Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 22.
Published in final edited form as: Best Pract Res Clin Haematol. 2011 Feb 23;24(1):83–92. doi: 10.1016/j.beha.2010.11.001

Mesenchymal Stem Cells in ex vivo Cord Blood Expansion

Simon N Robinson 1, Paul J Simmons 2, Hong Yang 1, Amin M Alousi 1, Marcos J de Lima 1, Elizabeth J Shpall 1,
PMCID: PMC3805362  NIHMSID: NIHMS276855  PMID: 21396596

Abstract

Umbilical cord blood (CB) is becoming an important source of haematopoietic support for transplant patients lacking human leukocyte antigen matched donors. The ethnic diversity, relative ease of collection, ready availability as cryopreserved units from CB banks, reduced incidence and severity of graft versus host disease and tolerance of higher degrees of HLA disparity between donor and recipient, are positive attributes when compared to bone marrow or cytokine-mobilized peripheral blood. However, CB transplantation is associated with significantly delayed neutrophil and platelet engraftment and an elevated risk of graft failure. These hurdles are thought to be due, at least in part, to low total nucleated cell and CD34+ cell doses transplanted. Here, current strategies directed at improving TNC and CD34+ cell doses at transplant are discussed, with particular attention paid to the use of a mesenchymal stem cell (MSC)/CB mononuclear cell ex vivo co-culture expansion system.

Keywords: Cord blood (CB) transplantation, ex vivo expansion, mesenchymal stem cells (MSC)

A. Background

Since the first CB transplant (CBT) was performed by Gluckman et al.[1] in 1988, >20,000 patients have received this procedure to support treatment for a variety of malignant and non-malignant diseases.[216] The reported event-free survival rates for such patients are comparable with those achieved following the transplantation of unrelated allogeneic bone marrow (BM), or mobilized peripheral blood progenitor cells (PBPCs).[15] In addition, there are many reports of lower rates of graft versus host disease (GvHD) than are commonly observed with BM and PBPC transplantation, particularly in pediatric patients. This reduced incidence of GvHD is observed despite the use of CB grafts with greater donor-recipient human leukocyte antigen (HLA) mismatching than would be tolerated by recipients of BM,[4;5;9;12] or PBPC allografts.[1719]

B. Challenges

One major challenge associated with the use of CB for transplantation is the relatively low cell dose available. This is thought to contribute, at least in part, to the slower engraftment and an elevated risk of engraftment failure that is associated with CBT.[2024] For example, the time required for a patient receiving CBT to achieve an absolute neutrophil count (ANC) of ≥0.5×109/L can range from 23 to 41 days. Similarly, the median time for a CBT patient to achieve a transfusion-independent platelet count of ≥20×109/L can range from 56 to >100 days. Further, while engraftment failure rates for CBT recipients as a whole (pediatric and adult CB recipients) can range from 12–20%,[5;9;13;14] those data for adult patients (>18 years old and/or >45 kg) are particularly poor with engraftment failure rates reaching of 20% or higher having been reported.

C. Evidence Of Threshold Doses For Effective Transplantation

Data from studies performed by Gluckman et al.[9] demonstrated that engraftment and survival were superior in CBT patients who received a transplant dose of ≥3.7×107 TNC/kg. These data suggest that there is a threshold CB total nucleated cell (TNC) dose above which time to engraftment is improved and graft failure rate reduced and below which time to engraftment is prolonged and graft failure rate increased. However, it is rare that a CBT cell dose of ≥3.7×107 TNC/kg is achieved. This is particularly true for CBT patients of >45 kg. Additional analyses of these data revealed that a lower, more readily realized target CBT dose ‘threshold’ of ≥1.0×107 TNC/kg was still associated with favorable engraftment rates and could be applied for this patient population.[2;9] However, analysis of data from patients who received myeloablative therapy and a single CB unit in North America or Europe through 2005 at the world’s three largest CBT registries: Center for International Blood and Marrow Transplant Research (CIBMTR), National Cord Blood Program (NCBP) and Eurocord,[25] revealed that a 100-day treatment-related mortality of approximately 44% was closely correlated with a CBT dose of <2.5×107 TNC/kg (P<0.0001) despite state-of-the-art care practices. These data underscore the urgent need to improve CBT strategies with the goal of improving neutrophil and platelet engraftment and reducing the risk of engraftment failure, especially for patients >45 kg.

D. Strategies To Enhance CBT Outcome

At the University of Texas M. D. Anderson Cancer Center Transplant Program, PBPC is the most commonly used source of unrelated haematopoietic support for cancer patients. PBPC remain the ‘gold standard’ against which the efficacy of CBT is compared. For comparison, patients who receive unrelated PBPC transplants achieve an ANC of ≥0.5×109/L at a median of 11 days post-transplant as compared to 23–41 days for CBT recipients. Similarly, while PBPC recipients achieve a transfusion-independent platelet count of ≥20×109/L at a median of 13 days post-transplant, patients receiving CBT might not achieve platelet engraftment until >100 days. Further, while PBPC recipients have an engraftment failure rate of <1%, rates of 12–20% are common for CBT. Given that these differences are likely a consequence of the limited cell dose associated with CBT, two major therapeutic strategies are being explored by different clinical centers to increase the cell dose at transplant and thereby improve time to neutrophil and platelet engraftment and reduce engraftment failure are (i) double CBT[2630] and (ii) ex vivo expansion.[26,27,31,32]

E. Double Cord Blood Transplantation

While double CBT does provide significantly more rapid neutrophil engraftment (23 days; range 15–41 days),[2830;33] when compared to single CBT, it continues to be associated with significantly delayed engraftment and elevated engraftment failure when compared to BM or PBPC transplantation.[27] The exact mechanisms by which double CBT improves the rate of engraftment over single CBT remain to be determined, however, it is worthy of note that while the progeny of both CB units are detectable for a short period after transplant, only one CB unit will ultimately predominate.[33] Measures that are predictive of which CB unit will ultimately predominate remain to be determined.

