1
|
Cheng P, Rashad A, Gangrade A, Barros NRD, Khademhosseini A, Tam J, Varadarajan P, Agrawal DK, Thankam FG. Stem Cell-Derived Cardiomyocyte-Like Cells in Myocardial Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:1-14. [PMID: 37294202 DOI: 10.1089/ten.teb.2023.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Myocardial infarction results in the significant loss of cardiomyocytes (CMs) due to the ischemic injury following coronary occlusion leading to impaired contractility, fibrosis, and ultimately heart failure. Stem cell therapy emerged as a promising regenerative strategy to replenish the otherwise terminally differentiated CM to restore cardiac function. Multiple strategies have been applied to successfully differentiate diverse stem cell populations into CM-like phenotypes characterized by the expression status of signature biomarkers and observable spontaneous contractions. This article discusses the current understanding and applications of various stem cell phenotypes to drive the differentiation machinery toward CM-like lineage. Impact Statement Ischemic heart disease (IHD) extensively affects a large proportion of the population worldwide. Unfortunately, current treatments for IHD are insufficient to restore cardiac effectiveness and functionality. A growing field in regenerative cardiology explores the potential for stem cell therapy following cardiovascular ischemic episodes. The thorough understanding regarding the potential and shortcomings of translational approaches to drive versatile stem cells to cardiomyocyte lineage paves the way for multiple opportunities for next-generation cardiac management.
Collapse
Affiliation(s)
- Pauline Cheng
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Ahmad Rashad
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Jonathan Tam
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Padmini Varadarajan
- University of California Riverside School of Medicine, Riverside, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
2
|
Askari MHA, Shahabi M, Kojabad AA, Zarif MN. Reconstruction of bone marrow microenvironment for expansion of hematopoietic stem cells by a histone deacetylase inhibitor. Cytotechnology 2023; 75:195-206. [PMID: 37187947 PMCID: PMC10167084 DOI: 10.1007/s10616-022-00564-w] [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: 07/25/2022] [Accepted: 12/06/2022] [Indexed: 05/17/2023] Open
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) is an approach for overcoming cell insufficiency for umbilical cord blood transplantation. It was suggested that in common ex vivo cultures, the stemness specificity of HSCs is rapidly reducing due to DNA hypermethylation. Here, Nicotinamide (NAM), a DNA methyltransferase and histone deacetylase inhibitor, is used with a bioengineered Bone Marrow-like niche (BLN) for HSC ex vivo expansion. The CFSE cell proliferation assay was used for tracking HSCs division. qRT-PCR was conducted to assay the HOXB4 mRNA expression levels. The morphology of BLN-cultured cells was analyzed using scanning electron microscopy (SEM). NAM boosted the induction of HSC proliferation in the BLN group compared to the control group. In addition, the ability of HSCs to colonize was more significant in the BLN group than in the control group. Our data suggest that the presence of NAM in bioengineered niches promotes HSC proliferation. The presented approach showed that small molecules could be used in the clinical setting to overcome the limited number of CD34+ cells in cord blood units.
Collapse
Affiliation(s)
- Maryam Haj Ali Askari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, IBTO Building, Hemmat Expressway, Tehran, Iran
| | - Majid Shahabi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, IBTO Building, Hemmat Expressway, Tehran, Iran
| | - Amir Asri Kojabad
- Department of Hematology and Blood Bank, Iran University of Medical Sciences, Tehran, Iran
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, IBTO Building, Hemmat Expressway, Tehran, Iran
- Department of Medicine, Center for Hematology and Regenerative Medicin, 14183 Stockholm, Sweden
- Cell Therapy Department, XNKtheraeutics Company, Stockholm, Sweden
| |
Collapse
|
3
|
Wang P, Liu C, Wei Z, Jiang W, Sun H, Wang Y, Hou J, Sun J, Huang Y, Wang H, Wang Y, He X, Wang X, Qian X, Zhai X. Nomogram for Predicting Early Mortality after Umbilical Cord Blood Transplantation in Children with Inborn Errors of Immunity. J Clin Immunol 2023:10.1007/s10875-023-01505-8. [PMID: 37155023 DOI: 10.1007/s10875-023-01505-8] [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: 03/24/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE Pediatric patients with inborn errors of immunity (IEI) undergoing umbilical cord blood transplantation (UCBT) are at risk of early mortality. Our aim was to develop and validate a prediction model for early mortality after UCBT in pediatric IEI patients based on pretransplant factors. METHODS Data from 230 pediatric IEI patients who received their first UCBT between 2014 and 2021 at a single center were analyzed retrospectively. Data from 2014-2019 and 2020-2021 were used as training and validation sets, respectively. The primary outcome of interest was early mortality. Machine learning algorithms were used to identify risk factors associated with early mortality and to build predictive models. The model with the best performance was visualized using a nomogram. Discriminative ability was measured using the area under the curve (AUC) and decision curve analysis. RESULTS Fifty days was determined as the cutoff for distinguishing early mortality in pediatric IEI patients undergoing UCBT. Of the 230 patients, 43 (18.7%) suffered early mortality. Multivariate logistic regression with pretransplant albumin, CD4 (absolute count), elevated C-reactive protein, and medical history of sepsis showed good discriminant AUC values of 0.7385 (95% CI, 0.5824-0.8945) and 0.827 (95% CI, 0.7409-0.9132) in predicting early mortality in the validation and training sets, respectively. The sensitivity and specificity were 0.5385 and 0.8154 for validation and 0.7667 and 0.7705 for training, respectively. The final model yielded net benefits across a reasonable range of risk thresholds. CONCLUSION The developed nomogram can predict early mortality in pediatric IEI patients undergoing UCBT.
Collapse
Affiliation(s)
- Ping Wang
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Chao Liu
- Yidu Cloud Technology Inc, Beijing, 100083, China
- Nanjing YiGenCloud Institute, Nanjing, 211899, China
| | - Zhongling Wei
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Wenjin Jiang
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Hua Sun
- Department of Gastroenterology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yuhuan Wang
- Department of Gastroenterology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Jia Hou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Ying Huang
- Department of Gastroenterology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Hongsheng Wang
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yao Wang
- Yidu Cloud Technology Inc, Beijing, 100083, China
| | - Xinjun He
- Yidu Cloud Technology Inc, Beijing, 100083, China
- Nanjing YiGenCloud Institute, Nanjing, 211899, China
| | - Xiaochuan Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xiaowen Qian
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xiaowen Zhai
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| |
Collapse
|
4
|
Hong J, Zheng W, Wang X, Hao Y, Cheng G. Biomedical polymer scaffolds mimicking bone marrow niches to advance in vitro expansion of hematopoietic stem cells. J Mater Chem B 2022; 10:9755-9769. [PMID: 36444902 DOI: 10.1039/d2tb01211a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hematopoietic stem cell (HSC) transplantation provides an effective platform for the treatment of hematological disorders. However, the donor shortage of HSCs and immune responses severely restrict the clinical applications of HSCs. Compared to allogeneic transplantation, autogenous transplantation poses less risk to the immune system, but the problem associated with insufficient HSCs remains a substantial challenge. A significant strategy for obtaining sufficient HSCs is to promote the expansion of HSCs. In vivo, a bone marrow microenvironment supports the survival and hematopoiesis of HSCs. Therefore, it is crucial to establish a platform that mimics the features of a bone marrow microenvironment for the in vitro expansion of HSCs. Three-dimensional (3D) scaffolds have emerged as the most powerful tools to mimic cellular microenvironments for the growth and proliferation of stem cells. Biomedical polymers have been widely utilized as cell scaffolds due to their advantageous features including favorable biocompatibility, biodegradability, as well as adjustable physical and chemical properties. This review focuses on recent advances in the study of biomedical polymer scaffolds that mimic bone marrow microenvironments for the in vitro expansion of HSCs. Bone marrow transplantation and microenvironments are first introduced. Then, biomedical polymer scaffolds for the expansion of HSCs and future prospects are summarized and discussed.
Collapse
Affiliation(s)
- Jing Hong
- Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong 528200, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu 215123, China. .,School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Anhui 230026, China
| | - Wenlong Zheng
- Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Jiangsu 215021, China
| | | | - Ying Hao
- Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong 528200, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu 215123, China. .,School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Anhui 230026, China
| | - Guosheng Cheng
- Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong 528200, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu 215123, China. .,School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Anhui 230026, China
| |
Collapse
|
5
|
Inflammatory monocytes promote pre-engraftment syndrome and tocilizumab can therapeutically limit pathology in patients. Nat Commun 2021; 12:4137. [PMID: 34230468 PMCID: PMC8260612 DOI: 10.1038/s41467-021-24412-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 06/15/2021] [Indexed: 12/01/2022] Open
Abstract
Unrelated cord blood transplantation (UCBT) is an effective treatment for hematopoietic disorders. However, this attractive approach is frequently accompanied by pre-engraftment syndrome (PES), severe cases of PES are associated with enhanced mortality and morbidity, but the pathogenesis of PES remains unclear. Here we show that GM-CSF produced by cord blood-derived inflammatory monocytes drives PES pathology, and that monocytes are the main source of IL-6 during PES. Further, we report the outcome of a single arm, single-center clinical study of tocilizumab in the treatment of steroid-refractory severe PES patients (www.chictr.org.cn ChiCTR1800015472). The study met the primary outcome measure since none of the patients was nonrelapse death during the 100 days follow-up. The study also met key secondary outcomes measures of neutrophil engraftment and hematopoiesis. These findings offer a therapeutic strategy with which to tackle PES and improve nonrelapse mortality. Pre-engraftment syndrome is a major consideration during clinical application of unrelated cord blood transfusion and monocytes represent a critical cell type in immune-pathogenesis. Here the authors further establish the role of monocytes and GM-CSF in pre-engraftment syndrome and show clinical administration of tocilizumab limits pathology in pre-engraftment syndrome pathology in patients.
