CD3 + T cells 19 hrs after μVS processing. Importantly, we show that processing cells via μVS does not negatively affect cell growth rates or alter cell states. We also demonstrate processing speeds of greater than 2.0 × 106 cells s-1 at volumes ranging from 0.1 to 1.5 milliliters. Altogether, these results highlight the use of μVS as a rapid and gentle delivery method with promising potential to engineer primary human cells for research and clinical applications.
Collapse
|
21
|
Hornberger K, Yu G, McKenna D, Hubel A. Cryopreservation of Hematopoietic Stem Cells: Emerging Assays, Cryoprotectant Agents, and Technology to Improve Outcomes. Transfus Med Hemother 2019; 46:188-196. [PMID: 31244587 DOI: 10.1159/000496068] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem cell (HSC) therapy is widely used to treat a growing number of hematological and non-hematological diseases. Cryopreservation of HSCs allows for cells to be transported from the site of processing to the site of clinical use, creates a larger window of time in which cells can be administered to patients, and allows sufficient time for quality control and regulatory testing. Currently, HSCs and other cell therapies conform to the same cryopreservation techniques as cells used for research purposes: cells are cryopreserved in dimethyl sulfoxide (DMSO) at a slow cooling rate. As a result, HSC therapy can result in numerous adverse symptoms in patients due to the infusion of DMSO. Efforts are being made to improve the cryopreservation of HSCs for clinical use. This review discusses advances in the cryopreservation of HSCs from 2007 to the present. The preclinical development of new cryoprotectants and new technology to eliminate cryoprotectants after thawing are discussed in detail. Additional cryopreservation considerations are included, such as cooling rate, storage temperature, and cell concentration. Preclinical cell assessment and quality control are discussed, as well as clinical studies from the past decade that focus on new cryopreservation protocols to improve patient outcomes.
Collapse
Affiliation(s)
- Kathlyn Hornberger
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guanglin Yu
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - David McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Allison Hubel
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
22
|
Roddie C, O'Reilly M, Dias Alves Pinto J, Vispute K, Lowdell M. Manufacturing chimeric antigen receptor T cells: issues and challenges. Cytotherapy 2019; 21:327-340. [PMID: 30685216 DOI: 10.1016/j.jcyt.2018.11.009] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Clinical trials of adoptively transferred CD19 chimeric antigen receptor (CAR) T cells have delivered unprecedented responses in patients with relapsed refractory B-cell malignancy. These results have prompted Food and Drug Administration (FDA) approval of two CAR T-cell products in this high-risk patient population. The widening range of indications for CAR T-cell therapy and increasing patient numbers present a significant logistical challenge to manufacturers aiming for reproducible delivery systems for high-quality clinical CAR T-cell products. This review discusses current and novel CAR T-cell processing methodologies and the quality control systems needed to meet the increasing clinical demand for these exciting new therapies.
Collapse
Affiliation(s)
- Claire Roddie
- Research Department of Haematology, University College London, London, UK; Department of Haematology, University College London Hospitals National Health Service (NHS) Foundation Trust, London.
| | - Maeve O'Reilly
- Research Department of Haematology, University College London, London, UK; Department of Haematology, University College London Hospitals National Health Service (NHS) Foundation Trust, London
| | | | - Ketki Vispute
- Research Department of Haematology, University College London, London, UK
| | - Mark Lowdell
- Research Department of Haematology, University College London, London, UK
| |
Collapse
|
23
|
Rohner N, Passweg JR, Tsakiris DA, Halter JP, Heim D, Buser AS, Infanti L, Holbro A. The value of the post-thaw CD34+ count with and without DMSO removal in the setting of autologous stem cell transplantation. Transfusion 2018; 59:1052-1060. [PMID: 30556582 DOI: 10.1111/trf.15107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND CD34+ cell count correlates with engraftment potency after autologous stem cell transplantation. Assessment of CD34+ mainly occurs after apheresis and before cryopreservation with dimethyl sulfoxide (DMSO). The influence of postthaw CD34+ cell numbers over time to engraftment is not well studied, and determination of postthaw CD34+ cell counts is challenging for a variety of reasons. The aim of this retrospective study was to systematically assess the value of postthaw CD34+ cell counts in autologous grafts with and without DMSO removal. STUDY DESIGN AND METHODS Between January 2008 and December 2015, 236 adult patients underwent a total of 292 autologous stem cell transplantations. Median age at transplantation was 56 years, and the main indication was multiple myeloma (60%). DMSO removal was done in 96 grafts (33%), either by centrifugation or by Sepax method. RESULTS Patients receiving grafts containing DMSO showed a significantly faster platelet (p = 0.02) and RBC (p = 0.001) engraftment. DMSO removal was not associated with fewer infusion-related adverse events. We observed a good correlation between CD34+ cell count after apheresis and CD34+ cell count after thawing/washing (r = 0.931). Ninety grafts (31%) showed a significant loss of viable CD34+ cells, which translated into a delayed engraftment. CONCLUSION DMSO removal was associated with delayed platelet and RBC engraftment without preventing adverse events. CD34+ cell enumeration after thawing remains difficult to perform, but grafts showing higher cell loss during cryopreservation and thawing are associated with slower engraftment. Prospective studies on the role of DMSO removal and postthaw CD34+ enumeration using defined protocols are needed.
