• No results found

Current Opinion in Pediatrics Cord Blood Banking and Transplantation: Advances and Controversies

N/A
N/A
Protected

Academic year: 2021

Share "Current Opinion in Pediatrics Cord Blood Banking and Transplantation: Advances and Controversies"

Copied!
22
0
0

Loading.... (view fulltext now)

Full text

(1)

Current Opinion in Pediatrics

Cord Blood Banking and Transplantation: Advances and Controversies

--Manuscript

Draft--Manuscript Number:

Full Title: Cord Blood Banking and Transplantation: Advances and Controversies

Article Type: Review Article

Corresponding Author: Mervin C Yoder

Indianapolis, IN UNITED STATES Corresponding Author Secondary

Information:

Corresponding Author's Institution: Corresponding Author's Secondary Institution:

First Author: Mervin C Yoder

First Author Secondary Information:

Order of Authors: Mervin C Yoder Order of Authors Secondary Information:

(2)

Cord Blood Banking and Transplantation: Advances and Controversies

Mervin C. Yoder, MD Distinguished Professor and

Richard and Pauline Klingler Professor of Pediatrics Department of Pediatrics

Indiana University School of Medicine Indianapolis, IN

Phone: 317-274-4738 myoder@iu.edu

(3)

Abstract

Purpose of review: A review of papers published since January 2012 on the topic of cord blood banking and cord blood stem cell transplantation was conducted for this the 25th anniversary year of the first cord blood transplant performed in a human subject.

Recent findings: Cord blood banking is performed throughout the world. Umbilical cord blood (UCB) transplantation is recognized as an acceptable alternative stem cell source for pediatric and adult subjects requiring a hematopoietic transplant, particularly for patients of racial and ethnic minorities. To further advance the use of UCB, methods to enhance UCB stem cell expansion, engraftment, and maintenance may be required. Controversy on the most effective and economically sustainable model for banking and storing an optimal UCB product continues to persist.

Summary: Cord blood banking and transplantation of cord blood stem cells has advanced rapidly over the initial 25 years as more than 30,000 patients have benefited from the therapy. New concepts on the use of methods to expand UCB stem cells for transplantation and use for non-hematopoietic indications may increase demand for UCB over the next few decades.

Keywords: Umbilical cord blood

Umbilical cord blood banking

(4)

Introduction

This has been a notable year for cord blood transplantation and banking, since it was 25 years ago that a child with Fanconi Anemia received an umbilical cord blood (UCB) transplant. Since that time, more than 600,000 UCB units have been cryopreserved and stored in banks throughout the world and >30,000 transplants have been performed using stored material [1]. We will briefly point out some notable achievements that have occurred over the past 25 years and highlight some recent advances in cord blood transplantation. We will also point out a few areas where UCB has been proposed as a novel source of stem cells that may provide repair or regeneration of tissue function in non-hematopoietic systems. Finally, we will discuss the ongoing challenge of providing education to the public as to the different types of cord blood banks that collect and preserve UCB under different economic and blood banking standards.

Overview of highlights from the first 25 years of cord blood transplantation and banking

The first UCB transplant was successfully performed through the efforts of a team of international investigators [1]. A child with FA who was suffering from severe aplastic anemia was deemed a potentially suitable candidate when a tissue matched sibling was born and UCB was isolated and cryopreserved in the laboratory of Dr. Hal Broxmeyer. Dr. Arleen Auerbach determined that the UCB of the donor sibling was unaffected by FA. Dr. Eliane Gluckman had previously developed pre-transplant conditioning regimens for patients with FA, and successfully performed the UCB transplant in October of 1988 after appropriate ethics review [1, 2]. The patient displayed signs of donor cell engraftment on day 22 post-transplant and subsequently achieved a full hematopoietic reconstitution with donor cells. Now 25 years later, this healthy patient continues to show complete hematologic and immunologic donor cell chimerism [1].

Several years after this epic event, the first public UCB bank was formed at the New York Blood Center [3] and in 1993 the first unrelated UCB transplant was performed [4, 5].