Collapse
|
6
|
Yoo KH. Strategies to enhance graft performance in cord blood transplantation. PRECISION AND FUTURE MEDICINE 2021. [DOI: 10.23838/pfm.2020.00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
7
|
Ghebes CA, Morhayim J, Kleijer M, Koroglu M, Erkeland SJ, Hoogenboezem R, Bindels E, van Alphen FPJ, van den Biggelaar M, Nolte MA, van der Eerden BCJ, Braakman E, Voermans C, van de Peppel J. Extracellular Vesicles Derived From Adult and Fetal Bone Marrow Mesenchymal Stromal Cells Differentially Promote ex vivo Expansion of Hematopoietic Stem and Progenitor Cells. Front Bioeng Biotechnol 2021; 9:640419. [PMID: 33718342 PMCID: PMC7947881 DOI: 10.3389/fbioe.2021.640419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/02/2021] [Indexed: 01/05/2023] Open
Abstract
Recently, we and others have illustrated that extracellular vesicles (EVs) have the potential to support hematopoietic stem and progenitor cell (HSPC) expansion; however, the mechanism and processes responsible for the intercellular communication by EVs are still unknown. In the current study, we investigate whether primary human bone marrow derived mesenchymal stromal cells (BMSC) EVs isolated from two different origins, fetal (fEV) and adult (aEV) tissue, can increase the relative low number of HSPCs found in umbilical cord blood (UCB) and which EV-derived components are responsible for ex vivo HSPC expansion. Interestingly, aEVs and to a lesser extent fEVs, showed supportive ex vivo expansion capacity of UCB-HSPCs. Taking advantage of the two BMSC sources with different supportive effects, we analyzed the EV cargo and investigated how gene expression is modulated in HSPCs after incubation with aEVs and fEVs. Proteomics analyses of the protein cargo composition of the supportive aEV vs. the less-supportive fEV identified 90% of the Top100 exosome proteins present in the ExoCarta database. Gene Ontology (GO) analyses illustrated that the proteins overrepresented in aEVs were annotated to oxidation-reduction process, mitochondrial ATP synthesis coupled proton transport, or protein folding. In contrast, the proteins overrepresented in fEVs were annotated to extracellular matrix organization positive regulation of cell migration or transforming growth factor beta receptor (TGFBR) signaling pathway. Small RNA sequencing identified different molecular signatures between aEVs and fEVs. Interestingly, the microRNA cluster miR-99b/let-7e/miR-125a, previously identified to increase the number of HSPCs by targeting multiple pro-apoptotic genes, was highly and significantly enriched in aEVs. Although we identified significant differences in the supportive effects of aEVs and fEVs, RNAseq analyses of the 24 h treated HSPCs indicated that a limited set of genes was differentially regulated when compared to cells that were treated with cytokines only. Together, our study provides novel insights into the complex biological role of EVs and illustrates that aEVs and fEVs differentially support ex vivo expansion capacity of UCB-HSPCs. Together opening new means for the application of EVs in the discovery of therapeutics for more efficient ex vivo HSPC expansion.
Collapse
Affiliation(s)
- Corina A Ghebes
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands
| | - Jess Morhayim
- Department of Hematology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Marion Kleijer
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands
| | - Merve Koroglu
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands
| | - Stefan J Erkeland
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Remco Hoogenboezem
- Department of Hematology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Eric Bindels
- Department of Hematology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | | | | | - Martijn A Nolte
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Department of Molecular Hematology, Sanquin Research, Amsterdam, Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Eric Braakman
- Department of Hematology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
8
|
A 3D engineered scaffold for hematopoietic progenitor/stem cell co-culture in vitro. Sci Rep 2020; 10:11485. [PMID: 32661289 PMCID: PMC7359311 DOI: 10.1038/s41598-020-68250-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/11/2020] [Indexed: 01/29/2023] Open
Abstract
Proliferation of HPSCs in vitro can promote its broad clinical therapeutic use. For in vitro co-culture, interaction between the stem cell and feeder cell as well as their spatial position are essential. To imitate the natural microenvironment, a 3D engineered scaffold for CD34+ cells co-culture was established via 3D bioprinting. Herein, the concentration of hydrogel and the ratio of two kinds of cells were optimized. Flow cytometry, real time PCR and RNA-seq technology were applied to analyze the effect of the engineered scaffold on expanded cells. After 10 days co-culture with the engineered scaffold, the expansion of CD34+CD38- cells can reach 33.57-folds and the expansion of CD34+CD184+ cells can reach 16.66-folds. Result of PCR and RNA-seq indicates that the CD34+ cells in 3D group exhibited a tendency of interaction with the engineered scaffold. Compared to 2D co-culture, this customizable 3D engineered scaffold can provide an original and integrated environment for HPSCs growth. Additionally, this scaffold can be modified for different cell co-culture or cell behavior study.
Collapse
|
9
|
Emiloju OE, Potdar R, Jorge V, Gupta S, Varadi G. Clinical Advancement and Challenges of ex vivo Expansion of Human Cord Blood Cells. Clin Hematol Int 2019; 2:18-26. [PMID: 34595439 PMCID: PMC8432338 DOI: 10.2991/chi.d.191121.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/16/2019] [Indexed: 02/02/2023] Open
Abstract
Apart from peripheral blood stem cell (PBSC), umbilical cord blood (UCB) is now a recognized source of stem cells for transplantation. UCB is an especially important source of stem cells for minority populations, which would otherwise be unable to find appropriately matched adult donors. UCB has fewer mature T lymphocytes compared with peripheral blood, thus making a UCB transplantation (UCBT) with a greater degree of HLA mismatch possible. The limited cell dose per UCB sample is however associated with delayed engraftment and a higher risk of graft failure, especially in adult recipients. This lower cell dose can be optimized by performing double unit UCBT, ex vivo UCB expansion prior to transplant and enhancement of the capabilities of the stem cells to home to the bone marrow. UCB contains naïve and immature T cells, thus posing significant challenges with increased risk of infections, graft versus host diseases (GVHD) and relapse following UCBT. Cell engineering techniques have been developed to circumnavigate the immaturity of the T cells, and include virus-specific cytotoxic T cells (VSTs), T cells transduced with disease-specific chimeric antigen receptor (CAR T cells) and regulatory T cell (Tregs) engineering. In this article, we review the advances in UCB ex vivo expansion and engineering to improve engraftment and reduce complications. As further research continues to find ways to overcome the current challenges, outcomes from UCBT will likely improve.
Collapse
Affiliation(s)
| | - Rashmika Potdar
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Vinicius Jorge
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Sorab Gupta
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Gabor Varadi
- Hematology and Oncology Department, Albert Einstein Medical Center, Philadelphia, PA, USA
| |
Collapse
|
10
|
Szostak B, Machaj F, Rosik J, Pawlik A. Umbilical cord blood transplantation and the impact of the CTLA4 genotype on outcomes. Expert Rev Hematol 2019; 12:1089-1094. [PMID: 31607188 DOI: 10.1080/17474086.2019.1680281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background: Umbilical cord blood transplantation is an effective method of curing multiple diseases when there is no donor available for allogeneic hematopoietic stem cell transplantation (AHSCT). It has been recently suggested that polymorphisms in genes affecting antigen presentation could potentially affect cord blood transplantation (CBT) outcomes.Areas covered: In this review, we present the results of the latest studies investigating the link between CTLA4 gene variability and umbilical cord blood transplantation outcomes.Expert opinion: The search for genetic variants that influence the immune response, both innate and adaptive immunity, may lead to more optimal therapies. Promising candidate genes are those that regulate the expression of proteins associated with T-cell activation. Many genetic variants could be therapeutically important, including those related to innate and adaptive immunity, cytokines, chemokines, drug-metabolizing enzymes, drug transporters, and inflammatory enzymes. The development of an algorithm that includes the determination of selected genetic variants could be helpful for an appropriate donor-recipient CBT matching.
Collapse
Affiliation(s)
- Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Filip Machaj
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
11
|
Liver Sinusoidal Endothelial Cells Promote the Expansion of Human Cord Blood Hematopoietic Stem and Progenitor Cells. Int J Mol Sci 2019; 20:ijms20081985. [PMID: 31018542 PMCID: PMC6515002 DOI: 10.3390/ijms20081985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/20/2019] [Accepted: 04/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cord blood (CB) is an attractive source of hematopoietic stem cells (HSCs) for hematopoietic cell transplantation. However, its application remains limited due to the low number of HSCs/progenitors in a single CB unit and its notoriously difficulty in expanding ex vivo. Here, we demonstrated that the human fetal liver sinusoidal endothelial cells engineered to constitutively express the adenoviral E4orf1 gene (hFLSECs-E4orf1) is capable of efficient expansion ex vivo for human CB hematopoietic stem and progenitor cells (HSPCs). Coculture of CD34+ hCB cells with hFLSECs-E4orf1 resulted in generation of substantially more total nucleated cells, CD34+CD38− and CD34+ CD38−CD90+ HSPCs in comparison with that of cytokines alone after 14 days. The multilineage differentiation potential of the expanded hematopoietic cells in coculture condition, as assessed by in vitro colony formation, was also significantly heightened. The CD34+ hCB cells amplified on hFLSECs-E4orf1 were capable of engraftment in vivo. Furthermore, hFLSECs-E4orf1 highly expressed hematopoiesis related growth factor and Notch receptors. Accordingly, the CD34+ hCB cells amplified on hFLSECs-E4orf1 exhibited Notch signaling activation. Taken together, our findings indicated that FLSECs may potentially be the crucial component of the microenvironment to support recapitulation of embryonic HSC amplification in vitro and allow identification of new growth factors responsible for collective regulation of hematopoiesis.
