Efficacy of ‘Off-the-Shelf’, Commercially-Available, Third-Party Mesenchymal Stem Cells (MSC) in Ex Vivo Cord Blood (CB) Co-Culture Expansion.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4106-4106
Author(s):  
Simon N. Robinson ◽  
Paul J. Simmons ◽  
Nathalie Brouard ◽  
Shannon Kidd ◽  
Hong Yang ◽  
...  

Abstract INTRODUCTION: Our previous studies have shown that clinically-relevant levels of hematopoietic stem and progenitor cell (HSPC) expansion are possible by ex vivo co-culture of cord blood (CB) mononuclear cells (MNC) with third-party bone marrow (BM)-derived mesenchymal stem cells (MSC) and growth factors.1 A recently activated M. D. Anderson protocol requires that BM from a haplo-identical family member be used for the de novo generation of sufficient MSC for subsequent co-culture, a process requiring ∼3 weeks. Time constraints, uncertainties associated with the identification of a suitable BM donor and potential variation in MSC performance make logistical execution of this strategy difficult. We therefore investigated the potential efficacy of ‘off-the-shelf’ commercially-available sources of MSC. Since MSC do not express HLA-II (DR) they are non-immunogenic, suggesting that this might be a valuable alternative strategy. We compared ex vivo CB HSPC expansion obtained following CB MNC co-culture with 2 commercially-available research-grade MSC isolated by density separation and plastic adherence (MSC#1, Cambrex, Walkersville, MD and MSC#2, Allcells, Emeryville, CA). A third MSC, isolated by Stro-12 selection (MSC#3, supplied by PJS) was also evaluated. METHODS: Two MDACC frozen CB units (CB#1&2) were thawed, washed and co-cultured with adherent monolayers from each MSC. Total nucleated cell (TNC) and HSPC (CD34+ cells and colony-forming units, CFU) numbers were measured at input (Day 0) and output (Day 14). RESULTS: TNC and HSPC numbers revealed that the 2 commercially-available research-grade MSC (MSC#1&2) supported ex vivo CB HSPC expansion. MSC TNC CB34+ CFU n/a - not available CB#1 #1 x 6 x23 n/a #2 x 3 x 8 x15 #3 x 6 x16 x23 CB#2 #1 x 7 x20 x31 #2 x 5 x10 x20 #3 x10 x16 x34 1 Robinson et al. x13 x14 x25 MSC#2 performed less well than MSC#1 for both CB units suggesting that variation may exist between individual MSC. These data suggest that the screening of clinical-grade MSC that perform optimally during ex vivo expansion co-culture might be warranted to best utilize this ‘off-the-shelf’ strategy. Data were similar to previous reports where TNC, CD34+ and CFU numbers were shown to increase approximately 13, 14 and 25-fold, respectively.1 Data were also similar for MSC#3, suggesting that the method used to isolate MSC does not appear to be an important variable for effective CB MNC/MSC co-culture. CONCLUSION: Although research-grade MSC were compared from different commercial sources, these data suggest that, in principle, commercially-available clinical-grade MSC might prove a valuable ‘off-the-shelf’ option, potentially reducing the time to therapy and addressing concerns associated with identifying a BM donor and variation in MSC performance. Future studies will evaluate FDA-compliant MSC that could be used clinically.

Cytotherapy ◽  
2004 ◽  
Vol 6 (4) ◽  
pp. 311-317 ◽  
Author(s):  
I. McNiece ◽  
J. Harrington ◽  
J. Turney ◽  
J. Kellner ◽  
E.J. Shpall

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4810-4810
Author(s):  
Olga Kulemina ◽  
Izida Minullina ◽  
Sergey Anisimov ◽  
Renata Dmitrieva ◽  
Andrey Zaritskey