F. Rationale For Ex Vivo expansion

The observation that there is a threshold dose below which CBT recipients have markedly delayed engraftment and elevated risk of graft failure suggests that suboptimal numbers of the cells responsible for rapid engraftment are being transplanted.[34] If this is the case, increasing the dose of those CB cell subpopulations responsible for rapid engraftment, should improve the time to neutrophil and platelet engraftment and reduce the risk of graft failure. This has been the rationale behind the development of ex vivo expansion strategies. However, it is important to emphasize that double CBT and ex vivo expansion are not mutually exclusive, with the hope that a combination of these strategies might provide the greatest benefit to CBT recipients.

E. Concerns Associated With Ex Vivo Expansion

The goal of increasing the numbers of haematopoietic progenitors available for transplant using ex vivo expansion has been explored in a PBPC setting.[3544] These PBPC studies set the stage for the use of this approach with CB. While there is evidence of functional and phenotypic heterogeneity within the primitive haematopoietic progenitor compartment,[4548] one concern associated with ex vivo expansion is that short-term reconstituting, lower ‘quality’ haematopoietic progenitors will be expanded at the expense of longer-term reconstituting, higher ‘quality’ haematopoietic progenitors, thereby significantly impacting the haematopoietic reserve of the graft.[49] Evidence primarily in animal models, suggest that this may occur under certain conditions.[5057] Clinically, while the absence of durable engraftment from ex vivo expanded CD34+ cells has been reported in some cases,[58] durable engraftment has been reported in other patients who received expanded autologous products as the sole source of haematopoietic support following high dose therapy.[51] This suggests that the primitive haematopoietic progenitor cell compartment in general,[5964] properties of homing[65] and the basic biologic and genetic characteristics of the primitive haematopoietic progenitor cells[66] are preserved following ex vivo expansion.

F. Methodologies

(a) Static Liquid Cultures

Static liquid culture was shown to first require the purification of CD34+ (or CD133+) cells from fresh or frozen tissue.[44;67;68] Although clinical grade immunomagnetic devices can be used for the isolation procedure,[68] the purification process is limited by low recovery of the target cells (median CD34+ cell recovery of 35%, range, 4–70%). Despite this loss, isolated CB CD34+ (or CD133+) cells have been expanded ex vivo and used in clinical trials at the M. D. Anderson Cancer Center.[27;69] Initially, positively selected CD133+ cells were cultured in medium supplemented with 100 ng/ml each of granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF) and thrombopoietin (TPO),[70] for 10 days which resulted in a 56-fold expansion of total nucleated cells (TNCs) and a 4-fold expansion of CD34+ cells. A two-step, 14 day ex vivo CB expansion protocol was subsequently developed[71] where the CD133+ cells were cultured for one week with the same SCF-G-CSF-TPO regimen in a small volume of 50 ml and then transferred to a larger 800 ml volume on day 7 with fresh media/growth factors and continued culture for the second week. This procedure yielded a >400-fold increased TNCs and >20-fold increased CD34+ cells. It should be noted that in some cases, due to the upfront cell losses incurred by the immunomagnetic selection procedure, even marked ex vivo expansion only gives rise to a cell product whose numbers are not markedly different from that of the original CB unit. However, the argument can be made that exposure to the ex vivo expansion cytokine milieu generated a cell product that is different in ‘quality’ to the original CB unit with cells possibly ‘primed’ by the ex vivo exposure to growth factors and subsequently better able to home, engraft and proliferate when transplanted.

(b) Mesenchymal Stem Cell Based Cultures

Ex vivo liquid culture removes the primitive haematopoietic cells from molecular cues provided by the haematopoietic microenvironment. As a consequence the addition of exogenous cytokines is required to prevent apoptosis and stimulate proliferation. An alternative approach is the ex vivo co-culture of haematopoietic cells with components of their haematopoietic microenvironment. The haematopoietic microenvironment contains the putative stem cell ‘niche’ and is composed of haematopoietic and non-haematopoietic, cellular and extracellular components thought to provide the complex molecular cues that direct primitive haematopoietic progenitor self-renewal, proliferation and differentiation.[7280] This would be consistent with the observation that ex vivo contact between primitive haematopoietic progenitors and stromal components of the haematopoietic microenvironment preserve stem cell activity.[8187] Mesenchymal stem cells (MSC) are one component of the haematopoietic microenvironment and can be isolated from a variety of fetal and adult tissues.(89–92) Phenotypically MSC express CD73, CD90, CD105, CD16, and HLA-ABC(I) and do not express CD31, CD34, CD45, CD80 and HLA-DR(II). MSC can be grown and expanded as adherent, contact-inhibited monolayers in tissue culture flasks, although the primary nature of the cell-type limits the degree of expansion that can be achieved before senescence occurs.

G. Ex Vivo CB MNC/MSC Co-Culture

The CB MNC/MSC ex vivo co-culture technique does not require the isolation of CD34+ (or CD133+) cells from the CB, thereby minimizing the losses associated with this procedure. Using a co-culture strategy with bone marrow-derived MSCs and a supportive growth factor regimen which included Flt3-Ligand (FLT3-L), SCF, GCSF, and TPO, CB MNCs were cultured in 50 ml medium on stroma for 7 days at which time the non-adherent cells were removed in cultured in a larger volume (800 ml) for an additional 7 days. The flasks containing the adherent cells were also re-fed with 50 ml of media and the FLT3L-SCF-GCSF-TPO regimen and cultured for the subsequent 7 days. On day 14 all of the cells from the adherent and non-adherent cultures were pooled for evaluation. Using this strategy a 10–20 fold increase in TNCs, a 7–18-fold increase in committed progenitor cells (colony-forming units, CFU), a 2–5-fold increase in primitive haematopoietic progenitors (high proliferative potential colony-forming cells) and a 16–37-fold increase in CD34+ cells was achieved.[88] Building on this experience, a CB MNC/MSC co-culture strategy was developed to maximize the available expanded cells dose for transplant.[89] In the research laboratory, ex vivo expansion culture was characterized by a 6-fold increase in TNC, 30-fold increase in CD133+ cells, 8-fold increase in CD34+ cells, >200-fold increase in CFU, 50-fold increase in cobblestone area-forming cells persisting in culture for 2 weeks (CAFCwk2) and thought to be representative of more mature haematopoietic progenitors, and a reduction in (0.05-fold) cobblestone area-forming cells persisting in culture for 6 weeks (CAFCwk6) and thought to be representative of more primitive haematopoietic progenitors. These CAFCwk6 data provide evidence of the expansion of the more mature haematopoietic progenitors at the expense of the more primitive haematopoietic progenitor cell population. The fold increases cited are over those numbers originally present in the CB unit prior to ex vivo expansion and therefore represent true expansion.