Collapse
|
12
|
Kanate AS, Szabo A, Raj RV, Bower K, Grulke R, Shah N, Ross KG, Cumpston A, Craig M, Pasquini MC, Shah N, Hari P, Hamadani M, Chhabra S. Comparison of Graft Acquisition and Early Direct Charges of Haploidentical Related Donor Transplantation versus Umbilical Cord Blood Transplantation. Biol Blood Marrow Transplant 2019; 25:1456-1464. [PMID: 30878605 DOI: 10.1016/j.bbmt.2019.03.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/10/2019] [Indexed: 11/18/2022]
Abstract
Alternative donor allogeneic hematopoietic cell transplants (HCTs), such as double umbilical cord blood transplants (dUCBT) and haploidentical related donor transplants (haplo-HCT), have been shown to be safe and effective in adult patients who do not have an HLA-identical sibling or unrelated donor available. Most transplant centers have committed to 1 of the 2 alternative donor sources, even with a lack of published randomized data directly comparing outcomes and comparative data on the cost-effectiveness of dUCBT versus haplo-HCT. We conducted a retrospective study to evaluate and compare the early costs and charges of haplo-HCT and dUCBT in the first 100 days at 2 US transplant centers. Forty-nine recipients of haplo-HCT (at 1 center) and 37 with dUCBT (at another center) were included in the analysis. We compared graft acquisition, inpatient/outpatient, and total charges in the first 100 days. The results of the analysis showed a significantly lower cost of graft acquisition and lower total charges (for 100-day HCT survivors) in favor of haplo-HCT. Importantly, to control for the obvious shortcomings of comparing costs at 2 different transplant centers, adjustments were made based on the current (2018) local wage index and inflation rate. In the absence of further guidance from a prospective study, the cost analysis in this study suggests that haplo-HCT may result in early cost savings over dUCBT and may be preferred by transplant centers and for patients with more limited resources.
Collapse
Affiliation(s)
- Abraham S Kanate
- Osborn Hematopoietic Malignancy and Cellular Therapy Program, West Virginia University, Morgantown, West Virginia
| | - Aniko Szabo
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Renju V Raj
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kathryn Bower
- Osborn Hematopoietic Malignancy and Cellular Therapy Program, West Virginia University, Morgantown, West Virginia
| | - Rachel Grulke
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nilay Shah
- Osborn Hematopoietic Malignancy and Cellular Therapy Program, West Virginia University, Morgantown, West Virginia
| | - Kelly G Ross
- Osborn Hematopoietic Malignancy and Cellular Therapy Program, West Virginia University, Morgantown, West Virginia
| | - Aaron Cumpston
- Osborn Hematopoietic Malignancy and Cellular Therapy Program, West Virginia University, Morgantown, West Virginia
| | - Michael Craig
- Osborn Hematopoietic Malignancy and Cellular Therapy Program, West Virginia University, Morgantown, West Virginia
| | - Marcelo C Pasquini
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nirav Shah
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Parameswaran Hari
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mehdi Hamadani
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
13
|
Du W, Liu W, Mizukawa B, Shang X, Sipple J, Wunderlich M, Geiger H, Davies S, Mulloy J, Pang Q, Zheng Y. A non-myeloablative conditioning approach for long-term engraftment of human and mouse hematopoietic stem cells. Leukemia 2018; 32:2041-2046. [PMID: 29959415 PMCID: PMC6128741 DOI: 10.1038/s41375-018-0200-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Wei Du
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, 1 Medical Center Dr., Morgantown, WV, 26506, USA.
| | - Wei Liu
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Benjamin Mizukawa
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Xun Shang
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Jared Sipple
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Mark Wunderlich
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Hartmut Geiger
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Stella Davies
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - James Mulloy
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Qishen Pang
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| | - Yi Zheng
- Cancer and Blood Diseases Institute, Molecular and Developmental Biology Graduate Program, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
14
|
Dircio‐Maldonado R, Flores‐Guzman P, Corral‐Navarro J, Mondragón‐García I, Hidalgo‐Miranda A, Beltran‐Anaya FO, Cedro‐Tanda A, Arriaga‐Pizano L, Balvanera‐Ortiz O, Mayani H. Functional Integrity and Gene Expression Profiles of Human Cord Blood-Derived Hematopoietic Stem and Progenitor Cells Generated In Vitro. Stem Cells Transl Med 2018; 7:602-614. [PMID: 29701016 PMCID: PMC6090508 DOI: 10.1002/sctm.18-0013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/29/2018] [Indexed: 12/16/2022] Open
Abstract
To date, different experimental strategies have been developed for the ex vivo expansion of human hematopoietic stem (HSCs) and progenitor (HPCs) cells. This has resulted in significant advances on the use of such expanded cells in transplantation settings. To this day, however, it is still unclear to what extent those stem and progenitor cells generated in vitro retain the functional and genomic integrity of their freshly isolated counterparts. In trying to contribute to the solving of this issue, in the present study we have selected and purified three different hematopoietic cell populations: HSCs (CD34+ CD38- CD45RA- CD71- Lin- cells), myeloid progenitor cells (CD34+ CD38+ CD45RA+ CD71- Lin- cells), and erythroid progenitor cells (CD34+ CD38+ CD45RA- CD71+ Lin- cells), obtained directly from fresh human umbilical cord blood (UCB) units or generated in vitro under particular culture conditions. We, then, compared their functional integrity in vitro and their gene expression profiles. Our results indicate that in spite of being immunophenotipically similar, fresh and in vitro generated cells showed significant differences, both in functional and genetic terms. As compared to their fresh counterparts, those HSCs generated in our culture system showed a deficient content of long-term culture-initiating cells, and a marked differentiation bias toward the myeloid lineage. In addition, in vitro generated HSCs and HPCs showed a limited expansion potential. Such functional alterations correlated with differences in their gene expression profiles. These observations are relevant in terms of HSC biology and may have implications in UCB expansion and transplantation. Stem Cells Translational Medicine 2018;7:602-614.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alberto Cedro‐Tanda
- National Ministry of HealthNational Institute of Genomic MedicineMexico CityMexico
| | - Lourdes Arriaga‐Pizano
- Immunochemistry Research Unit, Medical Specialties Hospital, IMSS National Medical CenterMexico CityMexico
| | | | - Hector Mayani
- Hematopoietic Stem Cells LaboratoryOncology Research Unit, Oncology Hospital
| |
Collapse
|
15
|
Ferreira MSV, Mousavi SH. Nanofiber technology in the ex vivo expansion of cord blood-derived hematopoietic stem cells. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:1707-1718. [PMID: 29753127 DOI: 10.1016/j.nano.2018.04.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/18/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
Umbilical cord blood (CB) can be used as an alternative source of hematopoietic stem cells (HSCs) for transplantation in hematological and non-hematological disorders. Despite several recognized advantages the limited cell number in CB one unit still restricts its clinical use. The success of transplantation greatly depends on the levels of total nucleated cell and CD34+ cell counts. Thus, many ex vivo strategies have been developed within the last decade in order to solve this obstacle, with more or less success, mainly determined by the degree of difficulty related with maintaining HSCs self-renewal and stemness properties after long-term expansion. Different research groups have developed very promising and diverse CB-derived HSC expansion strategies using nanofiber scaffolds. Here we review the state-of-the-art of nanofiber technology-based CB-derived HSC expansion.
Collapse
Affiliation(s)
- Mónica Sofia Ventura Ferreira
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Mousavi SH, Abroun S, Soleimani M, Mowla SJ. 3-Dimensional nano-fibre scaffold for ex vivo expansion of cord blood haematopoietic stem cells. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2018; 46:740-748. [PMID: 28685587 DOI: 10.1080/21691401.2017.1337026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/24/2017] [Accepted: 05/29/2017] [Indexed: 12/31/2022]
Abstract
Cord blood (CB) haematopoietic stem cell (HSC) is an alternative source of HSC transplantation. The limited cell number greatly restricts their clinic-scale therapeutic applications. The objective of this study was an ex vivo expansion of CB HSCs in a new three-dimensional polycaprolactone nano-scaffold coated with fibronectin (FN). First, we isolated CB CD34+ cells and cultured 10 days in presence of growth factors. The evaluation was performed by qRT-PCR, flow cytometry and clonogenicity. 3D PCL nano-scaffold coated with FN produced significantly higher total nucleated cells and CD34+ cells (p < .05) and also had significantly higher homing and self-renewality genes than 2D cell culture and before expansion (p < .05). The expression of CXCR-4, VLA-4, VLA-5 and LFA-1, and also HOXB-4, HOXA-9, BMI-1 and hTERT genes was higher in 3D than 2D. The CD13, CD14, CD33, CD34 and CD45 markers were significantly higher and CD2, CD3 and CD19 markers were significantly lower in 3D scaffold than 2D cell culture (p < .05). The type and number of colonies in 2D culture were lower than 3D culture medium (p > .05). 3D PCL nano-scaffold coated with FN could better keep specifications homing and self renewality of CB HSCs after expansion.
