Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Umbilical cord blood graft engineering: challenges and opportunities

Abstract

We are entering a very exciting era in umbilical cord blood transplantation (UCBT), where many of the associated formidable challenges may become treatable by ex vivo graft manipulation and/or adoptive immunotherapy utilizing specific cellular products. We envisage the use of double UCBT rather than single UCBT for most patients; this allows for greater ability to treat larger patients as well as to manipulate the graft. Ex vivo expansion and/or fucosylation of one cord will achieve more rapid engraftment, minimize the period of neutropenia and also give certainty that the other cord will provide long-term engraftment/immune reconstitution. The non-expanded (and future dominant) cord could be chosen for characteristics such as better HLA matching to minimize GvHD, or larger cell counts to enable part of the unit to be utilized for the development of specific cellular therapies such as the production of virus-specificT-cells or chimeric-antigen receptor T-cells which are reviewed in this study.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Similar content being viewed by others

References

  1. 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. N Engl J Med 1989; 321: 1174–1178.

    Article  CAS  PubMed  Google Scholar 

  2. Rocha V, Cornish J, Sievers EL, Filipovich A, Locatelli F, Peters C et al. Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia. Blood 2001; 97: 2962–2971.

    CAS  PubMed  Google Scholar 

  3. 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. N Engl J Med 2001; 344: 1815–1822.

    Article  CAS  PubMed  Google Scholar 

  4. Ballen KK, Gluckman E, Broxmeyer HE . Umbilical cord blood transplantation: the first 25 years and beyond. Blood 2013; 122: 491–498.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Brunstein CG, Gutman JA, Weisdorf DJ, Woolfrey AE, Defor TE, Gooley TA et al. Allogeneic hematopoietic cell transplantation for hematologic malignancy: relative risks and benefits of double umbilical cord blood. Blood 2010; 116: 4693–4699.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Henslee-Downey PJ, Abhyankar SH, Parrish RS, Pati AR, Godder KT, Neglia WJ et al. Use of partially mismatched related donors extends access to allogeneic marrow transplant. Blood 1997; 89: 3864–3872.

    CAS  PubMed  Google Scholar 

  7. 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. N Engl J Med 2000; 342: 1846–1854.

    CAS  PubMed  Google Scholar 

  8. Eapen M, Rocha V, Sanz G, Scaradavou A, Zhang MJ, Arcese W et al. Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis. Lancet Oncol 2010; 11: 653–660.

    PubMed  PubMed Central  Google Scholar 

  9. Brunstein CG, Barker JN, Weisdorf DJ, DeFor TE, Miller JS, Blazar BR et al. Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease. Blood 2007; 110: 3064–3070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gluckman E, Rocha V, Boyer-Chammard A, Locatelli F, Arcese W, Pasquini R 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.

    CAS  PubMed  Google Scholar 

  11. Rubinstein P, Carrier C, Scaradavou A, Kurtzberg J, Adamson J, Migliaccio AR et al. Outcomes among 562 recipients of placental-blood transplants from unrelated donors. N Engl J Med 1998; 339: 1565–1577.

    Article  CAS  PubMed  Google Scholar 

  12. 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: 1947–1954.

    Article  PubMed  Google Scholar 

  13. Wagner JE, Barker JN, DeFor TE, Baker KS, Blazar BR, Eide C et al. Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood 2002; 100: 1611–1618.

    CAS  PubMed  Google Scholar 

  14. Verneris MR, Brunstein CG, Barker J, MacMillan ML, DeFor T, McKenna DH et al. Relapse risk after umbilical cord blood transplantation: enhanced graft-versus-leukemia effect in recipients of 2 units. Blood 2009; 114: 4293–4299.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Petersdorf EW, Hansen JA, Martin PJ, Woolfrey A, Malkki M, Gooley T et al. Major-histocompatibility-complex class I alleles and antigens in hematopoietic-cell transplantation. N Engl J Med 2001; 345: 1794–1800.