H. Ex vivo CB MNC/MSC Co-Culture Expansion Trial at the M. D. Anderson Cancer Center

Clinical-scale CB MNC-MSC ex vivo expansion procedures were developed[89] and validated.[90] A trial was subsequently designed to test the clinical feasibility of transplanting the expansion product from CB MNC/MSC co-cultures into patients with haematologic malignancies. The trial, approved by the MD Anderson Institutional Review Board (IRB) Protocol 05-0781) and the U.S. Food and Drug Administration (FDA) (IND 13,034), details that patients will receive two CB units matched in at least 4/6 HLA antigens, with a minimum dose of 1×107 TNC/kg from each unit. For the initial cohort of 12 patients, a family member (matched at ≥2/6 antigens) serves as the third party haploidentical MSC donor.[90] Approximately 100 ml of marrow is aspirated from the donor and MSC isolated by plastic adherence. In preparation for co-culture with CB MNC, the MSC are grown to >70% confluence in 10 x T175 culture flasks. The CB unit with the lowest TNC dose is then thawed and divided equally between each of the 10 MSC layers, each in 50 ml of ex vivo expansion media containing 100 ng/ml each of SCF, Flt-3-ligand (Flt-3L), G-CSF and TPO. After 7 days of co-culture at 37°C in a 5% CO2-in-air fully humidified atmosphere, non-adherent cells are removed from each flask and each transferred into individual 10 × 1-liter Teflon-coated culture bags (American Fluoroseal) with fresh ex vivo expansion medium added to generate 800 ml. This liquid culture step (in the absence of MSC) is performed for an additional 7 days (14 days total). The original co-culture flasks also receive 50 mls of fresh medium. At the end of the ex vivo expansion procedure, all non-adherent cells from the culture bags and culture flasks are pooled, washed and prepared for transplantation. Samples are removed for prospective flow cytometric analysis and quality assurance testing.

I. Preliminary Clinical Results with Family Member-Derived MSCs

Patients were admitted on day −9 for hydration and received the designated preparative regimen on days −8 through −2. On day 0, the unmanipulated CB unit was thawed and infused, followed by infusion of the expanded CB cells. Median TNC and CD34+ cell expansions were 12-fold (range, 1–13) and 12-fold (range, 1–27), respectively. The mean expanded doses of 5.7×107 TNC/kg and 3.8×105 CD34+ cells/kg representing important increases compared to those achieved in our previous expansion studies. Furthermore, when the second unmanipulated CB unit is considered, patients on this trial received a total of 9.5×107 TNC/kg and 8.2×105 CD34+ cells/kg. Recipients of myeloablative therapy, engrafted neutrophils in a median of 14.5 days (range, 12–23) and platelets in 30 days (range, 25–51).

J. Rationale For The Use Of “Off-The-Shelf” MSC For The Clinical Protocol

The complex logistics of generating MSC from a patient’s family member have limited accrual to the clinical trial. Such limitations include:

  1. an appropriate family member was not always be available to donate marrow

  2. disease progression is rapid for selected patients with for example, acute leukemia who were relapsing during the 3 weeks it took to generate sufficient MSCs and 2 weeks to perform the CB MNC/MSC co-culture expansion (delaying transplant for 5 weeks total after patient enrolled on the trial). We postulated that the availability of an “off-the-shelf” source of Good Manufacturing Practice (GMP)-compliant, allogeneic MSC would alleviate this logistical problem with MSC essentially available for immediate use.

The development of master cell banks from young, healthy volunteers provides an optimal source of MSC and standardization of selection and isolation procedures ensures a reproducible, readily available MSC product. The Stro-1 antibody, developed by Dr. Paul Simmons,[91] allows prospective isolation of human bone marrow MSC without plastic adherence. Angioblast Systems, Inc., acquired the technology and developed the allogeneic MSC product Revascor for clinical use primarily in the treatment of ischaemic cardiovascular disease.[9295] To date, more than 20 congestive heart failure patients have received injections of Revascor with no adverse events related to the cells (Dr. Silviu Itescu, Angioblast Systems, Inc., personal communication). Angioblast has agreed to supply Revascor as an ‘off-the-shelf’ product for the M. D. Anderson CB MNC/MSC co-culture expansion trial.

Pre-Clinical Studies of Angioblast-MSC-CB Expansion

Preclinical studies have confirmed that the 10 flasks of MSC required for the ex vivo CB expansion protocol can be routinely generated in 4 days from a single vial (107 cells) of the Angioblast MSC product. Multiple experiments performed to compare the performance of the Angioblast MSC product with that of normal donor-derived MSC have revealed no difference in the expanded CB product generated.[96]

K. Preliminary Clinical Results with Angioblast-Derived MSCs

With the pre-clinical Angioblast data described above, the M.D. Anderson CB MNC/MSC ex vivo expansion Protocol 05-0781 and FDA IND 13,034 were amended to include a separate cohort of patients who would be treated identically to the first cohort, but who would receive CB cells expanded on the Angioblast product. Accrual to that cohort has been initiated. During CB MNC-MSC ex vivo expansion, a median expansion of 14-fold (range 1–30) for TNC and 40-fold (range 4–140) for the CD34+ cells was achieved. At transplant, the contribution of the unmanipulated CB included 2.35×107 (range 0.2–8.2) TNC/kg and 0.95 × 105 (range 0–4) CD34+ cells/kg, while the contribution of the ex vivo expanded CB unit was 5.8×107 (range, 0.3–14.4) TNC/kg and 8.7×105 (range, 0–93.4) CD34+ cells/kg. These were higher doses than we have ever infused into any of our recipients of unmanipulated double CBT, or CB expanded with our liquid culture system. As with the family member-derived MSCs, median time to neutrophil engraftment (500/μl) was 15 days (range 9–42) and platelet engraftment (>20,000/μl) was 38 days (range 13–62) with 26 patients (81%) of patients becoming platelet transfusion independant. On transplant day+21, the chimerism assays revealed that the MSC-expanded unit contributed to engraftment with a mean of 19% of the mononuclear cell, 16% of the T cell, and 14% of the myeloid fractions due to the expanded unit. Subsequently, hematopoiesis was increasingly derived from the unexpanded unit with long-term engraftment provided exclusively by the unexpanded unit by six months posttransplant in the vast majority of patients.