(5)

Subsequently UCB was increasingly utilized in pediatric patients. When comparing bone marrow versus UCB as a source of matched sibling donor stem cells`, transplantation of UCB was associated with delayed neutrophil and platelet time to engraftment`, reduced acute and chronic GVHD`, but similar survival [6]. In a recent analysis of 5 year outcomes of children with acute leukemia that received a sibling UCB transplant, the probability of disease free survival (DFS) was 44% [7]. In children with acute leukemia that received either an unrelated matched UCB or an unrelated matched bone marrow transplant, recipients of the UCB had better outcomes [8]. An increase in transplant related mortality (TRM) was detected in children receiving < 3 X 107 total nucleated cells per kilogram bodyweight and a 1 HLA-disparate match or in children given a 2 HLA-disparate UCB transplant, pointing out the importance of UCB unit selection for improving outcomes [8]. One approach to enhancing cell dose is to provide a double UCB transplant. A preliminary analysis of a clinical study directly comparing the use of a single versus a double UCB transplant in pediatric patients with a hematologic malignancy did not show a survival advantage [1, 9].

Use of UCB as a stem cell source in adult patients with leukemia failed to result in the promising results obtained in pediatric patients with concerns that the neutrophil and platelet recovery times were significantly delayed and there was high TRM [10]. However, improvements in patient selection, better supportive care, and the use of higher infused cell dose have been associated with increased survival. A recent analysis of factors that affect mortality following myeloablative UCB transplantation in adult subjects identified older age, advanced disease, and limited center experience correlated with worse survival [11]. However, a remarkable 60-70% 5 year DFS was reported by one group from Japan for adult patients with acute myelogenous leukemia (AML) that underwent myeloablative conditioning prior to receiving a single UCB transplant [12]. To improve outcomes in the United States, combining reduced intensity conditioning with double cord blood transplantation has resulted in 30-50% DFS in adult subjects with malignancy [13, 14]. Improvements in UCB unit selection are expected to

(6)

further enhance outcomes in adult subjects that need an UCB transplant [15]. Thus, one recent review recommends that UCB transplantation should be considered in all high-risk adult subjects with AML in whom an allogeneic stem cell transplant is indicated but who lack a matched related or unrelated donor [1].

Methods to expand UCB hematopoietic stem cells

Given the delayed donor cell reconstitution of UCB recipients and the associated increased incidence of late viral infections and other TRM [16], strategies to enhance the numbers of transplanted stem cells in each UCB unit have long been desired [17, 18]. Results from recent clinical trials testing the safety and feasibility of expanding an UCB unit ex vivo and then infusing with a second unmanipulated UCB unit have been reported.

Notch-mediated expansion of UCB capable of rapid myeloid reconstitution

Activation of endogenous Notch receptors on UCB CD34+ cells via co-culture on immobilized engineered Delta-like ligand 1 was known to increase the CD34+ content nearly 100-fold and to increase the engraftment of the Notch activated cells in immunodeficient mice [19]. In preliminary clinical studies, use of the same approach provided a significant expansion of UCB CD34+ cells and when co-infused with an unmanipulated UCB unit, reduced the time to an absolute neutrophil count (ANC) >500 cells per L to a median of 16 days. This time was significantly reduced compared to a median of 26 days for ANC recovery in patients concurrently treated at the same institution with a double UCB transplant [20]. A recent update to that report indicates that 17 patients have now been enrolled and the median time to ANC recovery has been shortened to 11 days compared to 25 days in a concurrently enrolled institutional cohort of 36 patients undergoing a double UCB transplant [18]. Since the expanded unit predominated during the first week post-transplant, evidence exists that the Notch activated UCB cells provide rapid myeloid recovery.

(7)

UCB engraftment with ex vivo mesenchymal-cell co-culture

Expansion of hematopoietic progenitor cells in unfractionated UCB cells has been known to be markedly enhanced upon co-culture with mesenchymal stromal cells [21]. In a recent report, 31 adult subjects with hematological cancer that received an UCB transplant with 1 expanded unit and 1 unmanipulated unit were compared to 80 historical control subjects who received 2 unmanipulated UCB units. The median time to neutrophil engraftment was 15 days in the patients that received the expanded unit plus the unmanipulated unit compared to 24 days in the control patients that received a double UCB unit transplant; a significant improvement [22]. The median time to platelet recovery was also significantly shortened from 49 days in the control patients to 42 days in the patients receiving the expanded plus unmanipulated UCB. Of interest, the dose of the CD34+ cells and total nucleated cells per kilogram bodyweight in the recipients of the expanded unit significantly correlated with the time to neutrophil recovery. Long-term engraftment of more than 1 year was produced primarily by the unit of unmanipulated UCB in these patients. These results support the hypothesis that transplantation of UCB cells expanded with mesenchymal cells shortens the time to neutrophil and platelet recovery in adult subjects [22].