Collapse
Affiliation(s)
- Seyed Hadi Mousavi
- a Department of Hematology, Faculty of Allied Medical Sciences , Tehran University of Medical Sciences , Tehran , Iran
| | - Saeid Abroun
- b Department of Hematology, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| | - Masoud Soleimani
- b Department of Hematology, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| | - Seyed Javad Mowla
- c Department of Molecular Genetics, Faculty of Biological Sciences , Tarbiat Modares University , Tehran , Iran
| |
Collapse
|
17
|
Zhao K, Zheng WW, Dong XM, Yin RH, Gao R, Li X, Liu JF, Zhan YQ, Yu M, Chen H, Ge CH, Ning HM, Yang XM, Li CY. EDAG promotes the expansion and survival of human CD34+ cells. PLoS One 2018; 13:e0190794. [PMID: 29324880 PMCID: PMC5764277 DOI: 10.1371/journal.pone.0190794] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/20/2017] [Indexed: 01/17/2023] Open
Abstract
EDAG is multifunctional transcriptional regulator primarily expressed in the linloc-kit+Sca-1+ hematopoietic stem cells (HSC) and CD34+ progenitor cells. Previous studies indicate that EDAG is required for maintaining hematopoietic lineage commitment balance. Here using ex vivo culture and HSC transplantation models, we report that EDAG enhances the proliferative potential of human cord blood CD34+ cells, increases survival, prevents cell apoptosis and promotes their repopulating capacity. Moreover, EDAG overexpression induces rapid entry of CD34+ cells into the cell cycle. Gene expression profile analysis indicate that EDAG knockdown leads to down-regulation of various positive cell cycle regulators including cyclin A, B, D, and E. Together these data provides novel insights into EDAG in regulation of expansion and survival of human hematopoietic stem/progenitor cells.
Collapse
Affiliation(s)
- Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wei-Wei Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Ming Dong
- Tianjin University, School of Chemical Engineering and Technology, Department of Pharmaceutical Engineering, Tianjin, China
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Rui Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiu Li
- An Hui Medical University, Hefei, China
| | - Jin-Fang Liu
- Guang Dong Pharmaceutical University, School of Pharmacy, Guangzhou, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Hui Ge
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hong-Mei Ning
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital to Academy of Military Medical Sciences, Beijing, China
- * E-mail: (HMN); (XMY); (CYL)
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- Tianjin University, School of Chemical Engineering and Technology, Department of Pharmaceutical Engineering, Tianjin, China
- * E-mail: (HMN); (XMY); (CYL)
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- Guang Dong Pharmaceutical University, School of Pharmacy, Guangzhou, China
- * E-mail: (HMN); (XMY); (CYL)
| |
Collapse
|
18
|
Chang HC. The role of policies and networks in development of cord blood usage in China. Regen Med 2017; 12:637-645. [DOI: 10.2217/rme-2017-0050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Research regarding the use of cord blood (CB) has focused on antigen match and the number of stem cells, with policies and networks related to its use being under researched. This article is based on fieldwork in China from 2013 to 2015 and examines ways that the studied CB bank enhances CB usage in China. This article identifies that in addition to finding a match, CB use is linked to the policies and networks, release fee and public awareness that enable CB usage development.
Collapse
Affiliation(s)
- Hung-Chieh Chang
- Institute of Health Policy & Management, National Taiwan University No. 17, Xu-Zhou Road, Taipei 100, Taiwan
| |
Collapse
|
19
|
Shen B, Zhang Y, Dai W, Ma Y, Jiang Y. Ex-vivo expansion of nonhuman primate CD34 + cells by stem cell factor Sall4B. Stem Cell Res Ther 2016; 7:152. [PMID: 27765075 PMCID: PMC5072326 DOI: 10.1186/s13287-016-0413-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Hematopoietic CD34+ stem cells are widely used in the clinical therapy of complicated blood diseases. Stem cell factor Sall4B is a zinc finger transcription factor that plays a vital role in hematopoietic stem cell expansion. The purpose of our current study is to further evaluate how Sall4B might affect the expansion of CD34+ cells derived from nonhuman primates. METHODS Sall4B was overexpressed in nonhuman primate bone marrow-derived CD34+ cells via a lentiviral transduction system. The granulocyte-erythrocyte-macrophage-megakaryocyte colony-forming unit (CFU) assay evaluated the differentiation potential of primate CD34+ cells that were expanded with Sall4B. Furthermore, an in-vivo murine system was employed to evaluate the hematopoietic potential of primate Sall4B-expanded CD34+ cells. RESULTS Overexpression of Sall4B promoted ex-vivo nonhuman primate CD34+ cell expansion by 9.21 ± 1.94-fold on day 9, whereas lentiviral transduction without Sall4B expanded cells by only 2.95 ± 0.77-fold. Sall4B maintained a significant percentage of CD34+ cells as well. The CFU assay showed that the Sall4B-expanded CD34+ cells still possessed multilineage differentiation potential. A study using nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice in vivo revealed that Sall4B led to an increase in the number of repopulating cells and the 9-day-old Sall4B-transduced CD34+ cells still possess self-renewal and multilineage differentiation capacity in vivo, which are similar stemness characteristics to those in freshly isolated primate bone marrow-derived CD34+ cells. CONCLUSIONS We investigated the expansion of nonhuman primate bone marrow-derived CD34+ cells using the Sall4B lentiviral overexpression approach; our findings provide a new perspective on mechanisms of rapid stem cell proliferation. The utilization of Sall4B to expand CD34+ cells on a large scale through use of suitable model systems would prove helpful towards preclinical trials of autologous transplantation.
Collapse
Affiliation(s)
- Bin Shen
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, 277 Qingqiu Street, Suzhou, 215126 China
| | - Yu Zhang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, 277 Qingqiu Street, Suzhou, 215126 China
| | - Wei Dai
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, 277 Qingqiu Street, Suzhou, 215126 China
- Environmental Medicine, NYU Langone Medical Center, Tuxedo, NY 10987 USA
| | - Yupo Ma
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, 277 Qingqiu Street, Suzhou, 215126 China
- Department of Pathology, BST-9C, The State University of New York at Stony Brook, Stony Brook, NY 11794 USA
| | - Yongping Jiang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, 277 Qingqiu Street, Suzhou, 215126 China
- Biopharmagen Corp, Suzhou, 215126 China
| |
Collapse
|
20
|
IL-10+ regulatory B cells are enriched in cord blood and may protect against cGVHD after cord blood transplantation. Blood 2016; 128:1346-61. [PMID: 27439912 DOI: 10.1182/blood-2016-01-695122] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 07/12/2016] [Indexed: 12/19/2022] Open
Abstract
Cord blood (CB) offers a number of advantages over other sources of hematopoietic stem cells, including a lower rate of chronic graft-versus-host disease (cGVHD) in the presence of increased HLA disparity. Recent research in experimental models of autoimmunity and in patients with autoimmune or alloimmune disorders has identified a functional group of interleukin-10 (IL-10)-producing regulatory B cells (Bregs) that negatively regulate T-cell immune responses. At present, however, there is no consensus on the phenotypic signature of Bregs, and their prevalence and functional characteristics in CB remain unclear. Here, we demonstrate that CB contains an abundance of B cells with immunoregulatory function. Bregs were identified in both the naive and transitional B-cell compartments and suppressed T-cell proliferation and effector function through IL-10 production as well as cell-to-cell contact involving CTLA-4. We further show that the suppressive capacity of CB-derived Bregs can be potentiated through CD40L signaling, suggesting that inflammatory environments may induce their function. Finally, there was robust recovery of IL-10-producing Bregs in patients after CB transplantation, to higher frequencies and absolute numbers than seen in the peripheral blood of healthy donors or in patients before transplant. The reconstituting Bregs showed strong in vitro suppressive activity against allogeneic CD4(+) T cells, but were deficient in patients with cGVHD. Together, these findings identify a rich source of Bregs and suggest a protective role for CB-derived Bregs against cGVHD development in CB recipients. This advance could propel the development of Breg-based strategies to prevent or ameliorate this posttransplant complication.
Collapse
|
21
|
Peberdy L, Young J, Kearney L. Health care professionals' knowledge, attitudes and practices relating to umbilical cord blood banking and donation: an integrative review. BMC Pregnancy Childbirth 2016; 16:81. [PMID: 27094796 PMCID: PMC4837592 DOI: 10.1186/s12884-016-0863-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 04/05/2016] [Indexed: 01/29/2023] Open
Abstract
Background Collection and storage of an infant’s cord blood at birth is an option available to many new parents. Antenatal health care providers have an important role in providing non-biased and evidence based information to expectant parents about cord blood and tissue banking options. The aim of this paper was to identify and review studies of health care professionals’ knowledge, attitudes and practices concerning cord blood banking and the sources by which healthcare professionals obtained their information on this topic. Methods An integrative review was conducted using several electronic databases to identify papers on health care professionals’ knowledge, attitudes and practices pertaining to cord blood banking. The CASP tool was used to determine validity and quality of the studies included in the review. Results The search of the international literature identified nine papers which met review inclusion criteria. The literature review identified that there was little focus placed on antenatal health care professionals’ knowledge of cord blood banking options despite these health care professionals being identified by expectant parents as their preferred, key source of information. Conclusion Limited high quality studies have investigated what health care professionals know and communicate to expectant parents regarding cord blood banking. Further research should focus on understanding the knowledge, attitudes and practices of healthcare professionals and how they communicate with expectant parents about this issue. In addition, how this knowledge influences professional practice around birth is also important, as this may positively or negatively impact the information that is provided to expectant parents.