L. Summary

Our initial CB expansion protocol (#02-407, IND#7166) involved culture of CD133+ CB cells in teflon bags for 14 days with media containing 100 ng/ml SCF, G-CSF and TPO. In this system the CD133+ CB cells are initially cultured in bags with 50 ml of media/growth factors for 7 days and then transferred to a bag with 800 ml of fresh media and growth factors for another 7 days at which time they are washed and infused. With this strategy our patients experienced a modest improvement in engraftment of 20 days for neutrophils and 65 days for platelets compared to recipients of double unmanipulated CB units who engrafted neutrophils in 22 days and platelets in 100 days. However, we experienced a loss of >70% of the CB CD34+ cells following the CD133-selection procedure, which stimulated us to investigate the MSC-based co-culture system, where we could culture the entire CB unit without need for positive selection.

The use of the MSC-based CB expansion protocol (#05-0781) used a similar 14-day strategy where for the first 7 days the cells are cultured in 50 ml, but this time in 10 flasks containing 10% of the CB unit plus MSCs that are 70% or more confluent (rather than the entire CD133+ fraction in one bag with the liquid culture system). The growth factor regimen for this trial included the addition of Flt3-ligand to the SCF-GCSF-TPO regimen. On day 7, the non-adherent cells in each of the 10 flasks are transferred to 10 teflon-coated bags with 800 ml of media and the four growth factors (rather than transfer to a single bag in the liquid culture system with three growth factors). Fifty ml of media/growth factors are added to the 10 flasks containing the adherent layers of MSC-CB, and both those flasks and the 10 bags are cultured for another 7 days. On day 14, all of the cells in the bags and flasks are pooled, washed and infused. This strategy has shown more promising results providing markedly higher TNC and CD34+ cell doses than have ever before been achieved. The improvments are likely due to the use of MSCs to recapitulate the hematopoietic milieu, the ability to culture the whole CB unit rather than the CD133+ fraction, minimizing the large upfront CD34+ cell losses due to the positive selection procedure, and possibly the addition of FLT3-ligand to the cultures. The improvement in median times to engraftment of neutrophils (14 days) and platelets (35 days) are encouraging and accrual to the trial continues.

Acknowledgments

This work was supported in part by NIH Grant #RO1-CA061508-15 and Cancer Prevention & Research Institute of Texas (CPRIT) Grant # RP100469.

The authors would like to acknowledge the dedicated researchers, clinicians and nurses of the M. D. Anderson Cancer Center.

Footnotes

Conflict of interest statement

The authors declare that they have no competing financial interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Contributor Information

Simon N. Robinson, Email: snrobins@mdanderson.org.

Paul J. Simmons, Email: Paul.J.Simmons@uth.tmc.edu.

Hong Yang, Email: hyang@mdanderson.org.

Amin M Alousi, Email: aalousi@mdanderson.org.

Marcos J. de Lima, Email: mdelima@mdanderson.org.

Elizabeth J. Shpall, Email: eshpall@mdanderson.org.