Other approaches to expand UCB

A variety of other approaches have been examined for expanding human UCB cells. One approach expands a portion of an UCB unit using selected growth factors in the presence of a copper chelating agent tetraethylpentamine and this strategy appeared to enhance single UCB engraftment in a phase I/II clinical trial [23]. A number of preclinical studies have provided promising pathways to expand UCB. A purine molecule name StemRegenin1 (SR1) was identified in an unbiased screen of 100,000 compounds to expand CD34+ cells. UCB cells treated with SR1 demonstrated a 50-fold increase in CD34+ cells and a 17-fold increase in UCB cells that repopulate immunodeficient mice and may be one promising agent to expand UCB [24]. In another approach, human umbilical vein endothelial cells (HUVECs) were infected with a

(8)

lentivirus encoding the E4ORF1 gene of adenoviruses to permit long term culture and survival of the infected HUVECs (E4ECs) in serum free/growth factor free conditions [25]. Co-culture of UCB CD34+ cells with the E4ECs in direct contact with minimal concentrations of hematopoietic cytokines resulted in a 150-fold expansion of hematopoietic progenitor cells that was 3-fold greater than the hematopoietic cytokines alone. Significantly greater primary and secondary engraftment of the expanded UCB CD34+ cells into immunodeficient mice was observed in the cells expanded on the E4ECs than in cultures with hematopoietic cytokines alone [25]. Another group has identified CD146+ perivascular mesenchymal cells derived from human adult adipose tissue to provide in vitro co-culture support to expand the number of UCB CD34+ cells and to enhance engraftment into immunodeficient mice [26]. Other approaches have been recently reviewed [17]. In sum, a host of approaches are seeking to expand the stem cell pool within the UCB that may permit greater utilization of UCB as an alternative stem cell source for adult subjects that lack suitable donors.

Methods to enhance UCB homing and engraftment

One way to enhance the seeding of UCB cells into the bone marrow niches that are specialized for stem cells, is to directly inject the UCB into the bone marrow cells [27]. Time to recovery of neutrophil and platelet counts has recently been shown to be significantly shortened in a comparison of patients undergoing an intra-bone UCB transplant versus a double UCB transplant [28]. A significant reduction in acute GVHD was also reported in patients receiving the intra-bone UCB transplant [28]. However, only a prospective study that employs a homogenous conditioning regimen and GVHD prophylaxis will permit determination of whether the intra-bone UCB versus double UCB transplantation approach provides a better outcome.

An alternative choice for enhancing UCB transplant outcomes would be to enhance the homing of the cells into the marrow niche. A stable prostaglandin E2 (PGE2) derivative 16,16-dimethyl PGE2 (dmPGE2) has been shown to enhance hematopoietic stem cell (HSC)

(9)

engraftment with some evidence for increased HSC homing and survival [29-32]. In a phase I trial recently completed, dmPGE2 treatment of a single UCB unit when administered with an unmanipulated UCB unit led to accelerated neutrophil recovery compared to historical controls and the dmPGE2 treated UCB unit outcompeted the unmanipulated UCB unit long term in 10 of 12 subjects [33]. Based upon these results, the authors are proposing additional clinical trials of dmPGE2 to enhance engraftment of UCB and even autologous adult peripheral blood stem cells for transplantation.

Homing of UCB cells may also be enhanced by treating the cells with fucosyltransferase-VI. UCB CD34+ cells displaying greater cell surface fucosylation displayed more rapid and higher levels of engraftment in immunodeficient mouse models compared to untreated UCB cells [34]. These results have spawned a human clinical trial in which patients will receive a double UCB unit transplant in which one of the units is fucosylated [18].

Dipeptidylpeptidase 4 (DPP4) is expressed on the cell surface of many cells, but inhibition of DPP4 on HSC enhances engraftment in preclinical studies [35]. In a recent phase I trial, patients undergoing a single unit UCB transplant were administered sitagliptin (DPP4 inhibitor) orally prior to the UCB cell infusion. Systemic DPP4 inhibition was achieved and was well tolerated. A correlation between DPP4 activity time curve and time to engraftment was observed. Optimization of the DPP4 inhibition through manipulation of the oral agent may permit improved outcomes [36].