Collapse
Affiliation(s)
- Lisa Peberdy
- University of the Sunshine Coast, Sippy Downs, QLD, Australia.
| | - Jeanine Young
- University of the Sunshine Coast, Sippy Downs, QLD, Australia
| | - Lauren Kearney
- University of the Sunshine Coast, Sippy Downs, QLD, Australia
| |
Collapse
|
22
|
Baron F, Ruggeri A, Nagler A. Methods of ex vivo expansion of human cord blood cells: challenges, successes and clinical implications. Expert Rev Hematol 2016; 9:297-314. [PMID: 26635058 DOI: 10.1586/17474086.2016.1128321] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
More than 40,000 unrelated cord blood transplantations (UCBT) have been performed worldwide as treatment for patients with malignant or non-malignant life threatening hematologic disorders. However, low absolute numbers of hematopoietic stem and progenitor cells (HSPCs) within a single cord blood unit has remained a limiting factor for this transplantation modality, particularly in adult recipients. Further, because UCB contains low numbers of mostly naïve T cells, immune recovery after UCBT is slow, predisposing patients to severe infections. Other causes of UCBT failure has included graft-versus-host disease (GVHD) and relapse of the underlying disease. In this article, we first review the current landscape of cord blood engineering aimed at improving engraftment. This includes approaches of UCB-HSPCs expansion and methods aimed at improving UCB-HSCPs homing. We then discuss recent approaches of cord blood engineering developed to prevent infection [generation of multivirus-specific cytotoxic T cells (VSTs) from UCB], relapse [transduction of UCB-T cells with tumor-specific chimeric receptor antigens (CARs)] and GVHD (expansion of regulatory T cells from UCB). Although many of these techniques of UCB engineering remain currently technically challenging and expensive, they are likely to revolutionize the field of UCBT in the next decades.
Collapse
Affiliation(s)
- Frédéric Baron
- a Division of Hematology, Department of Medicine , University and CHU of Liège , Liège , Belgium.,b GIGA-I3, Section of Hematology , University of Liège , Liège , Belgium
| | - Annalisa Ruggeri
- c Eurocord Hospital Saint Louis, AP-HP , Paris , France.,d Hospital Saint Antoine , Service d'Hématologie et Thérapie Cellulaire, AP-HP , Paris , France.,e Cord Blood Committee, Cellular Therapy and Immunobiology Working Party , EBMT , Leiden , Netherlands
| | - Arnon Nagler
- f Division of Hematology and Bone Marrow Transplantation , The Chaim Sheba Medical Center, Tel-Hashomer , Ramat-Gan , Israel.,g EBMT Paris Office , Hospital Saint Antoine , Paris , France.,h Université Pierre et Marie Curie , Paris , France.,i Tel Aviv University (TAU) , Tel Aviv , Israel
| |
Collapse
|
23
|
Bari S, Chu PPY, Lim A, Fan X, Bunte RM, Li S, Ghosh S, Chiu GNC, Hwang WYK. Mitochondrial superoxide reduction and cytokine secretion skewing by carbon nanotube scaffolds enhance ex vivo expansion of human cord blood hematopoietic progenitors. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1643-56. [DOI: 10.1016/j.nano.2015.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 05/27/2015] [Accepted: 06/06/2015] [Indexed: 01/25/2023]
|
24
|
Oubari F, Amirizade N, Mohammadpour H, Nakhlestani M, Zarif MN. The Important Role of FLT3-L in Ex Vivo Expansion of Hematopoietic Stem Cells following Co-Culture with Mesenchymal Stem Cells. CELL JOURNAL 2015. [PMID: 26199899 PMCID: PMC4503834 DOI: 10.22074/cellj.2016.3715] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Objective Hematopoietic stem cells (HSCs) transplantation using umbilical cord blood
(UCB) has improved during the last decade. Because of cell limitations, several studies focused on the ex vivo expansion of HSCs. Numerous investigations were performed to introduce the best cytokine cocktails for HSC expansion The majority used the Fms-related
tyrosine kinase 3 ligand (FLT3-L) as a critical component. According to FLT3-L biology, in
this study we have investigated the hypothesis that FLT3-L only effectively induces HSCs
expansion in the presence of a mesenchymal stem cell (MSC) feeder.
Materials and Methods In this experimental study, HSCs and MSCs were isolated from
UCB and placenta, respectively. HSCs were cultured in different culture conditions in the
presence and absence of MSC feeder and cytokines. After ten days of culture, total nucleated cell count (TNC), cluster of differentiation 34+(CD34+) cell count, colony forming
unit assay (CFU), long-term culture initiating cell (LTC-IC), homeobox protein B4 (HoxB4)
mRNA and surface CD49d expression were evaluated. The fold increase for some culture
conditions was compared by the t test.
Results HSCs expanded in the presence of cytokines and MSCs feeder. The rate of expansion in the co-culture condition was two-fold more than culture with cytokines (P<0.05).
FLT3-L could expand HSCs in the co-culture condition at a level of 20-fold equal to the
presence of stem cell factor (SCF), thrombopoietin (TPO) and FLT3-L without feeder cells.
The number of extracted colonies from LTC-IC and CD49d expression compared with a
cytokine cocktail condition meaningfully increased (P<0.05).
Conclusion FLT3-L co-culture with MSCs can induce high yield expansion of HSCs and
be a substitute for the universal cocktail of SCF, TPO and FLT3-L in feeder-free culture.
Collapse
Affiliation(s)
- Farhad Oubari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran ; Faulty of Paramedics, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Naser Amirizade
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Hemn Mohammadpour
- Department of Medical Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Mozhdeh Nakhlestani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
25
|
Peters EB, Liu B, Christoforou N, West JL, Truskey GA. Umbilical Cord Blood-Derived Mononuclear Cells Exhibit Pericyte-Like Phenotype and Support Network Formation of Endothelial Progenitor Cells In Vitro. Ann Biomed Eng 2015; 43:2552-68. [PMID: 25777295 DOI: 10.1007/s10439-015-1301-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/11/2015] [Indexed: 01/17/2023]
Abstract
Umbilical cord blood represents a promising cell source for pro-angiogenic therapies. The present study examined the potential of mononuclear cells (MNCs) from umbilical cord blood to support endothelial progenitor cell (EPC) microvessel formation. MNCs were isolated from the cord blood of 20 separate donors and selected for further characterization based upon their proliferation potential and morphological resemblance to human vascular pericytes (HVPs). MNCs were screened for their ability to support EPC network formation using an in vitro assay (Matrigel™) as well as a reductionist, coculture system consisting of no additional angiogenic cytokines beyond those present in serum. In less than 15% of the isolations, we identified a population of highly proliferative MNCs that phenotypically resembled HVPs as assessed by expression of PDGFR-β, NG2, α-SMA, and ephrin-B2. Within a Matrigel™ system, MNCs demonstrated pericyte-like function through colocalization to EPC networks and similar effects as HVPs upon total EPC tubule length (p = 0.95) and number of branch points (p = 0.93). In a reductionist coculture system, MNCs served as pro-angiogenic mural cells by supporting EPC network formation to a significantly greater extent than HVP cocultures, by day 14 of coculture, as evidenced through EPC total tubule length (p < 0.0001) and number of branch points (p < 0.0001). Our findings are significant as we demonstrate mural cell progenitors can be isolated from umbilical cord blood and develop culture conditions to support their use in microvascular tissue engineering applications.