Abstract Abstract 4810 Ex vivo expansion and manipulation of primitive hematopoietic cells has become a major goal in the experimental hematology, because of its potential relevance in the development of therapeutic strategies aimed at treating a diverse group of hematologic disorders. Osteoblasts, mesenchymal stem/progenitor cells (MSC/MPC), adipocytes, reticular cells, endothelial cells and other stromal cells, have been implicated in regulation of HSC maintenance in endosteal and perivascular niches. These niches facilitate the signaling networks that control the balance between self-renewal and differentiation. In the present study, we evaluated and compared the effects of three different stromal feeder layers on expansion of HSPC derived from BM and cord blood (CB): BM mesenchymal stem cells (MSC), osteoblast-differentiated BM mesenchymal stem cells (Ost-MSC) and adipocyte-differentiated BM mesenchymal stem cells (Ad-MSC). BM-MSC cultures were established from plastic adherent BM cell fractions and analyzed for immunophenotype, frequency of colony forming units (CFU-F), frequency of osteo- (CFU-Ost) and adipo- (CFU-Ad) lineage progenitors. Cultures with similar clonogenity (CFU-F: 26,4 ± 4,5%) and progenitors frequency (CFU-Ost: 14,7 ± 4,5%; CFU-Ad: 13,3 ± 4,5%) were selected for co-culture experiments. All MSC were positive for stromal cell-associated markers (CD105, CD90, CD166, CD73) and negative for hematopoietic lineage cells markers (CD34, CD19, CD14, CD45). CD34+ cells were separared from BM and CB samples by magnetic cell sorting (MACS) and analyzed for CD34, CD38 and CD45 expression. Feeder layers (MSC, Ost-MSC, Ad-MSC) were prepared in 24-well plates prior to co-culture experiments: MSCs (4×104 cells/well) were cultured for 24 h and either used for following experiments or stimulated to differentiate into either osteoblasts or adipoctes according to standard protocols. CD34+ cells (3500-10000 cells per well) were co-cultured in Stem Span media with or without a feeder layers and in the presence of cytokines (10 ng/mL Flt3-L, 10 ng/mL SCF, 10ng/mL IL-7) for 7 days. Expanded cells were analyzed for CD34, CD38 and CD45 expression. Results are shown on figures 1 and 2. As expected, CB-derived HSPC expanded much more effectively than BM-derived HSPC. The similar levels of expansion were observed for both, the total number of HSPC, and more primitive CD34+CD38- fraction in the presence of all three feeder layers. Ost-MSC supported CB-derived HSPC slightly better than MSC and Ad-MSC which is in a good agreement with data from literature (Mishima et.al., European Journal of Haematology, 2010), but difference was not statistically significant. In contrast, whereas BM-MSC feeder facilitated CD34+CD38- fraction in BM-derived HSPC, Adipocyte-differentiated MSC and osteoblast-differentiated MSC failed to support BM-derived CD34+CD38- expansion (11,4 ±.4 folds for MSC vs 0,9 ±.0,14 for Ad-MSC, n=5, p<0,01 and 0,92 ±.0,1 for Ost-MSC, n=5, p<0,01).Figure 1.Cord Blood HSPC ex vivo expansionFigure 1. Cord Blood HSPC ex vivo expansionFigure 2.Bone Marrow HSPC ex vivo expansionFigure 2. Bone Marrow HSPC ex vivo expansion Conclusion: BM- and CB-derived CD34+CD38- cells differ in their dependence of bone marrow stroma. Coctail of growth factors facilitate CB HSPC expansion irrespective of lineage differentiation of supporting MSC feeder layer. In contrast, primitive BM CD34+CD38- HSPC were able to expand only on not differentiated MSC. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 5061-5061
Author(s):  
Xiaoyan Zhang ◽  
Jianyong Li ◽  
Kejiang Cao ◽  
Hanxin Wu ◽  
Hua Lu ◽  
...  

Abstract Background: Mesenchymal stem cells(MSCs) can be isolated from bone-marrow and expanded ex-vivo, may support hematopoietic reconstruction and mitigate graft-versus-host disease (GVHD) in hematopoietic stem cells transplantation (HSCT). We hope to explore the feasibility and safety of cotransplantation culture-expanded MSCs and hematopoietic stem cells (HSCs) from the same human leucocyte antigen (HLA)-identical sibling donor in patients with hematologic diseases. Methods: Bone marrow mononuclear cells from healthy donors were cultured and expanded ex-vivo. Immunophenotype, karyotye, immunosuppressive property of the harvested MSCs were characterized. Patients were cotransplanted with HSCs and MSCs from the same donor. Hematopoietic reconstruction, complications and clinical outcomes after transplantation in these patients were observed. Results: (1.77±0.40)×106/kg (donor’s weight) MSCs were successfully expanded from 23.6±5.96ml bone marrow samples. They had normal karyotype and were CD73, CD90, CD105 positive and CD34, CD45, HLA-DR negative. They can inhibit mixed lymphocyte reactions (MLRs). Twelve patients were undergoing cotransplantation. No adverse response was observed during and after the infusion of allogenic MSCs. Hematopoietic reconstruction were rapid. Two patients developed grade II∼IV acute GVHD. Two patients developed systermatic chronic GVHD. Four patients suffered from cytomegalovirus (CMV) infection but were cured at last. Till now, seven patients have been alive for 29∼57 months and five patients died. Conclusion: MSCs identified by immunophenotype analysis can be isolated from human bone marrow, expanded effectively by culture. Their quality and quantity are suitable for clinical use. It is safe and feasible to cotransplant patients with allogenic culture-expanded MSCs and HSCs.


2007 ◽  
Vol 16 (6) ◽  
pp. 579-585 ◽  
Author(s):  
Guo-Ping Huang ◽  
Zhi-Jun Pan ◽  
Bing-Bing Jia ◽  
Qiang Zheng ◽  
Chun-Gang Xie ◽  
...  