References

  • *1.Gluckman E, Devergie A, Bourdeau-Esperou H, et al. Transplantation of umbilical cord blood in Fanconi’s anemia. Nouv Rev Fr Hematol. 1990;32:423–425. [PubMed] [Google Scholar]
  • 2.Wagner JE. Umbilical cord blood stem cell transplantation. Am J Pediatr Hematol Oncol. 1993;15:169–174. doi: 10.1097/00043426-199305000-00004. [DOI] [PubMed] [Google Scholar]
  • 3.Wagner JE, Kernan NA, Steinbuch M, et al. Allogeneic sibling umbilical-cord-blood transplantation in children with malignant and non-malignant disease. Lancet. 1995;346:214–219. doi: 10.1016/s0140-6736(95)91268-1. [DOI] [PubMed] [Google Scholar]
  • 4.Wagner JE, Rosenthal J, Sweetman R, et al. Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: analysis of engraftment and acute graft-versus-host disease. Blood. 1996;88:795–802. [PubMed] [Google Scholar]
  • *5.Kurtzberg J, Laughlin M, Graham ML, et al. Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients. N Engl J Med. 1996;335:157–166. doi: 10.1056/NEJM199607183350303. [DOI] [PubMed] [Google Scholar]
  • 6.Laporte JP, Gorin NC, Rubinstein P, et al. Cord-blood transplantation from an unrelated donor in an adult with chronic myelogenous leukemia. N Engl J Med. 1996;335:167–170. doi: 10.1056/NEJM199607183350304. [DOI] [PubMed] [Google Scholar]
  • 7.Laporte JP, Lesage S, Portnoi MF, et al. Unrelated mismatched cord blood transplantation in patients with hematological malignancies: a single institution experience. Bone Marrow Transplant. 1998;(Suppl 1):S76–S77. [PubMed] [Google Scholar]
  • 8.Cairo MS, Wagner JE. Placental and/or umbilical cord blood: an alternative source of hematopoietic stem cells for transplantation. Blood. 1997;90:4665–4678. [PubMed] [Google Scholar]
  • *9.Gluckman E, Rocha V, Boyer-Chammard A, et al. Outcome of cord-blood transplantation from related and unrelated donors. Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. N Engl J Med. 1997;337:373–381. doi: 10.1056/NEJM199708073370602. [DOI] [PubMed] [Google Scholar]
  • 10.Locatelli F, Rocha V, Chastang C, et al. Cord blood transplantation for children with acute leukemia. Eurocord Transplant Group. Bone Marrow Transplant. 1998;21 (Suppl 3):S63–S65. [PubMed] [Google Scholar]
  • 11.Locatelli F, Rocha V, Chastang C, et al. Factors associated with outcome after cord blood transplantation in children with acute leukemia. Eurocord-Cord Blood Transplant Group. Blood. 1999;93:3662–3671. [PubMed] [Google Scholar]
  • 12.Rocha V, Wagner JE, Jr, Sobocinski KA, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources. N Engl J Med. 2000;342:1846–1854. doi: 10.1056/NEJM200006223422501. [DOI] [PubMed] [Google Scholar]
  • 13.Long GD, Laughlin M, Madan B, et al. Unrelated umbilical cord blood transplantation in adult patients. Biol Blood Marrow Transplant. 2003;9:772–780. doi: 10.1016/j.bbmt.2003.08.007. [DOI] [PubMed] [Google Scholar]
  • 14.Laughlin MJ, Barker J, Bambach B, et al. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors. N Engl J Med. 2001;344:1815–1822. doi: 10.1056/NEJM200106143442402. [DOI] [PubMed] [Google Scholar]
  • 15.Barker JN, Davies SM, DeFor T, et al. Survival after transplantation of unrelated donor umbilical cord blood is comparable to that of human leukocyte antigen-matched unrelated donor bone marrow: results of a matched-pair analysis. Blood. 2001;97:2957–2961. doi: 10.1182/blood.v97.10.2957. [DOI] [PubMed] [Google Scholar]
  • 16.Eapen M, Rubinstein P, Zhang MJ, et al. Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: a comparison study. Lancet. 2007;369:1947–1954. doi: 10.1016/S0140-6736(07)60915-5. [DOI] [PubMed] [Google Scholar]
  • 17.Bensinger WI, Clift R, Martin P, et al. Allogeneic peripheral blood stem cell transplantation in patients with advanced hematologic malignancies: a retrospective comparison with marrow transplantation. Blood. 1996;88:2794–2800. [PubMed] [Google Scholar]
  • 18.Bensinger WI, Weaver CH, Appelbaum FR, et al. Transplantation of allogeneic peripheral blood stem cells mobilized by recombinant human granulocyte colony-stimulating factor. Blood. 1995;85:1655–1658. [PubMed] [Google Scholar]
  • 19.Bensinger WI, Martin PJ, Storer B, et al. Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers. N Engl J Med. 2001;344:175–181. doi: 10.1056/NEJM200101183440303. [DOI] [PubMed] [Google Scholar]
  • 20.McGlave P, Bartsch G, Anasetti C, et al. Unrelated donor marrow transplantation therapy for chronic myelogenous leukemia: initial experience of the National Marrow Donor Program. Blood. 1993;81:543–550. [PubMed] [Google Scholar]
  • 21.Kernan NA, Bartsch G, Ash RC, et al. Analysis of 462 transplantations from unrelated donors facilitated by the National Marrow Donor Program. N Engl J Med. 1993;328:593–602. doi: 10.1056/NEJM199303043280901. [DOI] [PubMed] [Google Scholar]
  • 22.Schiller G, Feig SA, Territo M, et al. Treatment of advanced acute leukaemia with allogeneic bone marrow transplantation from unrelated donors. Br J Haematol. 1994;88:72–78. doi: 10.1111/j.1365-2141.1994.tb04979.x. [DOI] [PubMed] [Google Scholar]
  • 23.Szydlo R, Goldman JM, Klein JP, et al. Results of allogeneic bone marrow transplants for leukemia using donors other than HLA-identical siblings. J Clin Oncol. 1997;15:1767–1777. doi: 10.1200/JCO.1997.15.5.1767. [DOI] [PubMed] [Google Scholar]
  • 24.Berthou C, Legros-Maida S, Soulie A, et al. Cord blood T lymphocytes lack constitutive perforin expression in contrast to adult peripheral blood T lymphocytes. Blood. 1995;85:1540–1546. [PubMed] [Google Scholar]