Collapse
Affiliation(s)
- Nicolai Rohner
- Division of Hematology, University Hospital Basel, Switzerland
| | - Jakob R Passweg
- Division of Hematology, University Hospital Basel, Switzerland
| | | | - Jörg P Halter
- Division of Hematology, University Hospital Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital Basel, Switzerland
| | - Andreas S Buser
- Division of Hematology, University Hospital Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| | - Laura Infanti
- Division of Hematology, University Hospital Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
24
|
Kraal KCJM, Timmerman I, Kansen HM, van den Bos C, Zsiros J, van den Berg H, Somers S, Braakman E, Peek AML, van Noesel MM, van der Schoot CE, Fiocco M, Caron HN, Voermans C, Tytgat GAM. Peripheral Stem Cell Apheresis is Feasible Post 131Iodine-Metaiodobenzylguanidine-Therapy in High-Risk Neuroblastoma, but Results in Delayed Platelet Reconstitution. Clin Cancer Res 2018; 25:1012-1021. [PMID: 30314967 DOI: 10.1158/1078-0432.ccr-18-1904] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/01/2018] [Accepted: 10/09/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Targeted radiotherapy with 131iodine-meta-iodobenzylguanidine (131I-MIBG) is effective for neuroblastoma (NBL), although optimal scheduling during high-risk (HR) treatment is being investigated. We aimed to evaluate the feasibility of stem cell apheresis and study hematologic reconstitution after autologous stem cell transplantation (ASCT) in patients with HR-NBL treated with upfront 131I-MIBG-therapy. EXPERIMENTAL DESIGN In two prospective multicenter cohort studies, newly diagnosed patients with HR-NBL were treated with two courses of 131I-MIBG-therapy, followed by an HR-induction protocol. Hematopoietic stem and progenitor cell (e.g., CD34+ cell) harvest yield, required number of apheresis sessions, and time to neutrophil (>0.5 × 109/L) and platelet (>20 × 109/L) reconstitution after ASCT were analyzed and compared with "chemotherapy-only"-treated patients. Moreover, harvested CD34+ cells were functionally (viability and clonogenic capacity) and phenotypically (CD33, CD41, and CD62L) tested before cryopreservation (n = 44) and/or after thawing (n = 19). RESULTS Thirty-eight patients (47%) were treated with 131I-MIBG-therapy, 43 (53%) only with chemotherapy. Median cumulative 131I-MIBG dose/kg was 0.81 GBq (22.1 mCi). Median CD34+ cell harvest yield and apheresis days were comparable in both groups. Post ASCT, neutrophil recovery was similar (11 days vs. 10 days), whereas platelet recovery was delayed in 131I-MIBG- compared with chemotherapy-only-treated patients (29 days vs. 15 days, P = 0.037). Testing of harvested CD34+ cells revealed a reduced post-thaw viability in the 131I-MIBG-group. Moreover, the viable CD34+ population contained fewer cells expressing CD62L (L-selectin), a marker associated with rapid platelet recovery. CONCLUSIONS Harvesting of CD34+ cells is feasible after 131I-MIBG. Platelet recovery after ASCT was delayed in 131I-MIBG-treated patients, possibly due to reinfusion of less viable and CD62L-expressing CD34+ cells, but without clinical complications. We provide evidence that peripheral stem cell apheresis is feasible after upfront 131I-MIBG-therapy in newly diagnosed patients with NBL. However, as the harvest of 131I-MIBG-treated patients contained lower viable CD34+ cell counts after thawing and platelet recovery after reinfusion was delayed, administration of 131I-MIBG after apheresis is preferred.