Collapse
|
26
|
Fan HC, Ho LI, Chi CS, Cheng SN, Juan CJ, Chiang KL, Lin SZ, Harn HJ. Current proceedings of cerebral palsy. Cell Transplant 2015; 24:471-85. [PMID: 25706819 DOI: 10.3727/096368915x686931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cerebral palsy (CP) is a complicated disease with varying causes and outcomes. It has created significant burden to both affected families and societies, not to mention the quality of life of the patients themselves. There is no cure for the disease; therefore, development of effective therapeutic strategies is in great demand. Recent advances in regenerative medicine suggest that the transplantation of stem cells, including embryonic stem cells, neural stem cells, bone marrow mesenchymal stem cells, induced pluripotent stem cells, umbilical cord blood cells, and human embryonic germ cells, focusing on the root of the problem, may provide the possibility of developing a complete cure in treating CP. However, safety is the first factor to be considered because some stem cells may cause tumorigenesis. Additionally, more preclinical and clinical studies are needed to determine the type of cells, route of delivery, cell dose, timing of transplantation, and combinatorial strategies to achieve an optimal outcome.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chaurasia P, Gajzer DC, Schaniel C, D'Souza S, Hoffman R. Epigenetic reprogramming induces the expansion of cord blood stem cells. J Clin Invest 2014; 124:2378-95. [PMID: 24762436 DOI: 10.1172/jci70313] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cord blood (CB) cells that express CD34 have extensive hematopoietic capacity and rapidly divide ex vivo in the presence of cytokine combinations; however, many of these CB CD34+ cells lose their marrow-repopulating potential. To overcome this decline in function, we treated dividing CB CD34+ cells ex vivo with several histone deacetylase inhibitors (HDACIs). Treatment of CB CD34+ cells with the most active HDACI, valproic acid (VPA), following an initial 16-hour cytokine priming, increased the number of multipotent cells (CD34+CD90+) generated; however, the degree of expansion was substantially greater in the presence of both VPA and cytokines for a full 7 days. Treated CD34+ cells were characterized based on the upregulation of pluripotency genes, increased aldehyde dehydrogenase activity, and enhanced expression of CD90, c-Kit (CD117), integrin α6 (CD49f), and CXCR4 (CD184). Furthermore, siRNA-mediated inhibition of pluripotency gene expression reduced the generation of CD34+CD90+ cells by 89%. Compared with CB CD34+ cells, VPA-treated CD34+ cells produced a greater number of SCID-repopulating cells and established multilineage hematopoiesis in primary and secondary immune-deficient recipient mice. These data indicate that dividing CB CD34+ cells can be epigenetically reprogrammed by treatment with VPA so as to generate greater numbers of functional CB stem cells for use as transplantation grafts.
Collapse
|
28
|
Roth JA, Bensink ME, O’Donnell PV, Fuchs EJ, Eapen M, Ramsey SD. Design of a cost-effectiveness analysis alongside a randomized trial of transplantation using umbilical cord blood versus HLA-haploidentical related bone marrow in advanced hematologic cancer. J Comp Eff Res 2014; 3:135-44. [PMID: 24645687 PMCID: PMC4036637 DOI: 10.2217/cer.13.95] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND BMT CTN 1101 is a Phase III randomized controlled trial evaluating the comparative effectiveness of double unrelated umbilical cord blood (dUCB) versus HLA-haploidentical related donor bone marrow (haplo-BM) donor cell sources for blood or bone marrow transplantation (BMT) in patients with hematologic malignancies. Herein, we present the rationale, design and methods of the first cost-effectiveness analysis to be conducted alongside a BMT trial. METHODS Consenting patients will provide health insurance information to allow calculation of direct medical costs from reimbursement records, and will provide out-of-pocket costs, time costs and health-related quality of life measures through an online survey. These outcomes will inform a cost-effectiveness analysis comparing dUCB and haplo-BM donor cell sources from patient, payer and societal perspectives. CONCLUSION Novel approaches may significantly change the cost, outcomes or availability of BMT. The results of this analysis will be the first to provide a comprehensive evaluation of the comparative effectiveness of these approaches from multiple perspectives.
Collapse
Affiliation(s)
- Joshua A Roth
- Public Health Sciences Division, Fred Hutchinson Cancer Research Centre, 1100 Fairview Avenue North, Seattle, WA 98109, USA
- Group Health Research Institute, Group Health Cooperative, 1730 Minor Avenue, Seattle, WA 98101, USA
| | - Mark E Bensink
- Public Health Sciences Division, Fred Hutchinson Cancer Research Centre, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Paul V O’Donnell
- Clinical Research Division, Fred Hutchinson Cancer Research Centre, 1100 Fairview Avenue North, Seattle, WA 98109, USA
- Department of Medicine, University of Washington, WA, USA
| | - Ephraim J Fuchs
- John Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD 21287, USA
| | - Mary Eapen
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Scott D Ramsey
- Public Health Sciences Division, Fred Hutchinson Cancer Research Centre, 1100 Fairview Avenue North, Seattle, WA 98109, USA
- Department of Medicine, University of Washington, WA, USA
| |
Collapse
|
29
|
Abstract
β-thalassemia is an inherited disorder due to mutations found in the β-globin gene, leading to anemia and requiring sporadic or chronic blood transfusions for survival. Without proper chelation, β-thalassemia results in iron overload. Ineffective erythropoiesis can lead to iron overload even in untransfused patients who are affected by β-thalassemia intermedia. Better understanding of the molecular biologic aspects of this disorder has led to improvements in population screening and prenatal diagnosis, which, in turn, have led to dramatic reductions in the number of children born with β-thalassemia major in the Mediterranean littoral. However, as a consequence of decreases in neonatal and childhood mortality in other geographical areas, β-thalassemia has become a worldwide clinical problem. A number of unsolved pathophysiological issues remain, such as ineffective erythropoieis, abnormal iron absorption, oxidative stress, splenomegaly and thrombosis. In the last few years, novel studies have the potential to introduce new therapeutic approaches that might reduce these problems and limit the need for blood transfusion.
Collapse
Affiliation(s)
- Stefano Rivella
- Weill College Medical Center, Department of Pediatrics, Division of Hematology, Oncology, 515 E 71st Street, S702, New York, NY 10021, USA, Tel.: +1 212 746 4941, ,
| | | |
Collapse
|
30
|
Abstract
A now 10-year-old Laotian female was delivered at 30-week gestation by cesarean section because of severe hydrops. Fetal blood sampling revealed homozygous α-thalassemia. After immediate resuscitation, the infant was supported with frequent red cell transfusions. At 44 months of age, she received a 5 of 6 human leukocyte antigen-matched unrelated cord blood transplantation. She was treated with phlebotomy and chelation therapy with Deferasirox for correction of hemosiderosis and has been transfusion-independent since 41 days after transplant. She is currently 6 years after transplantation with stable, 100% donor engraftment, resolved iron overload, and normal growth and development.
Collapse
|
31
|
Wang L, Lu M. Regulation and direction of umbilical cord blood mesenchymal stem cells to adopt neuronal fate. Int J Neurosci 2013; 124:149-59. [PMID: 23879374 DOI: 10.3109/00207454.2013.828055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Umbilical cord blood mesenchymal stem cells (UCB-MSCs) transplantation is becoming a promising and attractive cell-based treatment modality for repairing the damaged central nervous system due to its advantages of low immunogenicity, wide range of sources, and less ethical controversy. One of the limitations of this approach is that the proportion of neurons differentiated from UCB-MSCs still remains at low level. Thus, to induce UCB-MSCs to differentiate into neuron-like cells with a higher proportion is one of the key technologies of regenerative medicine and tissue engineering. Many induction protocols with remarkably higher differentiation rate to neurons have been reported. However, each protocol has its pros and cons and whether the neurons differentiated from UCB-MSCs under a certain protocol has normal nerve function remains controversial. Therefore, to guarantee the success of future clinical applications of UCB-MSCs, more investigations should be performed to improve the induction method and differentiation efficiency.
Collapse
Affiliation(s)
- Lei Wang
- 1Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA) , Changsha, Hunan , China
| | | |
Collapse
|
32
|
Soltanpour MS, Amirizadeh N, Zaker F, Oodi A, Nikougoftar M, Kazemi A. mRNA expression and promoter DNA methylation status of CDKi p21 and p57 genes inex vivoexpanded CD34+cells following co-culture with mesenchymal stromal cells and growth factors. Hematology 2013; 18:30-8. [DOI: 10.1179/1607845412y.0000000030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Mohammad Soleiman Soltanpour
- Department of Hematology and Blood BankingSchool of Allied Medical Sciences, Tehran University of Medical Science, Tehran, Iran
| | - Naser Amirizadeh
- High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research Center, Tehran, Iran
| | - Farhad Zaker
- Department of Hematology and Blood BankingSchool of Allied Medical Sciences, Tehran University of Medical Science, Tehran, Iran
| | - Arezoo Oodi
- High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research Center, Tehran, Iran
| | - Mahin Nikougoftar
- High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research Center, Tehran, Iran
| | - Ahmad Kazemi
- Department of Hematology and Blood BankingSchool of Allied Medical Sciences, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
33
|
Magnusson M, Sierra MI, Sasidharan R, Prashad SL, Romero M, Saarikoski P, Van Handel B, Huang A, Li X, Mikkola HKA. Expansion on stromal cells preserves the undifferentiated state of human hematopoietic stem cells despite compromised reconstitution ability. PLoS One 2013; 8:e53912. [PMID: 23342037 PMCID: PMC3547050 DOI: 10.1371/journal.pone.0053912] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/04/2012] [Indexed: 12/15/2022] Open
Abstract
Lack of HLA-matched hematopoietic stem cells (HSC) limits the number of patients with life-threatening blood disorders that can be treated by HSC transplantation. So far, insufficient understanding of the regulatory mechanisms governing human HSC has precluded the development of effective protocols for culturing HSC for therapeutic use and molecular studies. We defined a culture system using OP9M2 mesenchymal stem cell (MSC) stroma that protects human hematopoietic stem/progenitor cells (HSPC) from differentiation and apoptosis. In addition, it facilitates a dramatic expansion of multipotent progenitors that retain the immunophenotype (CD34+CD38-CD90+) characteristic of human HSPC and proliferative potential over several weeks in culture. In contrast, transplantable HSC could be maintained, but not significantly expanded, during 2-week culture. Temporal analysis of the transcriptome of the ex vivo expanded CD34+CD38-CD90+ cells documented remarkable stability of most transcriptional regulators known to govern the undifferentiated HSC state. Nevertheless, it revealed dynamic fluctuations in transcriptional programs that associate with HSC behavior and may compromise HSC function, such as dysregulation of PBX1 regulated genetic networks. This culture system serves now as a platform for modeling human multilineage hematopoietic stem/progenitor cell hierarchy and studying the complex regulation of HSC identity and function required for successful ex vivo expansion of transplantable HSC.
Collapse
Affiliation(s)
- Mattias Magnusson
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maria I. Sierra
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rajkumar Sasidharan
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Sacha L. Prashad
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Melissa Romero
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Pamela Saarikoski
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Ben Van Handel
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Andy Huang
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hanna K. A. Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
34
|
Flores-Guzman P, Fernandez-Sanchez V, Valencia-Plata I, Arriaga-Pizano L, Alarcon-Santos G, Mayani H. Comparative in vitro analysis of different hematopoietic cell populations from human cord blood: in search of the best option for clinically oriented ex vivo cell expansion. Transfusion 2012; 53:668-78. [PMID: 22845003 DOI: 10.1111/j.1537-2995.2012.03799.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Ex vivo expansion of hematopoietic stem and progenitor cells has become a priority in the experimental hematology arena. In this study we have obtained different hematopoietic cell populations from umbilical cord blood and simultaneously assessed their proliferation and expansion kinetics. Our main goal was to determine which one of these cell populations would be more suitable for clinical-grade ex vivo expansion. STUDY DESIGN AND METHODS By using immunomagnetic-negative selection and cell sorting, five cell populations were obtained: unseparated mononuclear cells (MNCs; I); two lineage-negative cell populations, one enriched for CD34+ CD38+ cells (II) and the other enriched for CD34+ CD38- cells (III); and two CD34+ cell fractions purified by fluorescence-activated cell sorting, one containing CD34+ CD38+ cells (IV) and the other containing CD34+ CD38- cells (V). The kinetics of such populations were analyzed in both relative and absolute terms. RESULTS No expansion was observed in Population I; in contrast, significant increments in the numbers of both progenitor and stem cells were observed in cultures of Populations II to V. Population V (reaching 12,800-fold increase in total cells; 1280-fold increase in CD34+ cells; 490-fold increase in colony-forming cells; and 12-fold increase in long-term culture-initiating cells) showed the highest proliferation and expansion potentials. CONCLUSION Our study suggests that the cell fraction containing greater than 98% CD34+ CD38- cells would be the ideal one for large-scale ex vivo expansion; however, based on our data, it seems that, except for MNCs, all other cell populations could also be used as input cell fractions.
Collapse
Affiliation(s)
- Patricia Flores-Guzman
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|
35
|
Ponce-Regalado MD, Ortuño-Sahagún D, Zarate CB, Gudiño-Cabrera G. Ensheathing cell-conditioned medium directs the differentiation of human umbilical cord blood cells into aldynoglial phenotype cells. Hum Cell 2012; 25:51-60. [PMID: 22529032 DOI: 10.1007/s13577-012-0044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 02/24/2012] [Indexed: 10/28/2022]
Abstract
Despite their similarities to bone marrow precursor cells (PC), human umbilical cord blood (HUCB) PCs are more immature and, thus, they exhibit greater plasticity. This plasticity is evident by their ability to proliferate and spontaneously differentiate into almost any cell type, depending on their environment. Moreover, HUCB-PCs yield an accessible cell population that can be grown in culture and differentiated into glial, neuronal and other cell phenotypes. HUCB-PCs offer many potential therapeutic benefits, particularly in the area of neural replacement. We sought to induce the differentiation of HUCB-PCs into glial cells, known as aldynoglia. These cells can promote neuronal regeneration after lesion and they can be transplanted into areas affected by several pathologies, which represents an important therapeutic strategy to treat central nervous system damage. To induce differentiation to the aldynoglia phenotype, HUCB-PCs were exposed to different culture media. Mononuclear cells from HUCB were isolated and purified by identification of CD34 and CD133 antigens, and after 12 days in culture, differentiation of CD34+ HUCB-PCs to an aldynoglia phenotypic, but not that of CD133+ cells, was induced in ensheathing cell (EC)-conditioned medium. Thus, we demonstrate that the differentiation of HUCB-PCs into aldynoglia cells in EC-conditioned medium can provide a new source of aldynoglial cells for use in transplants to treat injuries or neurodegenerative diseases.
Collapse
Affiliation(s)
- María Dolores Ponce-Regalado
- Laboratorio de Desarrollo y Regeneración Neural, Departamento de Biología Celular y Molecular, Instituto de Neurobiología, C.U.C.B.A, Universidad de Guadalajara, Apdo. Postal 52-126, 45021, Guadalajara, Jalisco, Mexico
| | | | | | | |
Collapse
|
36
|
|
37
|
Umbilical cord blood: lessons learned and lingering challenges after more than 20 years of basic and clinical research. Arch Med Res 2011; 42:645-51. [PMID: 22154618 DOI: 10.1016/j.arcmed.2011.11.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 11/16/2011] [Indexed: 12/17/2022]
Abstract
During the last 23 years, cord blood research has played important roles both in experimental and clinical hematology. Cord blood-derived hematopoietic stem and progenitor cells have been shown to possess particular biological features and their study has been very important in our understanding of hematopoietic development. Today, >20,000 umbilical cord blood (UCB) transplants have been performed worldwide and ∼460,000 UCB units are being stored in >47 UCB banks worldwide. Here a brief overview on some of the most relevant issues regarding cord blood research is presented.
Collapse
|
38
|
Marquez-Curtis LA, Turner AR, Sridharan S, Ratajczak MZ, Janowska-Wieczorek A. The ins and outs of hematopoietic stem cells: studies to improve transplantation outcomes. Stem Cell Rev Rep 2011; 7:590-607. [PMID: 21140298 DOI: 10.1007/s12015-010-9212-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Deciphering the mechanisms of hematopoietic stem/progenitor cell (HSPC) mobilization and homing is important for the development of strategies to enhance the efficacy of HSPC transplantation and achieve the full potential of HSPC-based cellular therapy. Investigation of these mechanisms has revealed interdependence among the various molecules, pathways and cellular components involved, and underscored the complex nature of these two processes. This review summarizes recent progress in identifying the specific factors implicated in HSPC mobilization and homing, with emphasis on our own work. Particularly, we will discuss our studies on stromal cell-derived factor-1 and its interaction with its receptor CXCR4, proteases (matrix metalloproteinases and carboxypeptidase M), complement proteins (C1q, C3a, C5a, membrane attack complex), sphingosine-1-phosphate, and pharmacologic agents such as the histone deacetylase inhibitor valproic acid and hyaluronic acid.
Collapse
Affiliation(s)
- Leah A Marquez-Curtis
- Research & Development, Canadian Blood Services, CBS Edmonton Centre, 8249-114 St. NW, Edmonton, T6G 2R8, Alberta, Canada
| | | | | | | | | |
Collapse
|
39
|
|
40
|
Goldstein G, Elhasid R, Bielorai B, Shimoni A, Yerushalmi R, Kassis I, Nagler A. Adults requiring cord blood transplants but have insufficient cell doses from a single cord blood unit can receive two units with successful engraftment kinetics similar to those of children receiving a single unit. Leuk Lymphoma 2011; 52:635-41. [PMID: 21338286 DOI: 10.3109/10428194.2010.548109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We retrospectively evaluated neutrophil engraftment kinetics in 29 single versus nine double unit cord blood transplants (CBTs). All single CBTs were performed in pediatric patients (non-malignant/malignant diseases, 19/10), while all double CBTs were performed in adults (n = 8) and an adolescent (n = 1) with hematological malignancies. Median follow-up time was 2.3 years (range, 0.1-13.5 years). Engraftment was achieved in 69% and 89% of the single and double cord blood (CB) groups, respectively. Similarly, median day of engraftment was not different for the single versus the double CBTs, at 19 and 23 days, respectively, and the neutrophil engraftment kinetics was similar in the two groups. Our data indicate that adults without sufficient nucleated cell doses in a single CB unit may receive two units with similar engraftment kinetics to those of children receiving only a single unit.
Collapse
Affiliation(s)
- Gal Goldstein
- Pediatric Hemato-oncology and Bone Marrow Transplantation Department, The Edmond and Lily Safra Children's Hospital, Tel Hashomer, Israel.
| | | | | | | | | | | | | |
Collapse
|
41
|
Arien-Zakay H, Lazarovici P, Nagler A. Tissue regeneration potential in human umbilical cord blood. Best Pract Res Clin Haematol 2011; 23:291-303. [PMID: 20837341 DOI: 10.1016/j.beha.2010.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative medicine is the process of creating functional tissue with the aid of stem cells, to repair loss of organ function. Possible targets for regenerative medicine include orthopaedic, cardiac, hepatic, pancreatic and central nervous system (CNS) applications. Umbilical cord blood (CB) has established itself as a legitimate source for haematopoietic stem cell transplantation. It is also considered an accessible and less immunogenic source for mesenchymal, unrestricted somatic and for other stem cells with pluri/multipotent properties. The latter are capable of differentiating into a wide variety of cell types including bone, cartilage, cardiomyocytes and neural. They also possess protective abilities that may contribute to tissue repair even if in vitro differentiation is excluded. In view of the absence of treatment for many devastating diseases, the elucidation of non-haematopoietic applications for CB will facilitate the development of pioneering relevant cell therapy approaches. This review focusses on current studies using human CB-derived cells for regenerative medicine.
Collapse
|
42
|
Long HM, Taylor GS, Rickinson AB. Immune defence against EBV and EBV-associated disease. Curr Opin Immunol 2011; 23:258-64. [PMID: 21269819 DOI: 10.1016/j.coi.2010.12.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 12/23/2010] [Indexed: 10/18/2022]
Abstract
Epstein-Barr virus (EBV), a B-lymphotropic herpesvirus widespread in the human population and normally contained as an asymptomatic infection by T cell surveillance, nevertheless causes infectious mononucleosis and is strongly linked to several types of human cancer. Here we describe new findings on the range of cellular immune responses induced by EBV infection, on viral strategies to evade those responses and on the links between HLA gene loci and EBV-induced disease. The success of adoptive T cell therapy for EBV-driven post-transplant lymphoproliferative disease is stimulating efforts to target other EBV-associated tumours by immunotherapeutic means, and has reawakened interest in the ultimate intervention strategy, a prophylactic EBV vaccine.
Collapse
Affiliation(s)
- Heather M Long
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | |
Collapse
|
43
|
Abstract
AbstractThe young human brain is highly plastic and thus early brain lesions can lead to aberrant development of connectivity and mapping of functions. This is why initially in cerebral palsy only subtle changes in spontaneous movements are seen after the time of lesion, followed by a progressive evolution of a movement disorder over many months and years. Thus we propose that interventions to treat cerebral palsy should be initiated as soon as possible in order to restore the nervous system to the correct developmental trajectory. One such treatment might be autologous stem cell transplantation either intracerebrally or intravenously. All babies come with an accessible supply of stem cells, the umbilical cord, which can supply cells that could theoretically replace missing neural cell types, or act indirectly by supplying trophic support or modulating inflammatory responses to hypoxia/ischaemia. However, for such radical treatment to be proposed, it is necessary to be able to detect and accurately predict the outcomes of brain injury from a very early age. This article reviews our current understanding of perinatal injuries that lead to cerebral palsy, how well modern imaging might predict outcomes, what stem cells are yielded from umbilical cord blood and experimental models of brain repair using stem cells.
Collapse
|
44
|
Griffith ML, Jagasia M, Jagasia SM. Diabetes mellitus after hematopoietic stem cell transplantation. Endocr Pract 2010; 16:699-706. [PMID: 20439241 DOI: 10.4158/ep10027.ra] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To review the current literature on posttransplant diabetes mellitus after hematopoietic stem cell transplantation, including its epidemiologic features, transplant-related risk factors, and treatment. METHODS A literature search was conducted in PubMed for articles on diabetes mellitus after hematopoietic stem cell transplantation and effects of immunosuppressants on glucose metabolism. RESULTS Within 2 years after hematopoietic stem cell transplantation, up to 30% of patients may have diabetes. Although some of these cases resolve, the rates of diabetes and metabolic syndrome remain elevated in comparison with those in the nontransplant patient population during long-term follow-up. Traditional risk factors for diabetes as well as features related to the transplantation process, including immunosuppressive medications, are associated with posttransplant diabetes. Cardiovascular risk also appears to be increased in this population. Limited data are available on hypoglycemic agents for posttransplant diabetes; thus, treatment decisions must be based on safety, efficacy, and tolerability, with consideration of each patient's transplant-related medications and comorbidities. CONCLUSION Treatment of diabetes mellitus in patients who have undergone hematopoietic stem cell transplantation necessitates attention to the posttransplant medication regimen and clinical course. Although no guidelines specific to treatment of posttransplant diabetes in this patient population currently exist, treatment to goals similar to those for nontransplant patients with diabetes should be considered in an attempt to help reduce long-term morbidity and mortality.
Collapse
Affiliation(s)
- Michelle L Griffith
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8148, USA
| | | | | |
Collapse
|
45
|
|
46
|
Arien-Zakay H, Lecht S, Nagler A, Lazarovici P. Human umbilical cord blood stem cells: rational for use as a neuroprotectant in ischemic brain disease. Int J Mol Sci 2010; 11:3513-28. [PMID: 20957109 PMCID: PMC2956109 DOI: 10.3390/ijms11093513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 09/15/2010] [Accepted: 09/15/2010] [Indexed: 01/19/2023] Open
Abstract
The use of stem cells for reparative medicine was first proposed more than three decades ago. Hematopoietic stem cells from bone marrow, peripheral blood and human umbilical cord blood (CB) have gained major use for treatment of hematological indications. CB, however, is also a source of cells capable of differentiating into various non-hematopoietic cell types, including neural cells. Several animal model reports have shown that CB cells may be used for treatment of neurological injuries. This review summarizes the information available on the origin of CB-derived neuronal cells and the mechanisms proposed to explain their action. The potential use of stem/progenitor cells for treatment of ischemic brain injuries is discussed. Issues that remain to be resolved at the present stage of preclinical trials are addressed.
Collapse
Affiliation(s)
- Hadar Arien-Zakay
- The School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; E-Mails: (H.A.-Z.); (S.L.)
- Division of Hematology and Cord Blood Bank, Chaim Sheba Medical Center, Tel-Hashomer, Israel; E-Mail: (A.N.)
| | - Shimon Lecht
- The School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; E-Mails: (H.A.-Z.); (S.L.)
| | - Arnon Nagler
- Division of Hematology and Cord Blood Bank, Chaim Sheba Medical Center, Tel-Hashomer, Israel; E-Mail: (A.N.)
| | - Philip Lazarovici
- The School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; E-Mails: (H.A.-Z.); (S.L.)
- * Author to whom correspondence should be addressed: E-Mail: ; Tel.: 972-2-6758-729; Fax: 972-2-6757-490
| |
Collapse
|
47
|
Guerra-Marquez A, Novelo-Garza B, Malagón-Martínez A, Limon-Flores A, Luna-Bautista F, Juan-Shum L, Montero-Ponce I, Sanchez-Valle E, Peñaflor K, Vélez-Ruelas MA, Romero-Juárez Y, Mayani H. Cord blood banking and transplantation at the Mexican Institute of Social Security: the first 5 years. Transfusion 2010; 51:328-32. [DOI: 10.1111/j.1537-2995.2010.02875.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
48
|
Tung S, Parmar S, Robinson S, De Lima M, Shpall E. Ex vivo expansion of umbilical cord blood for transplantation. Best Pract Res Clin Haematol 2010; 23:245-57. [DOI: 10.1016/j.beha.2010.06.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Shi CJ, Wang F, Ren MF, Mi YJ, Yan YY, To KKW, Dai CL, Wang YS, Chen LM, Tong XZ, Liang YJ, Fu LW. Up-regulation of ABCB1/P-glycoprotein by escaping promoter hypermethylation indicates poor prognosis in hematologic malignancy patients with and without bone marrow transplantation. Leuk Res 2010; 35:73-9. [PMID: 20488541 DOI: 10.1016/j.leukres.2010.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 04/20/2010] [Accepted: 04/23/2010] [Indexed: 11/26/2022]
Abstract
We investigated the correlation between MDR1 promoter methylation status and MDR1 expression in 228 hematologic malignancies patients and 90 healthy controls. High level of MDR1 mRNA correlated to promoter hypomethylation and strongly associated with poor prognosis indicated by 2-year survival rates, poor CR rate (without BMT) and high relapse rate (with BMT). Furthermore, relative luciferase activity of methylated MDR1 at promoter -50 region was significantly higher than that of the unmethylated. In addition, MDR1 in K562 cells elevated significantly after 5-Aza-dC treatment. In summary, MDR1 promoter hypomethylation conferred its up-regulation and indicated poor prognosis in patients with and without BMT.
Collapse
Affiliation(s)
- Cheng-Jun Shi
- State Key Laboratory of Oncology in Southern China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Factors affecting mortality following myeloablative cord blood transplantation in adults: a pooled analysis of three international registries. Bone Marrow Transplant 2010; 46:70-6. [PMID: 20436518 DOI: 10.1038/bmt.2010.83] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A retrospective analysis was conducted to examine factors affecting early mortality after myeloablative, single-unit cord blood transplantation (CBT) for hematological malignancies in adolescents and adults. Data were collected from the three main CBT registries pooling 514 records of unrelated, single, unmanipulated, first myeloablative allogeneic CBTs conducted in North America or Europe from 1995 to 2005, with an HLA match ≥ 4/6 loci, in patients aged 12-55. Overall 100-day, 180-day and 1-year survival (Kaplan-Meier method) were 56, 46 and 37%, respectively, with no significant heterogeneity across registries. Multivariate analysis showed cell dose < 2.5 × 10⁷/kg (odds ratio (OR) 2.76, P < 0.0001), older age (P = 0.002), advanced disease (P = 0.02), positive CMV sero-status (OR 1.37 P = 0.11), female gender (OR 1.43, P = 0.07) and limited CBT center experience (< 10 records contributed, OR 2.08, P = 0.0003) to be associated with higher 100-day mortality. A multivariate model predictive of 1-year mortality included similar prognostic factors except female gender. Transplant year did not appear as a significant independent predictor. This is the first analysis to pool records from three major CBT registries in the United States and Europe. In spite of some differences in practice patterns, survival was remarkably homogeneous. The resulting model may contribute to better understanding factors affecting CBT outcomes.
Collapse
|