    CAS  PubMed  Google Scholar 

  16. Rocha V, Gluckman E, Eurocord-Netcord r, European B, Marrow Transplant g. Improving outcomes of cord blood transplantation: HLA matching, cell dose and other graft- and transplantation-related factors. Br J Haematol 2009; 147: 262–274.

    CAS  PubMed  Google Scholar 

  17. MacMillan ML, Weisdorf DJ, Brunstein CG, Cao Q, DeFor TE, Verneris MR et al. Acute graft-versus-host disease after unrelated donor umbilical cord blood transplantation: analysis of risk factors. Blood 2009; 113: 2410–2415.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ruggeri A, Rocha V, Masson E, Labopin M, Cunha R, Absi L et al. Impact of donor-specific anti-HLA antibodies on graft failure and survival after reduced intensity conditioning-unrelated cord blood transplantation: a Eurocord, Societe Francophone d'Histocompatibilite et d'Immunogenetique (SFHI) and Societe Francaise de Greffe de Moelle et de Therapie Cellulaire (SFGM-TC) analysis. Haematologica 2013; 98: 1154–1160.

    PubMed  PubMed Central  Google Scholar 

  19. Spellman S, Bray R, Rosen-Bronson S, Haagenson M, Klein J, Flesch S et al. The detection of donor-directed, HLA-specific alloantibodies in recipients of unrelated hematopoietic cell transplantation is predictive of graft failure. Blood 2010; 115: 2704–2708.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Stevens CE, Carrier C, Carpenter C, Sung D, Scaradavou A . HLA mismatch direction in cord blood transplantation: impact on outcome and implications for cord blood unit selection. Blood 2011; 118: 3969–3978.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Barker JN, Weisdorf DJ, DeFor TE, Blazar BR, McGlave PB, Miller JS 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.

    CAS  PubMed  Google Scholar 

  22. Ballen KK, Spitzer TR, Yeap BY, McAfee S, Dey BR, Attar E et al. Double unrelated reduced-intensity umbilical cord blood transplantation in adults. Biol Blood Marrow Transplant 2007; 13: 82–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Gutman JA, Turtle CJ, Manley TJ, Heimfeld S, Bernstein ID, Riddell SR et al. Single-unit dominance after double-unit umbilical cord blood transplantation coincides with a specific CD8+ T-cell response against the nonengrafted unit. Blood 2010; 115: 757–765.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Majhail NS, Brunstein CG, Wagner JE . Double umbilical cord blood transplantation. Curr Opin Immunol 2006; 18: 571–575.

    CAS  PubMed  Google Scholar 

  25. Smith AR, Wagner JE . Alternative haematopoietic stem cell sources for transplantation: place of umbilical cord blood. Br J Haematol 2009; 147: 246–261.

    PubMed  PubMed Central  Google Scholar 

  26. Laughlin MJ, Eapen M, Rubinstein P, Wagner JE, Zhang MJ, Champlin RE et al. Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia. N Engl J Med 2004; 351: 2265–2275.

    CAS  PubMed  Google Scholar 

  27. Locatelli F, Rocha V, Reed W, Bernaudin F, Ertem M, Grafakos S et al. Related umbilical cord blood transplantation in patients with thalassemia and sickle cell disease. Blood 2003; 101: 2137–2143.

    CAS  PubMed  Google Scholar 

  28. Rocha V, Labopin M, Sanz G, Arcese W, Schwerdtfeger R, Bosi A et al. Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia. N Engl J Med 2004; 351: 2276–2285.

    CAS  PubMed  Google Scholar 

  29. Chao NJ, Liu CX, Rooney B, Chen BJ, Long GD, Vredenburgh JJ et al. Nonmyeloablative regimen preserves ‘niches’ allowing for peripheral expansion of donor T-cells. Biol Blood Marrow Transplant 2002; 8: 249–256.

    PubMed  Google Scholar 

  30. 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 Transplant 2006; 37: 359–366.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. de Lima M, McNiece I, Robinson SN, Munsell M, Eapen M, Horowitz M et al. Cord-blood engraftment with ex vivo mesenchymal-cell coculture. N Engl J Med 2012; 367: 2305–2315.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Milner LA, Kopan R, Martin DI, Bernstein ID . A human homologue of the Drosophila developmental gene, Notch, is expressed in CD34+ hematopoietic precursors. Blood 1994; 83: 2057–2062.

    CAS  PubMed  Google Scholar 

  33. Varnum-Finney B, Xu L, Brashem-Stein C, Nourigat C, Flowers D, Bakkour S et al. Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive Notch1 signaling. Nature Med 2000; 6: 1278–1281.

    CAS  PubMed  Google Scholar 

  34. 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: 2693–2699.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ohishi K, Varnum-Finney B, Bernstein ID . Delta-1 enhances marrow and thymus repopulating ability of human CD34(+)CD38(-) cord blood cells. J Clin Invest 2002; 110: 1165–1174.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. 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 Med 2010; 16: 232–236.

    CAS  PubMed  Google Scholar 

  37. McEver RP, Moore KL, Cummings RD . Leukocyte trafficking mediated by selectin-carbohydrate interactions. J Biol Chem 1995; 270: 11025–11028.

    CAS  PubMed  Google Scholar 

  38. Xia L, McDaniel JM, Yago T, Doeden A, McEver RP . Surface fucosylation of human cord blood cells augments binding to P-selectin and E-selectin and enhances engraftment in bone marrow. Blood 2004; 104: 3091–3096.

    CAS  PubMed  Google Scholar 

  39. Hidalgo A, Frenette PS . Enforced fucosylation of neonatal CD34+ cells generates selectin ligands that enhance the initial interactions with microvessels but not homing to bone marrow. Blood 2005; 105: 567–575.

    CAS  PubMed  Google Scholar 

  40. Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J Immunol 2005; 174: 6477–6489.

    CAS  PubMed  Google Scholar 

  41. 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. Exp Hematol 2012; 40: 445–456.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kobzdej MM, Leppanen A, Ramachandran V, Cummings RD, McEver RP . Discordant expression of selectin ligands and sialyl Lewis x-related epitopes on murine myeloid cells. Blood 2002; 100: 4485–4494.

    CAS  PubMed  Google Scholar 

  43. Oran B, Hosing CM, Kebriaei P, Rezvani K, Parmar S, Shah N et al. Ex Vivo fucosylation of cord blood accelerates neutrophil and platelet engraftment. Blood 2013; 122: 691.

    Google Scholar 

  44. 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: 3074–3081.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. 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: 445–458.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Hoggatt J, Singh P, Sampath J, Pelus LM . Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood 2009; 113: 5444–5455.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Locatelli F, Rocha V, Chastang C, Arcese W, Michel G, Abecasis M 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.

    CAS  PubMed  Google Scholar 

  48. 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. N Engl J Med 1996; 335: 157–166.

    CAS  PubMed  Google Scholar 

  49. Zorn E, Kim HT, Lee SJ, Floyd BH, Litsa D, Arumugarajah S et al. Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease. Blood 2005; 106: 2903–2911.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Rieger K, Loddenkemper C, Maul J, Fietz T, Wolff D, Terpe H et al. Mucosal FOXP3+ regulatory T cells are numerically deficient in acute and chronic GvHD. Blood 2006; 107: 1717–1723.

    CAS  PubMed  Google Scholar 

  51. Rezvani K, Mielke S, Ahmadzadeh M, Kilical Y, Savani BN, Zeilah J et al. High donor FOXP3-positive regulatory T-cell (Treg) content is associated with a low risk of GVHD following HLA-matched allogeneic SCT. Blood 2006; 108: 1291–1297.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Miura Y, Thoburn CJ, Bright EC, Phelps ML, Shin T, Matsui EC et al. Association of Foxp3 regulatory gene expression with graft-versus-host disease. Blood 2004; 104: 2187–2193.

    CAS  PubMed  Google Scholar 

  53. Dong S, Maiella S, Xhaard A, Pang Y, Wenandy L, Larghero J et al. Multiparameter single-cell profiling of human CD4+FOXP3+ regulatory T-cell populations in homeostatic conditions and during graft-versus-host disease. Blood 2013; 122: 1802–1812.

    CAS  PubMed  Google Scholar 

  54. Rubtsov YP, Niec RE, Josefowicz S, Li L, Darce J, Mathis D et al. Stability of the regulatory T cell lineage in vivo. Science 2010; 329: 1667–1671.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Koreth J, Ritz J . Tregs, HSCT, and acute GVHD: up close and personal. Blood 2013; 122: 1690–1691.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Wing K, Ekmark A, Karlsson H, Rudin A, Suri-Payer E . Characterization of human CD25+ CD4+ T cells in thymus, cord and adult blood. Immunology 2002; 106: 190–199.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Hao L, Gao L, Chen XH, Zou ZM, Zhang X, Kong PY et al. Human umbilical cord blood-derived stromal cells prevent graft-versus-host disease in mice following haplo-identical stem cell transplantation. Cytotherapy 2011; 13: 83–91.

    CAS  PubMed  Google Scholar 

  58. Bresatz S, Sadlon T, Millard D, Zola H, Barry SC . Isolation, propagation and characterization of cord blood derived CD4+ CD25+ regulatory T cells. J Immunol Methods 2007; 327: 53–62.

    CAS  PubMed  Google Scholar 

  59. Godfrey WR, Spoden DJ, Ge YG, Baker SR, Liu B, Levine BL et al. Cord blood CD4(+)CD25(+)-derived T regulatory cell lines express FoxP3 protein and manifest potent suppressor function. Blood 2005; 105: 750–758.

    CAS  PubMed  Google Scholar 

  60. Lee CC, Lin SJ, Cheng PJ, Kuo ML . The regulatory function of umbilical cord blood CD4(+) CD25(+) T cells stimulated with anti-CD3/anti-CD28 and exogenous interleukin (IL)-2 or IL-15. Pediatr Allergy Immunol 2009; 20: 624–632.

    PubMed  Google Scholar 

  61. Asanuma S, Tanaka J, Sugita J, Kosugi M, Shiratori S, Wakasa K et al. Expansion of CD4(+)CD25 (+) regulatory T cells from cord blood CD4(+) cells using the common gamma-chain cytokines (IL-2 and IL-15) and rapamycin. Ann Hematol 2011; 90: 617–624.

    CAS  PubMed  Google Scholar 

  62. Yang J, Fan H, Hao J, Ren Y, Chen L, Li G et al. Amelioration of acute graft-versus-host disease by adoptive transfer of ex vivo expanded human cord blood CD4+CD25+ forkhead box protein 3+ regulatory T cells is associated with the polarization of Treg/Th17 balance in a mouse model. Transfusion 2012; 52: 1333–1347.

    CAS  PubMed  Google Scholar 

  63. Parmar S, Liu X, Tung SS, Robinson SN, Rodriguez G, Cooper LJ et al. Third-party umbilical cord blood-derived regulatory T cells prevent xenogenic graft-versus-host disease. Cytotherapy 2014; 16: 90–100.

    CAS  PubMed  Google Scholar 

  64. Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S . Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med 2002; 196: 389–399.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Nguyen VH, Zeiser R, Dasilva DL, Chang DS, Beilhack A, Contag CH et al. In vivo dynamics of regulatory T-cell trafficking and survival predict effective strategies to control graft-versus-host disease following allogeneic transplantation. Blood 2007; 109: 2649–2656.

    CAS  PubMed  Google Scholar 

  66. Nguyen VH, Shashidhar S, Chang DS, Ho L, Kambham N, Bachmann M et al. The impact of regulatory T cells on T-cell immunity following hematopoietic cell transplantation. Blood 2008; 111: 945–953.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Taylor PA, Lees CJ, Blazar BR . The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft-versus-host disease lethality. Blood 2002; 99: 3493–3499.

    CAS  PubMed  Google Scholar 

  68. Gaidot A, Landau DA, Martin GH, Bonduelle O, Grinberg-Bleyer Y, Matheoud D et al. Immune reconstitution is preserved in hematopoietic stem cell transplantation coadministered with regulatory T cells for GVHD prevention. Blood 2011; 117: 2975–2983.

    CAS  PubMed  Google Scholar 

  69. Ustun C, Miller JS, Munn DH, Weisdorf DJ, Blazar BR . Regulatory T cells in acute myelogenous leukemia: is it time for immunomodulation? Blood 2011; 118: 5084–5095.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Zhou Q, Bucher C, Munger ME, Highfill SL, Tolar J, Munn DH et al. Depletion of endogenous tumor-associated regulatory T cells improves the efficacy of adoptive cytotoxic T-cell immunotherapy in murine acute myeloid leukemia. Blood 2009; 114: 3793–3802.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Trenado A, Charlotte F, Fisson S, Yagello M, Klatzmann D, Salomon BL et al. Recipient-type specific CD4+CD25+ regulatory T cells favor immune reconstitution and control graft-versus-host disease while maintaining graft-versus-leukemia. J Clin Invest 2003; 112: 1688–1696.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S et al. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nature Med 2003; 9: 1144–1150.

    CAS  PubMed  Google Scholar 

  73. Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood 2011; 117: 1061–1070.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Saavedra S, Sanz GF, Jarque I, Moscardo F, Jimenez C, Lorenzo I et al. Early infections in adult patients undergoing unrelated donor cord blood transplantation. Bone Marrow Transplant 2002; 30: 937–943.

    CAS  PubMed  Google Scholar 

  75. Ruggeri A, Peffault de Latour R, Carmagnat M, Clave E, Douay C, Larghero J et al. Outcomes, infections, and immune reconstitution after double cord blood transplantation in patients with high-risk hematological diseases. Transplant Infect Dis 2011; 13: 456–465.

    CAS  Google Scholar 

  76. Morecki S, Gelfand Y, Nagler A, Or R, Naparstek E, Varadi G et al. Immune reconstitution following allogeneic stem cell transplantation in recipients conditioned by low intensity vs myeloablative regimen. Bone Marrow Transplant 2001; 28: 243–249.

    CAS  PubMed  Google Scholar 

  77. Brunstein CG, Weisdorf DJ, DeFor T, Barker JN, Tolar J, van Burik JA 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Kennedy-Nasser AA, Bollard CM, Myers GD, Leung KS, Gottschalk S, Zhang Y et al. Comparable outcome of alternative donor and matched sibling donor hematopoietic stem cell transplant for children with acute lymphoblastic leukemia in first or second remission using alemtuzumab in a myeloablative conditioning regimen. Biol Blood Marrow Transplant 2008; 14: 1245–1252.

    CAS  PubMed  Google Scholar 

  79. Boeckh M, Ljungman P . How we treat cytomegalovirus in hematopoietic cell transplant recipients. Blood 2009; 113: 5711–5719.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Heslop HE . How I treat EBV lymphoproliferation. Blood 2009; 114: 4002–4008.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Lindemans CA, Leen AM, Boelens JJ . How I treat adenovirus in hematopoietic stem cell transplant recipients. Blood 2010; 116: 5476–5485.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD . Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 1992; 257: 238–241.

    CAS  PubMed  Google Scholar 

  83. Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med 1995; 333: 1038–1044.

    CAS  PubMed  Google Scholar 

  84. Peggs KS, Verfuerth S, Pizzey A, Khan N, Guiver M, Moss PA et al. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet 2003; 362: 1375–1377.

    PubMed  Google Scholar 

  85. Doubrovina E, Oflaz-Sozmen B, Prockop SE, Kernan NA, Abramson S, Teruya-Feldstein J et al. Adoptive immunotherapy with unselected or EBV-specific T cells for biopsy-proven EBV+ lymphomas after allogeneic hematopoietic cell transplantation. Blood 2012; 119: 2644–2656.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 2010; 115: 925–935.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Leen AM, Myers GD, Sili U, Huls MH, Weiss H, Leung KS et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nature Med 2006; 12: 1160–1166.

    CAS  PubMed  Google Scholar 

  88. Cobbold M, Khan N, Pourgheysari B, Tauro S, McDonald D, Osman H et al. Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J Exp Med 2005; 202: 379–386.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Uhlin M, Okas M, Gertow J, Uzunel M, Brismar TB, Mattsson J . A novel haplo-identical adoptive CTL therapy as a treatment for EBV-associated lymphoma after stem cell transplantation. Cancer Immunol Immunother 2010; 59: 473–477.

    PubMed  Google Scholar 

  90. Feuchtinger T, Opherk K, Bethge WA, Topp MS, Schuster FR, Weissinger EM et al. Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood 2010; 116: 4360–4367.

    CAS  PubMed  Google Scholar 

  91. Moosmann A, Bigalke I, Tischer J, Schirrmann L, Kasten J, Tippmer S et al. Effective and long-term control of EBV PTLD after transfer of peptide-selected T cells. Blood 2010; 115: 2960–2970.

    CAS  PubMed  Google Scholar 

  92. Icheva V, Kayser S, Wolff D, Tuve S, Kyzirakos C, Bethge W et al. Adoptive transfer of epstein-barr virus (EBV) nuclear antigen 1-specific t cells as treatment for EBV reactivation and lymphoproliferative disorders after allogeneic stem-cell transplantation. J Clin Oncol 2013; 31: 39–48.

    CAS  PubMed  Google Scholar 

  93. Feuchtinger T, Matthes-Martin S, Richard C, Lion T, Fuhrer M, Hamprecht K et al. Safe adoptive transfer of virus-specific T-cell immunity for the treatment of systemic adenovirus infection after allogeneic stem cell transplantation. Br J Haematol 2006; 134: 64–76.

    PubMed  Google Scholar 

  94. Komanduri KV, St John LS, de Lima M, McMannis J, Rosinski S, McNiece I et al. Delayed immune reconstitution after cord blood transplantation is characterized by impaired thymopoiesis and late memory T-cell skewing. Blood 2007; 110: 4543–4551.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Leen AM, Christin A, Myers GD, Liu H, Cruz CR, Hanley PJ et al. Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein-Barr virus infections after haploidentical and matched unrelated stem cell transplantation. Blood 2009; 114: 4283–4292.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Leen A, Gee AP, Leung K, Martinez C, Krance RA, Liu H et al. Multi-virus-specific T-cell therapy for patients after hematopoietic stem cell and cord blood transplantation. Blood 2013; 122: 140.

    Google Scholar 

  97. Leen AM, Bollard CM, Mendizabal AM, Shpall EJ, Szabolcs P, Antin JH et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 2013; 121: 5113–5123.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Melenhorst JJ, Leen AM, Bollard CM, Quigley MF, Price DA, Rooney CM et al. Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood 2010; 116: 4700–4702.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Katari UL, Gerdemann U, Martinez C, Leung K, Carrum G, Gee AP et al. Safety and clinical efficacy of rapidly-generated virus-specific T cells with activity against Adv, EBV, CMV, HHV6 and BK virus administered after allogeneic hematopoietic stem cell transplant. Blood 2013; 122: 148.

    Google Scholar 

  100. Versluis J, Labopin M, Niederwieser D, Socie G, Schlenk RF, Milpied N et al. Prediction of non-relapse mortality in recipients of reduced intensity conditioning allo-HSCT with acute myeloid leukemia in first complete remission: integrating the seattle comorbidity index (HCT-CI) and EBMT scoring systems. ASH Annu Meet Abstr 2012; 120: 734.

    Google Scholar 

  101. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010; 116: 4099–4102.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011; 118: 4817–4828.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Porter DL, Levine BL, Kalos M, Bagg A, June CH . Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365: 725–733.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012; 119: 2709–2720.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013; 368: 1509–1518.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Micklethwaite KP, Savoldo B, Ramos CA, Lam S, Ku S, Diouf O et al. Infusion of donor-derived CD19-redirected-virus-specific T cells for b-cell malignancies relapsed after allogeneic stem cell transplant: a phase I study. Blood 2013; 122: 152.

    Google Scholar 

  107. Micklethwaite KP, Savoldo B, Hanley PJ, Leen AM, Demmler-Harrison GJ, Cooper LJ et al. Derivation of human T lymphocytes from cord blood and peripheral blood with antiviral and antileukemic specificity from a single culture as protection against infection and relapse after stem cell transplantation. Blood 2010; 115: 2695–2703.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Kebriaei PHH, Singh H, Olivares S, Figliola M, Kumar PR, Jena B et al. Adoptive immunotherapy following umbilical cord blood transplantation using the sleeping beauty system and artificial antigen presenting cells to generate donor-derived T cells expressing a CD19-specific chimeric antigen receptor. ASH Annu Meet Abstr 2013; 122: 4208.

    Google Scholar 

  109. Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 2013; 122: 4129–4139.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002; 295: 2097–2100.

    CAS  PubMed  Google Scholar 

  111. Childs RW, Berg M . Bringing natural killer cells to the clinic: ex vivo manipulation. Hematology Am Soc Hematol Educ Program 2013; 2013: 234–246.

    PubMed  PubMed Central  Google Scholar 

  112. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005; 105: 3051–3057.

    CAS  PubMed  Google Scholar 

  113. Frei GM, Persi N, Lador C, Peled A, Cohen YC, Nagler A et al. Nicotinamide, a form of vitamin B3, promotes expansion of natural killer cells that display increased in vivo survival and cytotoxic activity. ASH Annu Meet Abstr 2011; 118: 4035.

    Google Scholar 

  114. Berg M, Lundqvist A, Betters D, Childs RW . In vitro expanded NK cells have increased natural cytotoxity receptors, TRAIL and NKG2D expression, and superior tumor cytotoxicity compared to short-term IL-2-activated NK cells. ASH Annu Meet Abstr 2009; 114: 463.

    Google Scholar 

  115. Zhang H, Cui Y, Voong N, Sabatino M, Stroncek DF, Morisot S et al. Activating signals dominate inhibitory signals in CD137L/IL-15 activated natural killer cells. J Immunother 2011; 34: 187–195.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Lapteva N, Durett AG, Sun J, Rollins LA, Huye LL, Fang J et al. Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications. Cytotherapy 2012; 14: 1131–1143.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Imai C, Iwamoto S, Campana D . Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 2005; 106: 376–383.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res 2009; 69: 4010–4017.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Baird K, Fry TJ, Merchant M, Richards K, Delbrook C, Wayne AS et al. Early clinical experience of aAPC activated NK-DLI following allogeneic PBSCT in pediatric solid tumor patients. ASH Annu Meet Abstr 2012; 120: 3013.

    Google Scholar 

  120. Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM, Johnson JL et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PloS One 2012; 7: e30264.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Liu Y, Wu HW, Sheard MA, Sposto R, Somanchi SS, Cooper LJ et al. Growth and activation of natural killer cells ex vivo from children with neuroblastoma for adoptive cell therapy. Clin Cancer Res 2013; 19: 2132–2143.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Spanholtz J, Preijers F, Tordoir M, Trilsbeek C, Paardekooper J, de Witte T et al. Clinical-grade generation of active NK cells from cord blood hematopoietic progenitor cells for immunotherapy using a closed-system culture process. PloS One 2011; 6: e20740.

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Xing D, Ramsay AG, Gribben JG, Decker WK, Burks JK, Munsell M et al. Cord blood natural killer cells exhibit impaired lytic immunological synapse formation that is reversed with IL-2 ex vivo expansion. J Immunother 2010; 33: 684–696.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Gill S, Vasey AE, De Souza A, Baker J, Smith AT, Kohrt HE et al. Rapid development of exhaustion and down-regulation of eomesodermin limit the antitumor activity of adoptively transferred murine natural killer cells. Blood 2012; 119: 5758–5768.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Curti A, Ruggeri L, D'Addio A, Bontadini A, Dan E, Motta MR et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood 2011; 118: 3273–3279.

    CAS  PubMed  Google Scholar 

  126. Szmania S, Lapteva N, Garg TK, Lingo JD, Greenway AD, Bost A et al. Expanded natural killer (NK) cells for immunotherapy: fresh and made to order. ASH Annu Meet Abstr 2012; 120: 1912.

    Google Scholar 

  127. Skeate R, Singh C, Cooley S, Geller M, Northouse J, Welbig J et al. Hemolytic anemia due to passenger lymphocyte syndrome in solid malignancy patients treated with allogeneic natural killer cell products. Transfusion 2013; 53: 419–423.

    CAS  PubMed  Google Scholar 

  128. Berg M, Lundqvist A, McCoy P Jr, Samsel L, Fan Y, Tawab A et al. Clinical-grade ex vivo-expanded human natural killer cells up-regulate activating receptors and death receptor ligands and have enhanced cytolytic activity against tumor cells. Cytotherapy 2009; 11: 341–355.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Somanchi SS, Senyukov VV, Denman CJ, Lee DA . Expansion, purification, and functional assessment of human peripheral blood NK cells. J Vis Exp 2011; 2: 2540.

    Google Scholar 

  130. Somanchi SS, Somanchi A, Cooper LJ, Lee DA . Engineering lymph node homing of ex vivo-expanded human natural killer cells via trogocytosis of the chemokine receptor CCR7. Blood 2012; 119: 5164–5172.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Miller JS . Therapeutic applications: natural killer cells in the clinic. Hematology Am Soc Hematol Educ Program 2013; 2013: 247–253.

    PubMed  Google Scholar 

  132. Brandenburg S, Takahashi T, de la Rosa M, Janke M, Karsten G, Muzzulini T et al. IL-2 induces in vivo suppression by CD4(+)CD25(+)Foxp3(+) regulatory T cells. Eur J Immunol 2008; 38: 1643–1653.

    CAS  PubMed  Google Scholar 

  133. Stoklasek TA, Schluns KS, Lefrancois L . Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J Immunol 2006; 177: 6072–6080.

    CAS  PubMed  Google Scholar 

  134. Huntington ND, Legrand N, Alves NL, Jaron B, Weijer K, Plet A et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J Exp Med 2009; 206: 25–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Cooley S, Trachtenberg E, Bergemann TL, Saeteurn K, Klein J, Le CT et al. Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood 2009; 113: 726–732.

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Venstrom JM, Pittari G, Gooley TA, Chewning JH, Spellman S, Haagenson M et al. HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1. N Engl J Med 2012; 367: 805–816.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to E J Shpall.

Ethics declarations

Competing interests

LC has served as a consultant for Targazyme (formerly American Stem cells), GE Healthcare, Ferring Pharmaceuticals, Fate Therapeutics, Janssen Pharmaceuticals, and Bristol-Myers Squibb. LC owns equity/stock in Targazyme, has received lecture fees from Miltenyi Biotec and served on the Scientific Advisory Board of Cellectis. CMH has received grant support from Celgene. NDS has served on a Celgene Advisory Board and received grant support from Celgene.

Additional information

This article was published as part of a supplement, supported by WIS-CSP Foundation, in collaboration with Gilead, Milteny Biotec, Gamida cell, Adienne Pharma and Biotech, Medac hematology, Kiadis Pharma and Almog Diagnostic.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thompson, P., Rezvani, K., Hosing, C. et al. Umbilical cord blood graft engineering: challenges and opportunities. Bone Marrow Transplant 50 (Suppl 2), S55–S62 (2015). https://doi.org/10.1038/bmt.2015.97

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/bmt.2015.97

This article is cited by

Search

Quick links