Human mesenchymal stem cells (MSCs) are multipotential and are detected in bone marrow (BM), adipose tissue, placenta, and umbilical cord blood (UCB). In this study, we examined the ability of UCB-derived MSCs (UCB-MSCs) to support ex vivo expansion of hematopoietic stem/progenitor cells (HSPCs) from UCB and the engraftment of expanded HSPCs in NOD/SCID mice. The result showed that UCB-MSCs supported the proliferation and differentiation of CD34+ cells in vitro. The number of expanded total nucleated cells (TNCs) in MSC-based culture was twofold higher than cultures without MSC (control cultures). UCB-MSCs increased the expansion capabilities of CD34+ cells, long-term culture-initiating cells (LTC-ICs), granulocyte-macrophage colony-forming cells (GM-CFCs), and high proliferative potential colony-forming cells (HPP-CFCs) compared to control cultures. The expanded HSPCs were transplanted into lethally irradiated NOD/SCID mice to assess the effects of expanded cells on hematopoietic recovery. The number of white blood cells (WBCs) in the peripheral blood of mice transplanted with expanded cells from both the MSC-based and control cultures returned to pretreatment levels at day 25 posttransplant and then decreased. The WBC levels returned to pretreatment levels again at days 45–55 posttransplant. The level of human CD45+ cell engraftment in primary recipients transplanted with expanded cells from the MSC-based cultures was significantly higher than recipients transplanted with cells from the control cultures. Serial transplantation demonstrated that the expanded cells could establish long-term engraftment of hematopoietic cells. UCB-MSCs similar to those derived from adult bone marrow may provide novel targets for cellular and gene therapy.


Author(s):  
Viviana Rodríguez-Pardo ◽  
Jean Vernot

AbstractThe purpose of this study was to evaluate the influence of bone marrow-mesenchymal stem cells (BM-MSC) and exogenously added cytokines on the proliferation, primitive cell subpopulation maintenance (including the c-kit+ marker) and clonogenic capacity of hematopoietic stem cells (HSC). BM-MSC were collected from volunteer donors, isolated and characterized. Umbilical cord blood (UCB) samples were collected from healthy full-term deliveries. UCB-CD34+ cells were cultured in the presence or absence of BM-MSC and/or cytokines for 3 and 7 days. CD34+ cell proliferation was evaluated using the CSFE method and cell phenotype was determined by CD34, c-kit, CD33, CD38, HLA-DR, cyCD22 and cyCD3 detection. Cell clonogenic ability was also assessed. Exogenously added SCF, TPO and FLT3L increasedCD34+ cell proliferation in the presence or absence of BM-MSC, but with concomitant cell differentiation. Without any added cytokines, BM-MSC are able to increase the percentage of primitive progenitors as evaluated by c-kit expression and CFU-GEMM increase. Interestingly, this latter effect was dependent on both cell-cell interactions and secreted factors. A 7-day co-culture period will be optimal for obtaining an increased primitive HSC level. Including c-kit as a marker for primitive phenotype evaluation has shown the relevance of BM-MSC and their secreted factors on UCB-HSC stemness function. This effect could be dissociated from that of the addition of exogenous cytokines, which induced cellular differentiation instead.


2017 ◽  
Vol 14 (3) ◽  
pp. 923-931
Author(s):  
Nooshin Barikrow ◽  
Naser Amirizadeh ◽  
Nasim Hayati Roodbari ◽  
Mahin Nikougoftar

ABSTRACT: Because of insufficient number of umbilical cord blood hematopoietic stem cells (UCB-HSCs), expansion of these cells seems to be important for clinical application in adults. The aim of this study was to co-culture of UCB-HSCs with the amniotic membrane derived mesenchymal stem cells (AMMSCs) as a feeder layer in order to expand hematopoietic stem cells (HSCs). UCBs and amniotic membrane were collected from concern mothers. Ex vivo culture of UCB-HSCs were performed in four culture conditions: cytokine cocktail with MSCs feeder layer, cytokine cocktail, stem cell factor, and co-culture with MSCs without any cytokine. The number of total nucleated cells (TNC) was counted by hemocytometer. The HSC count and immunophenotyping of Mesenchymal stem cells (MSCs) and expanded HSC were evaluated by flow cytometry. Colony forming unit (CFU) assay was used to evaluate the potential of expanded HSCs for production of different lineage colonies. The mean fold changes of total nucleated cells (TNC) and CD34+ cells in the cytokine culture with feeder layer were higher than the cytokine culture without MSCs. However, in the co-culture system without cytokine, TNC and CD34+ cell numbers were increased up to 8 folds, but cell viability was more than 80% and differentiation rate was low. Our results demonstrated that we could increase the number of CD34+ cells of UCB that were used as primary HSC for transplantation.


Gene Reports ◽  
2019 ◽  
Vol 17 ◽  
pp. 100490
Author(s):  
Mohammadhosein Esmaeili ◽  
Vahid Niazi ◽  
Ali Akbar Pourfathollah ◽  
Mir Kamran Mousavi Hosseini ◽  
Mozhdeh Nakhlestani ◽  
...  

Author(s):  
Valentina Orticelli ◽  
Andrea Papait ◽  
Elsa Vertua ◽  
Patrizia Bonassi Signoroni ◽  
Pietro Romele ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document