  • 25.Cohen YC, Scaradavou A, Stevens CE, et al. Factors affecting mortality following myeloablative cord blood transplantation in adults: a pooled analysis of three international registries. Bone Marrow Transplant. 2010 doi: 10.1038/bmt.2010.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Delaney C, Varnum-Finney B, Aoyama K, et al. Dose-dependent effects of the Notch ligand Delta1 on ex vivo differentiation and in vivo marrow repopulating ability of cord blood cells. Blood. 2005;106:2693–2699. doi: 10.1182/blood-2005-03-1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.de Lima M, McMannis J, Komanduri K, et al. Randomized study of double cord blood transplantation (CBT) with versus without ex-vivo expansion. Blood. 2007;110:2014a. [Google Scholar]
  • 28.Brunstein CG, Weisdorf DJ, DeFor T, et al. Marked increased risk of Epstein-Barr virus-related complications with the addition of antithymocyte globulin to a nonmyeloablative conditioning prior to unrelated umbilical cord blood transplantation. Blood. 2006;108:2874–2880. doi: 10.1182/blood-2006-03-011791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Barker JN, Weisdorf DJ, Wagner JE. Creation of a double chimera after the transplantation of umbilical-cord blood from two partially matched unrelated donors. N Engl J Med. 2001;344:1870–1871. doi: 10.1056/NEJM200106143442417. [DOI] [PubMed] [Google Scholar]
  • 30.Barker J, Defor T, Davies S. Impact of multiple unit unrelated donor umbilical cord blood transplantation in adults: Preliminary analysis of safety and efficacy. Blood. 2001;98:666a. [Google Scholar]
  • 31.Kurtzberg J, Balber A, Mendizabal A. Preliminary results of a pilot trial of unrelated umbilical cord blood transplantation (UCBT) augmented with cytokine-primed aldehyde dehydrogenase-bright cells (ALDHbr) Blood. 2006;108:3641a. [Google Scholar]
  • 32.Shpall E, de Lima M, Chan K. Transplantation of cord blood expanded ex vivo with copper chelator. Blood. 2004;104:281a. [Google Scholar]
  • 33.Barker JN, Weisdorf DJ, DeFor TE, et al. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005;105:1343–1347. doi: 10.1182/blood-2004-07-2717. [DOI] [PubMed] [Google Scholar]
  • 34.Broxmeyer HE, Hangoc G, Cooper S, et al. Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation in adults. Proc Natl Acad Sci U S A. 1992;89:4109–4113. doi: 10.1073/pnas.89.9.4109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Haylock DN, To LB, Dowse TL, et al. Ex vivo expansion and maturation of peripheral blood CD34+ cells into the myeloid lineage. Blood. 1992;80:1405–1412. [PubMed] [Google Scholar]
  • 36.Haylock DN, To LB, Dowse TL, et al. The Stem Cell Manual. Alpha Med Press; 1994. Ex vivo expansion of hematopoietic stem cells; pp. 291–301. [Google Scholar]
  • 37.Simmons PJ, Haylock DN. Use of hematopoietic growth factors for in vitro expansion of precursor cell populations. Curr Opin Hematol. 1995;2:189–195. doi: 10.1097/00062752-199502030-00006. [DOI] [PubMed] [Google Scholar]
  • 38.Haylock DN, Horsfall MJ, Dowse TL, et al. Increased recruitment of hematopoietic progenitor cells underlies the ex vivo expansion potential of FLT3 ligand. Blood. 1997;90:2260–2272. [PubMed] [Google Scholar]
  • 39.Makino S, Haylock DN, Dowse T, et al. Ex vivo culture of peripheral blood CD34+ cells: effects of hematopoietic growth factors on production of neutrophilic precursors. J Hematother. 1997;6:475–489. doi: 10.1089/scd.1.1997.6.475. [DOI] [PubMed] [Google Scholar]
  • *40.Prince HM, Simmons PJ, Whitty G, et al. Improved haematopoietic recovery following transplantation with ex vivo-expanded mobilized blood cells. Br J Haematol. 2004;126:536–545. doi: 10.1111/j.1365-2141.2004.05081.x. [DOI] [PubMed] [Google Scholar]
  • 41.Andrews RG, Briddell RA, Hill R, et al. Engraftment of primates with G-CSF mobilized peripheral blood CD34+ progenitor cells expanded in G-CSF, SCF and MGDF decreases the duration and severity of neutropenia. Stem Cells. 1999;17:210–218. doi: 10.1002/stem.170210. [DOI] [PubMed] [Google Scholar]
  • 42.Boiron JM, Dazey B, Cailliot C, et al. Large-scale expansion and transplantation of CD34(+) hematopoietic cells: in vitro and in vivo confirmation of neutropenia abrogation related to the expansion process without impairment of the long-term engraftment capacity. Transfusion. 2006;46:1934–1942. doi: 10.1111/j.1537-2995.2006.01001.x. [DOI] [PubMed] [Google Scholar]
  • 43.McAlister IB, Teepe M, Gillis S, Williams DE. Ex vivo expansion of peripheral blood progenitor cells with recombinant cytokines. Exp Hematol. 1992;20:626–628. [PubMed] [Google Scholar]
  • 44.Purdy MH, Hogan CJ, Hami L, et al. Large volume ex vivo expansion of CD34-positive hematopoietic progenitor cells for transplantation. J Hematother. 1995;4:515–525. doi: 10.1089/scd.1.1995.4.515. [DOI] [PubMed] [Google Scholar]
  • 45.Dick JE, Guenechea G, Gan OI, Dorrell C. In vivo dynamics of human stem cell repopulation in NOD/SCID mice. Ann N Y Acad Sci. 2001;938:184–190. doi: 10.1111/j.1749-6632.2001.tb03588.x. [DOI] [PubMed] [Google Scholar]
  • 46.Guenechea G, Gan OI, Dorrell C, Dick JE. Distinct classes of human stem cells that differ in proliferative and self-renewal potential. Nat Immunol. 2001;2:75–82. doi: 10.1038/83199. [DOI] [PubMed] [Google Scholar]
  • 47.Lemischka IR, Jordan CT. The return of clonal marking sheds new light on human hematopoietic stem cells. Nat Immunol. 2001;2:11–12. doi: 10.1038/83115. [DOI] [PubMed] [Google Scholar]
  • 48.Hogan CJ, Shpall EJ, Keller G. Differential long-term and multilineage engraftment potential from subfractions of human CD34+ cord blood cells transplanted into NOD/SCID mice. Proc Natl Acad Sci U S A. 2002;99:413–418. doi: 10.1073/pnas.012336799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Williams DA. Ex vivo expansion of hematopoietic stem and progenitor cells--robbing Peter to pay Paul? Blood. 1993;81:3169–3172. [PubMed] [Google Scholar]