UCB use for non-hematological disorders

While the above discussion has outlined the important role UCB can play as a source of cells to repopulate the hematopoietic system in patients requiring such a therapy, UCB has been demonstrated to contain a variety of other cell types with stem cell properties [37]. Mesenchymal stem/stromal cells (MSCs), multipotent adult progenitor cells, unrestricted somatic stem cells, and endothelial colony forming cells have all been isolated and

(10)

cryopreserved from UCB [38]. A recent analysis of clinical trials using UCB for non-hematologic disorders has identified 31 unique trials for 15 different clinical conditions (Table) [38]. Essentially all of the clinical trials have arisen from preclinical data obtained using animal models of human disease and in many cases, UCB or UCB-derived stem cell populations as therapeutic agents. Great interest has arisen over the potential use of autologous UCB to treat human neonates that suffer hypoxic-ischemic injury [39]. Human clinical trials using this approach are underway, however, no reports of the outcomes of these trials are yet available [40].

UCB banking ongoing controversies

UCB continues to be banked largely in two separate systems: public or private cord blood banks. Public banks incur all the costs for collection, transportation, processing, testing, and cryopreservation. The inventory of the UCB units is listed through national and international registries and is intended to provide patients with a high quality unit if needed and matched. If a unit is selected for transplant, the public bank is compensated as a cost-recovery. Private cord blood banks charge a fee to parents for the collection, transportation, processing, testing, and cryopreservation of the material and some type of annual fee for the ongoing storage of the UCB unit. A comparison of the public and private banking processes has recently been published [41].

Some controversy continues to exist around the cost-effectiveness of the private UCB banking system. One study has argued that use of autologous UCB units from private cord blood banks is extremely rare compared to use of allogeneic units from public banks and overall this leads to a much higher cost per unit used from the private over public banks [4, 42]. Furthermore, given the fact that most private banks are not subject to the same regulatory review as public banks, one cannot be assured that the UCB stored is of comparable quantity and quality when comparing private to public units. Hybrid UCB banking models have been

(11)

considered but also suffer from many ethical, regulatory, economic, and social concerns [4]. However, hybrid UCB banking has been selected as a preferred model among some current and potential UCB donors [43]. Some challenges persist in developing directed-family cord blood banking for those families who are pregnant and have an existing child or a known risk for producing a child affected by a disease that could be cured by allogeneic HSC transplantation [4].

Conclusion

Cord blood transplantation and banking have matured greatly in the first 25 years of application. It is obvious that cord blood banking models continue to be controversial and may continue to morph as the need for stored units increases worldwide. How UCB expansion technologies impact patient outcomes and usage may also factor into banking demands. As new fields of regenerative medicine test UCB as a potential source of reparative stem cells, even greater demand may be placed on the banking industry. It is anticipated that cord blood transplantation and banking will continue to advance along with the scientific progress.

(12)

Acknowledgements

(13)

References

**1. Ballen KK, Gluckman E, Broxmeyer HE. Umbilical cord blood transplantation: the first 25 years and beyond. Blood. 2013;122(4):491-8.

Overview of the field of cord blood banking with suggested areas for continuing improvement. Provides concise review of the first patient to undergo a cord blood transplant and descibes how the field quickly emerged and evolved to the current state. 2. Gluckman E, Broxmeyer HA, Auerbach AD, Friedman HS, Douglas GW, Devergie A, et

al. Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical-cord blood from an HLA-identical sibling. The New England Journal of Medicine. 1989;321(17):1174-8.

3. Rubinstein P, Adamson JW, Stevens C. The Placental/Umbilical Cord Blood Program of the New York Blood Center. A Progress Report. Annals of the New York Academy of Sciences. 1999;872:328-34; discussion 34-5.

4. Gluckman E, Ruggeri A, Rocha V, Baudoux E, Boo M, Kurtzberg J, et al. Family-directed umbilical cord blood banking. Haematologica. 2011;96(11):1700-7.

5. Kurtzberg J, Laughlin M, Graham ML, Smith C, Olson JF, Halperin EC, et al. Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients. The New England Journal of Medicine. 1996;335(3):157-66.

6. Rocha V, Wagner JE, Jr., Sobocinski KA, Klein JP, Zhang MJ, Horowitz MM, 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. The New England Journal of Medicine. 2000;342(25):1846-54.