Collapse
Affiliation(s)
- Kathelijne C J M Kraal
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| | - Ilse Timmerman
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands.,Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Hannah M Kansen
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands.,Department of Paediatric Pulmonology and Allergology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Cor van den Bos
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| | - Jozsef Zsiros
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| | - Henk van den Berg
- Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| | - Sebastiaan Somers
- Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| | - Eric Braakman
- Department of Hematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Annemarie M L Peek
- Department of Pediatric Oncology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Marta Fiocco
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.,Mathematical Institute, Leiden University, Leiden, the Netherlands
| | - Huib N Caron
- Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology (PMC), Utrecht, the Netherlands. .,Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, the Netherlands
| |
Collapse
|
25
|
Nath K, Boles R, McCutchan A, Vangaveti V, Birchley A, Irving I. The relationship between CD34+ stem cell dose and time to neutrophil recovery in autologous haematopoietic stem cell recipients-A single centre experience. Transfus Apher Sci 2018; 57:532-536. [PMID: 29933906 DOI: 10.1016/j.transci.2018.05.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/27/2018] [Accepted: 05/31/2018] [Indexed: 10/14/2022]
Abstract
A retrospective, observational study was performed of 112 patients who underwent autologous haematopoietic stem cell transplantation (ASCT) to determine the relationship between CD34+ stem cell dose and neutrophil engraftment. Importantly, a novel approach to more accurately calculate time to neutrophil engraftment was employed. The results demonstrated that a higher CD34+ stem cell dose was associated with faster neutrophil recovery (P < 0.05). CD34+ stem cell dose using actual and ideal patient body weight were both equally predictive of neutrophil engraftment as were absolute and viable CD34+ measurements. The clinical implications for this relationship are limited with an increase in CD34+ stem cell dose by 1 × 106/kg reducing the neutrophil engraftment time by only 3 h and 50 min. The median time to neutrophil recovery was 217 h (9 days and 1 h) and this relatively early engraftment time may be related to an early initiation of granulocyte colony-stimulating factor (G-CSF) on day +1 post-transplant. Female patients engrafted 17 h faster than their male counterparts on multi-variate analysis (P < 0.05). Conditioning chemotherapy, bacteraemia, G-CSF dose/kg body weight and increasing age had no impact on time to neutrophil recovery.
Collapse
Affiliation(s)
- Karthik Nath
- Department of Haematology and Bone Marrow Transplantation, Townsville Hospital, Townsville, Australia.
| | - Rachael Boles
- Department of Haematology and Bone Marrow Transplantation, Townsville Hospital, Townsville, Australia
| | - Andrew McCutchan
- Department of Haematology and Bone Marrow Transplantation, Townsville Hospital, Townsville, Australia
| | - Venkat Vangaveti
- College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Andrew Birchley
- Department of Haematology and Bone Marrow Transplantation, Townsville Hospital, Townsville, Australia
| | | |
Collapse
|
26
|
Holbro A, Baldomero H, Lanza F, Chabannon C, Snowden JA, Buser A, Infanti L, Worel N, Sureda A, Badoglio M, Passweg J, Bonini C. Handling, processing and disposal of stem cell products in Europe: A survey by the cellular therapy and immunobiology working party of the European Society for Blood and Marrow Transplantation. Cytotherapy 2018; 20:453-460. [DOI: 10.1016/j.jcyt.2017.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/03/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
|
27
|
Ko DW, Yoon JK, Ahn JI, Lee M, Yang WS, Ahn JY, Lim JM. The importance of post-thaw subculture for standardizing cellular activity of fresh or cryopreserved mouse embryonic stem cells. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2017; 31:335-343. [PMID: 28823125 PMCID: PMC5838338 DOI: 10.5713/ajas.17.0294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/08/2017] [Accepted: 08/07/2017] [Indexed: 12/25/2022]
Abstract
Objective Remarkable difference in cellular activity was found between early and late subpassaged embryonic stem cell (ESCs) lines, which can be created by subtle changes in cell manipulation protocol. This study subsequently examined whether post-thaw subculture of early subpassaged ESC lines could further affect the activity of the ESCs. Methods Fresh (as a control treatment) or cryopreserved F1 hybrid (B6CBAF1) early ESC lines (C57BL/6xCBA) of the 4 (P4) or the 19 passage (P19) were subcultured once, twice or six times under the same condition. The post-thaw survival of the ESCs was monitored after the post-treatment subculture and the ability of cell proliferation, reactive oxygen species (ROS) generation, apoptosis and mitochondrial ATP synthesis was subsequently examined. Results Regardless of the subculture number, P19 ESCs showed better (p<0.05) doubling time and less ATP production than P4 ESCs and such difference was not influenced by fresh or cryopreservation. The difference between P4 and P19 ESC lines became decreased as the post-treatment subculture was increased and the six times subculture eliminated such difference. Similarly, transient but prominent difference in ROS production and apoptotic cell number was detected between P4 and P19 ESCs only at the 1st subculture after treatment, but no statistical differences between two ESC lines was detected in other observations. Conclusion The results of this study suggest that post-thaw subculture of ESCs under the same environment is recommended for standardizing their cellular activity. The activity of cell proliferation ability and ATP synthesis can be used as parameters for quality control of ESCs.
Collapse
Affiliation(s)
- Dong Woo Ko
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Research Institutes of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jung Ki Yoon
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Research Institutes of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jong Il Ahn
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Research Institutes of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Myungook Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Research Institutes of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Woo Sub Yang
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Research Institutes of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Ji Yeon Ahn
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Jeong Mook Lim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Research Institutes of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.,GreenBio Research Institute, Seoul National University, Pyeongchang 25354, Korea
| |
Collapse
|
28
|
Analysis of hematopoietic recovery after autologous transplantation as method of quality control for long-term progenitor cell cryopreservation. Bone Marrow Transplant 2017. [DOI: 10.1038/bmt.2017.113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Wang X, Rivière I. Genetic Engineering and Manufacturing of Hematopoietic Stem Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:96-105. [PMID: 28480310 PMCID: PMC5415326 DOI: 10.1016/j.omtm.2017.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The marketing approval of genetically engineered hematopoietic stem cells (HSCs) as the first-line therapy for the treatment of severe combined immunodeficiency due to adenosine deaminase deficiency (ADA-SCID) is a tribute to the substantial progress that has been made regarding HSC engineering in the past decade. Reproducible manufacturing of high-quality, clinical-grade, genetically engineered HSCs is the foundation for broadening the application of this technology. Herein, the current state-of-the-art manufacturing platforms to genetically engineer HSCs as well as the challenges pertaining to production standardization and product characterization are addressed in the context of primary immunodeficiency diseases (PIDs) and other monogenic disorders.
Collapse
Affiliation(s)
- Xiuyan Wang
- Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle Rivière
- Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
30
|
Morgenstern DA, Gray P, Prudhoe A, Watts M, Wheeler K. Developing quality assurance for pediatric autologous stem cell transplants in England: results of a 3-year national audit of activity and engraftment by treatment centre. Bone Marrow Transplant 2017; 52:922-925. [DOI: 10.1038/bmt.2017.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Watts MJ, Linch DC. Optimisation and quality control of cell processing for autologous stem cell transplantation. Br J Haematol 2016; 175:771-783. [DOI: 10.1111/bjh.14378] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Michael J. Watts
- University College London Hospitals; London UK
- University College London; London UK
| | - David C. Linch
- University College London Hospitals; London UK
- University College London; London UK
| |
Collapse
|
32
|
Hokland P, Cotter F. Honest reporting and elucidation of very serious adverse events: a lesson to us all. Br J Haematol 2016; 174:341-2. [PMID: 27340811 DOI: 10.1111/bjh.14159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Peter Hokland
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | - Finbarr Cotter
- Centre for Haemato-Oncology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|