  • 50.McNiece IK, Almeida-Porada G, Shpall EJ, Zanjani E. Ex vivo expanded cord blood cells provide rapid engraftment in fetal sheep but lack long-term engrafting potential. Exp Hematol. 2002;30:612–616. doi: 10.1016/s0301-472x(02)00805-6. [DOI] [PubMed] [Google Scholar]
  • *51.McNiece I, Jones R, Bearman SI, Cagnoni P, et al. Ex vivo expanded peripheral blood progenitor cells provide rapid neutrophil recovery after high-dose chemotherapy in patients with breast cancer. Blood. 2000;96:3001–3007. [PubMed] [Google Scholar]
  • 52.Shimizu Y, Ogawa M, Kobayashi M, et al. Engraftment of cultured human hematopoietic cells in sheep. Blood. 1998;91:3688–3692. [PubMed] [Google Scholar]
  • 53.Tisdale JF, Hanazono Y, Sellers SE, et al. Ex vivo expansion of genetically marked rhesus peripheral blood progenitor cells results in diminished long-term repopulating ability. Blood. 1998;92:1131–1141. [PubMed] [Google Scholar]
  • 54.Abkowitz JL, Taboada MR, Sabo KM, Shelton GH. The ex vivo expansion of feline marrow cells leads to increased numbers of BFU-E and CFU-GM but a loss of reconstituting ability. Stem Cells. 1998;16:288–293. doi: 10.1002/stem.160288. [DOI] [PubMed] [Google Scholar]
  • 55.Von Drygalski A, Alespeiti G, Ren L, Adamson JW. Murine bone marrow cells cultured ex vivo in the presence of multiple cytokine combinations lose radioprotective and long-term engraftment potential. Stem Cells Dev. 2004;13:101–111. doi: 10.1089/154732804773099308. [DOI] [PubMed] [Google Scholar]
  • 56.Peters SO, Kittler EL, Ramshaw HS, Quesenberry PJ. Ex vivo expansion of murine marrow cells with interleukin-3 (IL-3), IL-6, IL-11, and stem cell factor leads to impaired engraftment in irradiated hosts. Blood. 1996;87:30–37. [PubMed] [Google Scholar]
  • 57.Peters SO, Kittler EL, Ramshaw HS, Quesenberry PJ. Murine marrow cells expanded in culture with IL-3, IL-6, IL-11, and SCF acquire an engraftment defect in normal hosts. Exp Hematol. 1995;23:461–469. [PubMed] [Google Scholar]
  • 58.Holyoake TL, Alcorn MJ, Richmond L, et al. CD34 positive PBPC expanded ex vivo may not provide durable engraftment following myeloablative chemoradiotherapy regimens. Bone Marrow Transplant. 1997;19:1095–1101. doi: 10.1038/sj.bmt.1700799. [DOI] [PubMed] [Google Scholar]
  • 59.Piacibello W, Sanavio F, Severino A, et al. Engraftment in nonobese diabetic severe combined immunodeficient mice of human CD34(+) cord blood cells after ex vivo expansion: evidence for the amplification and self-renewal of repopulating stem cells. Blood. 1999;93:3736–3749. [PubMed] [Google Scholar]
  • 60.Lewis ID, Almeida-Porada G, Du J, Lemischka IR, et al. Umbilical cord blood cells capable of engrafting in primary, secondary, and tertiary xenogeneic hosts are preserved after ex vivo culture in a noncontact system. Blood. 2001;97:3441–3449. doi: 10.1182/blood.v97.11.3441. [DOI] [PubMed] [Google Scholar]
  • 61.Guenechea G, Segovia JC, Albella B, Lamana M, et al. Delayed engraftment of nonobese diabetic/severe combined immunodeficient mice transplanted with ex vivo-expanded human CD34(+) cord blood cells. Blood. 1999;93:1097–1105. [PubMed] [Google Scholar]
  • 62.Traycoff CM, Cornetta K, Yoder MC, et al. Ex vivo expansion of murine hematopoietic progenitor cells generates classes of expanded cells possessing different levels of bone marrow repopulating potential. Exp Hematol. 1996;24:299–306. [PubMed] [Google Scholar]
  • 63.Muench MO, Moore MA. Accelerated recovery of peripheral blood cell counts in mice transplanted with in vitro cytokine-expanded hematopoietic progenitors. Exp Hematol. 1992;20:611–618. [PubMed] [Google Scholar]
  • 64.Holyoake TL, Freshney MG, McNair L, et al. Ex vivo expansion with stem cell factor and interleukin-11 augments both short-term recovery posttransplant and the ability to serially transplant marrow. Blood. 1996;87:4589–4595. [PubMed] [Google Scholar]
  • 65.Zhai QL, Qiu LG, Li Q, Meng HX, et al. Short-term ex vivo expansion sustains the homing-related properties of umbilical cord blood hematopoietic stem and progenitor cells. Haematologica. 2004;89:265–273. [PubMed] [Google Scholar]
  • 66.Tian H, Huang S, Gong F, et al. Karyotyping, immunophenotyping, and apoptosis analyses on human hematopoietic precursor cells derived from umbilical cord blood following long-term ex vivo expansion. Cancer Genet Cytogenet. 2005;157:33–36. doi: 10.1016/j.cancergencyto.2004.06.003. [DOI] [PubMed] [Google Scholar]
  • 67.Briddell RA, Kern BP, Zilm KL, et al. Purification of CD34+ cells is essential for optimal ex vivo expansion of umbilical cord blood cells. J Hematother. 1997;6:145–150. doi: 10.1089/scd.1.1997.6.145. [DOI] [PubMed] [Google Scholar]
  • 68.McNiece IK, Stoney GB, Kern BP, Briddell RA. CD34+ cell selection from frozen cord blood products using the Isolex 300i and CliniMACS CD34 selection devices. J Hematother. 1998;7:457–461. doi: 10.1089/scd.1.1998.7.457. [DOI] [PubMed] [Google Scholar]
  • 69.de Lima M, McMannis J, Gee A, et al. Transplantation of ex vivo expanded cord blood cells using the copper chelator tetraethylenepentamine: a phase I/II clinical trial. Bone Marrow Transplant. 2008;41:771–778. doi: 10.1038/sj.bmt.1705979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *70.Shpall EJ, Quinones R, Giller R, et al. Transplantation of ex vivo expanded cord blood. Biol Blood Marrow Transplant. 2002;8:368–376. doi: 10.1053/bbmt.2002.v8.pm12171483. [DOI] [PubMed] [Google Scholar]
  • 71.McNiece I, Kubegov D, Kerzic P, et al. Increased expansion and differentiation of cord blood products using a two-step expansion culture. Exp Hematol. 2000;28:1181–1186. doi: 10.1016/s0301-472x(00)00520-8. [DOI] [PubMed] [Google Scholar]
  • 72.Lemischka IR, Moore KA. Stem cells: interactive niches. Nature. 2003;425:778–779. doi: 10.1038/425778a. [DOI] [PubMed] [Google Scholar]