7. Herr AL, Kabbara N, Bonfim CM, Teira P, Locatelli F, Tiedemann K, et al. Long-term follow-up and factors influencing outcomes after related HLA-identical cord blood

(14)

transplantation for patients with malignancies: an analysis on behalf of Eurocord-EBMT. Blood. 2010;116(11):1849-56.

8. Eapen M, Rubinstein P, Zhang MJ, Stevens C, Kurtzberg J, Scaradavou A, 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(9577):1947-54. 9. Wagner JE, Eapen M, Carter SL, Haut PR, Peres E, Schultz KR, et al. No Survival

Advantage After Double Umbilical Cord Blood (UCB) Compared to Single UCB Transplant in Children with Hematological Malignancy: Results of the Blood and Marrow Transplant Clinical Trials Network (BMT CTN 0501) Randomized Trial. ASH Annual Meeting Abstracts. 2012;120(21):359.

10. Laughlin MJ, Barker J, Bambach B, Koc ON, Rizzieri DA, Wagner JE, et al. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors. The New England Journal of Medicine. 2001;344(24):1815-22.

11. Cohen YC, Scaradavou A, Stevens CE, Rubinstein P, Gluckman E, Rocha V, et al. Factors affecting mortality following myeloablative cord blood transplantation in adults: a pooled analysis of three international registries. Bone Marrow Transplantation. 2011;46(1):70-6.

*12. Ooi J, Takahashi S, Tomonari A, Tsukada N, Konuma T, Kato S, et al. Unrelated cord blood transplantation after myeloablative conditioning in adults with acute myelogenous leukemia. Biology of blood and marrow transplantation : Journal of the American Society for Blood and Marrow Transplantation. 2008;14(12):1341-7.

Provides a brief overview of changes in cord blood unit selection and methods to expand cord blood stem cells for transplantation.

*13. Cutler C, Ballen KK. Improving outcomes in umbilical cord blood transplantation: State of the Art. Blood Reviews. 2012;26(6):241-6.

(15)

Provides a brief overview of changes in cord blood unit selection and methods to expand cord blood stem cells for transplantation.

14. Cutler C, Stevenson K, Kim HT, Brown J, McDonough S, Herrera M, et al. Double umbilical cord blood transplantation with reduced intensity conditioning and sirolimus-based GVHD prophylaxis. Bone Marrow Transplantation. 2011;46(5):659-67.

15. Barker JN, Byam C, Scaradavou A. How I treat: the selection and acquisition of unrelated cord blood grafts. Blood. 2011;117(8):2332-9.

16. Sauter C, Abboud M, Jia X, Heller G, Gonzales AM, Lubin M, et al. Serious infection risk and immune recovery after double-unit cord blood transplantation without antithymocyte globulin. Biology of blood and marrow transplantation : Journal of the American Society for Blood and Marrow Transplantation. 2011;17(10):1460-71.

*17. Broxmeyer HE. Enhancing engraftment of cord blood cells via insight into the biology of stem/progenitor cell function. Annals of the New York Academy of Sciences. 2012;1266:151-60.

Provides an overview of multiple strategies for expanding cord blood stem cells primarily in preclinical model systems.

**18. Delaney C, Bollard CM, Shpall EJ. Cord blood graft engineering. Biology of blood and marrow transplantation : Journal of the American Society for Blood and Marrow Transplantation. 2013;19(1 Suppl):S74-8.

An outstanding overview of the current approaches to expanding cord blood stem cells ex vivo to enhance in vivo engraftment.

19. Delaney C, Varnum-Finney B, Aoyama K, Brashem-Stein C, Bernstein ID. 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(8):2693-9.

(16)

20. Delaney C, Heimfeld S, Brashem-Stein C, Voorhies H, Manger RL, Bernstein ID. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nature Medicine. 2010;16(2):232-6.

21. Robinson SN, Ng J, Niu T, Yang H, McMannis JD, Karandish S, et al. Superior ex vivo cord blood expansion following co-culture with bone marrow-derived mesenchymal stem cells. Bone Marrow Transplantation. 2006;37(4):359-66.

*22. de Lima M, McNiece I, Robinson SN, Munsell M, Eapen M, Horowitz M, et al. Cord-blood engraftment with ex vivo mesenchymal-cell coculture. The New England Journal of Medicine. 2012;367(24):2305-15.

First human clinical trial to demonstrate improved engraftment neutrophils and platelet times for cord blood stem cells expanded on mesenchymal cells and then transplanted into adult patients.

23. de Lima M, McMannis J, Gee A, Komanduri K, Couriel D, Andersson BS, et al. Transplantation of ex vivo expanded cord blood cells using the copper chelator tetraethylenepentamine: a phase I/II clinical trial. Bone Marrow Transplantation. 2008;41(9):771-8.

24. Boitano AE, Wang J, Romeo R, Bouchez LC, Parker AE, Sutton SE, et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 2010;329(5997):1345-8.

*25. Butler JM, Gars EJ, James DJ, Nolan DJ, Scandura JM, Rafii S. Development of a vascular niche platform for expansion of repopulating human cord blood stem and progenitor cells. Blood. 2012;120(6):1344-7.

Novel preclinical study approach to expanding cord blood stem cells using modified human umbilical vein endothelial cells as a supportive monolayer.

26. Corselli M, Chin CJ, Parekh C, Sahaghian A, Wang W, Ge S, et al. Perivascular support of human hematopoietic stem/progenitor cells. Blood. 2013;121(15):2891-901.

(17)

27. Frassoni F, Gualandi F, Podesta M, Raiola AM, Ibatici A, Piaggio G, et al. Direct intrabone transplant of unrelated cord-blood cells in acute leukaemia: a phase I/II study. The Lancet Oncology. 2008;9(9):831-9.

*28. Rocha V, Labopin M, Ruggeri A, Podesta M, Gallamini A, Bonifazi F, et al. Unrelated cord blood transplantation: outcomes after single-unit intrabone injection compared with double-unit intravenous injection in patients with hematological malignancies. Transplantation. 2013;95(10):1284-91.

Report on the results of a direct comparison of cord blood cells administered via direct intra-bone injections versus a double cord blood unit transplant. Discusses how the present results need to be further evaluated using more standard approaches to patient care and transplantation protocols.

29. Goessling W, Allen RS, Guan X, Jin P, Uchida N, Dovey M, et al. Prostaglandin E2 enhances human cord blood stem cell xenotransplants and shows long-term safety in preclinical nonhuman primate transplant models. Cell Stem Cell. 2011;8(4):445-58. 30. Hoggatt J, Singh P, Sampath J, Pelus LM. Prostaglandin E2 enhances hematopoietic

stem cell homing, survival, and proliferation. Blood. 2009;113(22):5444-55.

31. North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature. 2007;447(7147):1007-11.

32. Pelus LM, Hoggatt J, Singh P. Pulse exposure of haematopoietic grafts to prostaglandin E2 in vitro facilitates engraftment and recovery. Cell Proliferation. 2011;44 Suppl 1:22-9. *33. Cutler C, Multani P, Robbins D, Kim HT, Le T, Hoggatt J, et al. Prostaglandin-modulated

umbilical cord blood hematopoietic stem cell transplantation. Blood. 2013;122(17):3074-81.

Very recent report on the improved engraftment provided by prostaglandin treated cord blood stem cells upon transplantation into human subjects.

(18)

34. Robinson SN, Simmons PJ, Thomas MW, Brouard N, Javni JA, Trilok S, et al. Ex vivo fucosylation improves human cord blood engraftment in NOD-SCID IL-2Rgamma(null) mice. Experimental Hematology. 2012;40(6):445-56.

35. Christopherson KW, 2nd, Hangoc G, Mantel CR, Broxmeyer HE. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science. 2004;305(5686):1000-3.

36. Farag SS, Srivastava S, Messina-Graham S, Schwartz J, Robertson MJ, Abonour R, et al. In vivo DPP-4 inhibition to enhance engraftment of single-unit cord blood transplants in adults with hematological malignancies. Stem cells and development. 2013;22(7):1007-15.

*37. Pelosi E, Castelli G, Testa U. Human umbilical cord is a unique and safe source of various types of stem cells suitable for treatment of hematological diseases and for regenerative medicine. Blood Cells, Molecules & Diseases. 2012;49(1):20-8.

Concise overview of various stem cell populations for multiple lineages of cells that may be useful in regenerative medicine treatments.

**38. Ilic D, Miere C, Lazic E. Umbilical cord blood stem cells: clinical trials in non-hematological disorders. British Medical Bulletin. 2012;102:43-57.

Outstanding concise overview of the clinical trials of umbilical cord blood cells for non-hematologic conditions.

*39. Carroll J. Human cord blood for the hypoxic-ischemic neonate. Pediatric Research. 2012;71(4 Pt 2):459-63.

Presents evidence from preclinical studies performed with umbilical cord blood in rodents exposed to hypoxia-ischemia that supports consideration of human clinical trials. *40. Liao Y, Cotten M, Tan S, Kurtzberg J, Cairo MS. Rescuing the neonatal brain from hypoxic injury with autologous cord blood. Bone Marrow Transplantation. 2013;48(7):890-900.

(19)

Timely analysis of the results of preclinical animal models of human hypoxia-ischemia and provides rationale for considering human clinical trials of autologous cord blood transplantation for neonates with hypoxic-ischemic injury.

41. Guindi, E.S. in Broxmeyer, H. E.(ed), Cord Blood: Biology, Transplantation, Banking, and Regulation. Bethesda, MD. AABB Press, 2011, pp. 595-631.

42. Rosenthal J, Woolfrey AE, Pawlowska A, Thomas SH, Appelbaum F, Forman S. Hematopoietic cell transplantation with autologous cord blood in patients with severe aplastic anemia: an opportunity to revisit the controversy regarding cord blood banking for private use. Pediatric Blood & Cancer. 2011;56(7):1009-12.

43. Wagner AM, Krenger W, Suter E, Ben Hassem D, Surbek DV. High acceptance rate of hybrid allogeneic-autologous umbilical cord blood banking among actual and potential Swiss donors. Transfusion. 2013;53(7):1510-9.

(20)

Table. Clinical trials using UCB stem cells in therapy of non-hematological disorders.

Condition ClinicalTrials.gov Identifier No

Alzheimer’s disease NCT01297218 1

Autism NCT01343511 1

Bronchopulmonary dysplasia NCT01297205 1

Burns NCT01443689 1

Cartilage injury, osteoarthritis NCT01041001 1

Cerebral palsy NCT01072370; NCT01193660 2

Critical limb ischemia NCT01019681 1

Diabetes NCT01350219; NCT01415726; NCT00873925;

NCT00989547

4

Epidermolysis bullosa NCT01033552; NCT00881556; NCT00478244 3

Hearing loss NCT01343394 1

Inborn metabolic disorders NCT00950846a; NCT00920972a; NCT01238328;

NCT00668564;NCT00654433; NCT00383448; NCT00176917; NCT00176904 8 Osteopetrosis NCT00775931; NCT01087398; NCT00638820 3 Solid tumors NCT00436761; NCT00112645 2 Stroke NCT01438593 1

SLE, systemic sclerosis NCT00684255 1

SLE, systemic lupus erythematosus.

a

Clinical trials that involve both hematological and non-hematological disorders.

reprinted with permission from Ilic D, et al. Umbilical cord blood stem cells: clinical trials in non-hematological disorders. British medical bulletin. 2012;102:43-57.

(21)

LWW Copyright Transfer and Disclosure Form

(22)

Table Copyright Permission

References

Related documents

Spleen cells from mice vaccinated with the three different vaccine formulations produced significant levels of IL-2 (Figure 1A) and IFN-c (Figure 1B) after in vitro stimulation

Apart from the methods of public offering or sale to qualified investors, it is allowed that Asset or Mort- gage-Backed Securities may be sold by private place- ment provided

Greedy Perimeter Stateless Routing (GPSR)[1] is a geographic or location based rout- ing protocol that uses the geographic positions of routers and packets destination to make

Students were asked to further rank the tools they have used in the last 6 months to contact their teachers, 37% identified their usage of face to face consultation whilst email

18Mcmfi govms &amp; ~r~on of the computer within one cycle of the computer’s internal clock; it is part of the computers oper~~g sYstem. @W@~ility of mic~e WM upheld in NEC COT.

To meet the aims of this review, a study had to meet the following criteria: (1) use a randomized controlled design; (2) utilize a nontreatment, treatment as usual,

Its principal advantages relative to these instruments are the inclusion of scales devoted to the diagnosis of the personality disorders included within the fourth edition of

Data collection methods will depend on the type of objectives that have been identified. Most likely, objectives will fall into categories related to a) the adoption of smoke-free