  • 73.Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and their niche. Cell. 2004;116:769–778. doi: 10.1016/s0092-8674(04)00255-7. [DOI] [PubMed] [Google Scholar]
  • 74.Hackney JA, Charbord P, Brunk BP, et al. A molecular profile of a hematopoietic stem cell niche. Proc Natl Acad Sci U S A. 2002;99:13061–13066. doi: 10.1073/pnas.192124499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Etheridge SL, Spencer GJ, Heath DJ, Genever PG. Expression profiling and functional analysis of wnt signaling mechanisms in mesenchymal stem cells. Stem Cells. 2004;22:849–860. doi: 10.1634/stemcells.22-5-849. [DOI] [PubMed] [Google Scholar]
  • 76.Kadereit S, Deeds LS, Haynesworth SE, et al. Expansion of LTC-ICs and maintenance of p21 and BCL-2 expression in cord blood CD34(+)/CD38(−) early progenitors cultured over human MSCs as a feeder layer. Stem Cells. 2002;20:573–582. doi: 10.1634/stemcells.20-6-573. [DOI] [PubMed] [Google Scholar]
  • 77.Rattis FM, Voermans C, Reya T. Wnt signaling in the stem cell niche. Curr Opin Hematol. 2004;11:88–94. doi: 10.1097/01.moh.0000133649.61121.ec. [DOI] [PubMed] [Google Scholar]
  • 78.Zhang J, Niu C, Ye L, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425:836–841. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
  • 79.Majumdar MK, Keane-Moore M, Buyaner D, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci. 2003;10:228–241. doi: 10.1007/BF02256058. [DOI] [PubMed] [Google Scholar]
  • 80.Majumdar MK, Thiede MA, Mosca JD, et al. Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol. 1998;176:57–66. doi: 10.1002/(SICI)1097-4652(199807)176:1<57::AID-JCP7>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  • 81.Yildirim S, Boehmler AM, Kanz L, Mohle R. Expansion of cord blood CD34+ hematopoietic progenitor cells in coculture with autologous umbilical vein endothelial cells (HUVEC) is superior to cytokine-supplemented liquid culture. Bone Marrow Transplant. 2005;36:71–79. doi: 10.1038/sj.bmt.1705001. [DOI] [PubMed] [Google Scholar]
  • 82.Breems DA, Blokland EA, Siebel KE, et al. Stroma-contact prevents loss of hematopoietic stem cell quality during ex vivo expansion of CD34+ mobilized peripheral blood stem cells. Blood. 1998;91:111–117. [PubMed] [Google Scholar]
  • 83.Brandt JE, Galy AH, Luens KM, et al. Bone marrow repopulation by human marrow stem cells after long-term expansion culture on a porcine endothelial cell line. Exp Hematol. 1998;26:950–961. [PubMed] [Google Scholar]
  • 84.Rafii S, Shapiro F, Rimarachin J, et al. Isolation and characterization of human bone marrow microvascular endothelial cells: hematopoietic progenitor cell adhesion. Blood. 1994;84:10–19. [PubMed] [Google Scholar]
  • 85.Rafii S, Shapiro F, Pettengell R, et al. Human bone marrow microvascular endothelial cells support long-term proliferation and differentiation of myeloid and megakaryocytic progenitors. Blood. 1995;86:3353–3363. [PubMed] [Google Scholar]
  • 86.Chute JP, Saini AA, Chute DJ, et al. Ex vivo culture with human brain endothelial cells increases the SCID-repopulating capacity of adult human bone marrow. Blood. 2002;100:4433–4439. doi: 10.1182/blood-2002-04-1238. [DOI] [PubMed] [Google Scholar]
  • 87.Davis TA, Robinson DH, Lee KP, Kessler SW. Porcine brain microvascular endothelial cells support the in vitro expansion of human primitive hematopoietic bone marrow progenitor cells with a high replating potential: requirement for cell-to-cell interactions and colony-stimulating factors. Blood. 1995;85:1751–1761. [PubMed] [Google Scholar]
  • *88.McNiece I, Harrington J, Turney J, et al. Ex vivo expansion of cord blood mononuclear cells on mesenchymal stem cells. Cytotherapy. 2004;6:311–317. doi: 10.1080/14653240410004871. [DOI] [PubMed] [Google Scholar]
  • *89.Robinson SN, Ng J, Niu T, et al. Superior ex vivo cord blood expansion following co-culture with bone marrow-derived mesenchymal stem cells. Bone Marrow Transplant. 2006;37:359–366. doi: 10.1038/sj.bmt.1705258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.de Lima M, McMannis JD, Hosing CM, et al. Rapid engraftment of neutrophils and platelets with mesenchymal stem cell based cord blood expansion. Blood. 2009;114:3394a. [Google Scholar]
  • *91.Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood. 1991;78:55–62. [PubMed] [Google Scholar]
  • 92.Martens TP, See F, Schuster MD, et al. Mesenchymal lineage precursor cells induce vascular network formation in ischemic myocardium. Nat Clin Pract Cardiovasc Med. 2006;3 (Suppl 1):S18–S22. doi: 10.1038/ncpcardio0404. [DOI] [PubMed] [Google Scholar]
  • 93.Hasnat AK, van d V, Hon JK, Yacoub MH. Reproducible model of post-infarction left ventricular dysfunction: haemodynamic characterization by conductance catheter. Eur J Cardiothorac Surg. 2003;24:98–104. doi: 10.1016/s1010-7940(03)00261-6. [DOI] [PubMed] [Google Scholar]
  • 94.Moainie SL, Gorman JH, III, Guy TS, et al. An ovine model of postinfarction dilated cardiomyopathy. Ann Thorac Surg. 2002;74:753–760. doi: 10.1016/s0003-4975(02)03827-4. [DOI] [PubMed] [Google Scholar]
  • 95.Charles CJ, Elliott JM, Nicholls MG, et al. Myocardial infarction with and without reperfusion in sheep: early cardiac and neurohumoral changes. Clin Sci. 2000;98:703–711. [PubMed] [Google Scholar]
  • 96.Robinson SN, Simmons PJ, Brouard N, et al. Efficacy of off-the-shelf, commercially-available, third-party mesenchymal stem cells (MSC) in ex vivo cord blood (CB) co-culture expansion. Blood. 2007;110:4106a. [Google Scholar]
  • 97.Yang H, Robinson SN, Lu J, et al. CD3(+) and/or CD14(+) depletion from cord blood mononuclear cells before ex vivo expansion culture improves total nucleated cell and CD34(+) cell yields. Bone Marrow Transplant. 2010;45:1000–1007. doi: 10.1038/bmt.2009.289. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES