1,697
Views
57
CrossRef citations to date
0
Altmetric
Reviews

Current and future approaches to treat graft failure after allogeneic hematopoietic stem cell transplantation

, &

Bibliography

  • Olsson R, Remberger M, Schaffer M, et al. Graft failure in the modern era of allogeneic hematopoietic SCT. Bone Marrow Transplant 2013;48(4):537-43
  • Schriber J, Agovi MA, Ho V, et al. Second unrelated donor hematopoietic cell transplantation for primary graft failure. Biol Blood Marrow Transplant 2010;16(8):1099-106
  • Kato M, Matsumoto K, Suzuki R, et al. Salvage allogeneic hematopoietic SCT for primary graft failure in children. Bone Marrow Transplant 2013;48:1173-8
  • Breuer S, Preuner S, Fritsch G, et al. Early recipient chimerism testing in the T- and NK-cell lineages for risk assessment of graft rejection in pediatric patients undergoing allogeneic stem cell transplantation. Leukemia 2012;26(3):509-19
  • Andreani M, Nesci S, Lucarelli G, et al. Long-term survival of ex-thalassemic patients with persistent mixed chimerism after bone marrow transplantation. Bone Marrow Transplant 2000;25(4):401-4
  • Lisini D, Zecca M, Giorgiani G, et al. Donor/recipient mixed chimerism does not predict graft failure in children with beta-thalassemia given an allogeneic cord blood transplantation from an HLA-identical sibling. Haematologica-the Hematol J 2008;93(12):1859-67
  • Baron F, Maris MB, Sandmaier BM, et al. Graft-versus-tumor effects after allogeneic hematopoietic cell transplantation with nonmyeloablative conditioning. J Clin Oncol 2005;23(9):1993-2003
  • Mohamedbhai SG, Edwards N, Morris EC, et al. Predominant or complete recipient T-cell chimerism following alemtuzumab-based allogeneic transplantation is reversed by donor lymphocytes and not associated with graft failure. Br J Haematol 2012;156(4):516-22
  • Mohty M, Avinens O, Faucher C, et al. Predictive factors and impact of full donor T-cell chimerism after reduced intensity conditioning allogeneic stem cell transplantation. Haematologica 2007;92(7):1004-6
  • Shaw BE, Byrne JL, Das-Gupta E, et al. The impact of chimerism patterns and predonor leukocyte infusion lymphopenia on survival following T cell-depleted reduced intensity conditioned transplants. Biol Blood Marrow Transplant 2007;13(5):550-9
  • Thomson KJ, Morris EC, Milligan D, et al. T-cell-depleted reduced-intensity transplantation followed by donor leukocyte infusions to promote graft-versus-lymphoma activity results in excellent long-term survival in patients with multiply relapsed follicular lymphoma. J Clin Oncol 2010;28(23):3695-700
  • Locatelli F, Pagliara D. Allogeneic hematopoietic stem cell transplantation in children with sickle cell disease. Pediatr Blood Cancer 2012;59(2):372-6
  • Walters MC, Patience M, Leisenring W, et al. Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anemia. Biol Blood Marrow Transplant 2001;7(12):665-73
  • Lucarelli G, Gaziev J. Advances in the allogeneic transplantation for thalassemia. Blood Rev 2008;22(2):53-63
  • Booth C, Veys P. T cell depletion in paediatric stem cell transplantation. Clin Exp Immunol 2013;172(2):139-47
  • Champlin RE, Horowitz MM, van Bekkum DW, et al. Graft failure following bone marrow transplantation for severe aplastic anemia: risk factors and treatment results. Blood 1989;73(2):606-13
  • Gyger M, Baron C, Forest L, et al. Quantitative assessment of hematopoietic chimerism after allogeneic bone marrow transplantation has predictive value for the occurrence of irreversible graft failure and graft-vs.-host disease. Exp Hematol 1998;26(5):426-34
  • Hill RS, Petersen FB, Storb R, et al. Mixed hematologic chimerism after allogeneic marrow transplantation for severe aplastic anemia is associated with a higher risk of graft rejection and a lessened incidence of acute graft-versus-host disease. Blood 1986;67(3):811-16
  • Locatelli F. Improving cord blood transplantation in children. Br J Haematol 2009;147(2):217-26
  • Rocha V, Gluckman E. Improving outcomes of cord blood transplantation: HLA matching, cell dose and other graft- and transplantation-related factors. Br J Haematol 2009;147(2):262-74
  • Storb R, Thomas ED. Allogeneic bone-marrow transplantation. Immunol Rev 1983;71:77-102
  • Bernaudin F, Socie G, Kuentz M, et al. Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease. Blood 2007;110(7):2749-56
  • Slavin S, Aker M, Shapira MY, et al. Reduced-intensity conditioning for the treatment of malignant and life-threatening non-malignant disorders. Clin Transpl 2003;275-82
  • Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases. Blood 1998;91(3):756-63
  • Khouri IF, Keating M, Korbling M, et al. Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies. J Clin Oncol 1998;16(8):2817-24
  • Focosi D, Zucca A, Scatena F. The role of anti-HLA antibodies in hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2011;17(11):1585-8
  • Nagata S, Okano S, Yonemitsu Y, et al. Critical roles of memory T cells and antidonor immunoglobulin in rejection of allogeneic bone marrow cells in sensitized recipient mice. Transplantation 2006;82(5):689-98
  • Taylor PA, Ehrhardt MJ, Roforth MM, et al. Preformed antibody, not primed T cells, is the initial and major barrier to bone marrow engraftment in allosensitized recipients. Blood 2007;109(3):1307-15
  • Xu H, Chilton PM, Tanner MK, et al. Humoral immunity is the dominant barrier for allogeneic bone marrow engraftment in sensitized recipients. Blood 2006;108(10):3611-19
  • Bacigalupo A, Socie G, Lanino E, et al. Fludarabine, cyclophosphamide, antithymocyte globulin, with or without low dose total body irradiation, for alternative donor transplants, in acquired severe aplastic anemia: a retrospective study from the EBMT-SAA working party. Haematologica-the Hematol J 2010;95(6):976-82
  • Lucarelli G, Clift RA, Galimberti M, et al. Marrow transplantation for patients with thalassemia: results in class 3 patients. Blood 1996;87(5):2082-8
  • Flomenberg N, Baxter-Lowe LA, Confer D, et al. Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome. Blood 2004;104(7):1923-30
  • Lee SJ, Klein J, Haagenson M, et al. High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation. Blood 2007;110(13):4576-83
  • Petersdorf EW, Gooley TA, Anasetti C, et al. Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient. Blood 1998;92(10):3515-20
  • Woolfrey A, Klein JP, Haagenson M, et al. HLA-C antigen mismatch is associated with worse outcome in unrelated donor peripheral blood stem cell transplantation. Biol Blood Marrow Transplant 2011;17(6):885-92
  • Fleischhauer K, Locatelli F, Zecca M, et al. Graft rejection after unrelated donor hematopoietic stem cell transplantation for thalassemia is associated with nonpermissive HLA-DPB1 disparity in host-versus-graft direction. Blood 2006;107(7):2984-92
  • Fleischhauer K, Shaw BE, Gooley T, et al. Effect of T-cell-epitope matching at HLA-DPB1 in recipients of unrelated-donor haemopoietic-cell transplantation: a retrospective study. Lancet Oncol 2012;13(4):366-74
  • Reisner Y, Ben-Bassat I, Douer D, et al. Demonstration of clonable alloreactive host T cells in a primate model for bone marrow transplantation. Proc Natl Acad Sci USA 1986;83(11):4012-15
  • Amrolia PJ, Muccioli-Casadei G, Huls H, et al. Adoptive immunotherapy with allodepleted donor T-cells improves immune reconstitution after haploidentical stem cell transplantation. Blood 2006;108(6):1797-808
  • Brunstein CG, Miller JS, Cao Q, et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood 2010;
  • Ciceri F, Bonini C, Stanghellini MT, et al. Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised phase I-II study. Lancet Oncol 2009;10(5):489-500
  • Di Ianni M, Falzetti F, Carotti A, et al. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood 2011;117(14):3921-8
  • Dodero A, Carniti C, Raganato A, et al. Haploidentical stem cell transplantation after a reduced-intensity conditioning regimen for the treatment of advanced hematologic malignancies: posttransplantation CD8-depleted donor lymphocyte infusions contribute to improve T-cell recovery. Blood 2009;113(19):4771-9
  • Pira GL, Landi F, Filippini P, et al. Negative depletion of b cells and t cells expressing the alpha beta chain of the t-cell receptor (tcr) for haploidentical stem cell transplantation. Blood 2012;120(21):2
  • 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(9577):1947-54
  • Kurtzberg J, Prasad VK, Carter SL, et al. Results of the Cord Blood Transplantation Study (COBLT): clinical outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with hematologic malignancies. Blood 2008;112(10):4318-27
  • Rocha V, Cornish J, Sievers EL, et al. Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia. Blood 2001;97(10):2962-71
  • Rubinstein P, Carrier C, Scaradavou A, et al. Outcomes among 562 recipients of placental-blood transplants from unrelated donors. N Engl J Med 1998;339(22):1565-77
  • Wagner JE, Barker JN, DeFor TE, 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(5):1611-18
  • van der Loo JC, Ploemacher RE. Marrow- and spleen-seeding efficiencies of all murine hematopoietic stem cell subsets are decreased by preincubation with hematopoietic growth factors. Blood 1995;85(9):2598-606
  • Castello S, Podesta M, Menditto VG, et al. Intra-bone marrow injection of bone marrow and cord blood cells: an alternative way of transplantation associated with a higher seeding efficiency. Exp Hematol 2004;32(8):782-7
  • Frassoni F, Gualandi F, Podesta M, et al. Direct intrabone transplant of unrelated cord-blood cells in acute leukaemia: a phase I/II study. Lancet Oncol 2008;9(9):831-9
  • Rocha V, Labopin M, Ruggeri A, 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
  • Gluckman E, Horowitz MM, Champlin RE, et al. Bone-marrow transplantation for severe aplastic-anemia - influence of conditioning and graft-versus-host disease prophylaxis regimens on outcome. Blood 1992;79(1):269-75
  • Jones RJ, Barber JP, Vala MS, et al. Assessment of aldehyde dehydrogenase in viable cells. Blood 1995;85(10):2742-6
  • Luznik L, O'Donnell PV, Fuchs EJ. Post-transplantation cyclophosphamide for tolerance induction in HLA-haploidentical bone marrow transplantation. Semin Oncol 2012;39(6):683-93
  • Bolanos-Meade J, Fuchs EJ, Luznik L, et al. HLA-haploidentical bone marrow transplantation with posttransplant cyclophosphamide expands the donor pool for patients with sickle cell disease. Blood 2012;120(22):4285-91
  • Storb R, Etzioni R, Anasetti C, et al. Cyclophosphamide combined with antithymocyte globulin in preparation for allogeneic marrow transplants in patients with aplastic anemia. Blood 1994;84(3):941-9
  • Storb R, Weiden PL, Sullivan KM, et al. Second marrow transplants in patients with aplastic anemia rejecting the first graft: use of a conditioning regimen including cyclophosphamide and antithymocyte globulin. Blood 1987;70(1):116-21
  • Horan JT, Liesveld JL, Fenton P, et al. Hematopoietic stem cell transplantation for multiply transfused patients with sickle cell disease and thalassemia after low-dose total body irradiation, fludarabine, and rabbit anti-thymocyte globulin. Bone Marrow Transplant 2005;35(2):171-7
  • Klangsinsirikul P, Carter GI, Byrne JL, et al. Campath-1G causes rapid depletion of circulating host dendritic cells (DCs) before allogeneic transplantation but does not delay donor DC reconstitution. Blood 2002;99(7):2586-91
  • Wing MG, Moreau T, Greenwood J, et al. Mechanism of first-dose cytokine-release syndrome by CAMPATH 1-H: involvement of CD16 (FcgammaRIII) and CD11a/CD18 (LFA-1) on NK cells. J Clin Invest 1996;98(12):2819-26
  • Strahm B, Locatelli F, Bader P, et al. Reduced intensity conditioning in unrelated donor transplantation for refractory cytopenia in childhood. Bone Marrow Transplant 2007;40(4):329-33
  • Ahmed N, Leung KS, Rosenblatt H, et al. Successful treatment of stem cell graft failure in pediatric patients using a submyeloablative regimen of campath-1H and fludarabine. Biol Blood Marrow Transplant 2008;14(11):1298-304
  • Weisdorf DJ, Verfaillie CM, Davies SM, et al. Hematopoietic growth factors for graft failure after bone marrow transplantation: a randomized trial of granulocyte-macrophage colony-stimulating factor (GM-CSF) versus sequential GM-CSF plus granulocyte-CSF. Blood 1995;85(12):3452-6
  • Sierra J, Terol MJ, Urbano-Ispizua A, et al. Different response to recombinant human granulocyte-macrophage colony-stimulating factor in primary and secondary graft failure after bone marrow transplantation. Exp Hematol 1994;22(7):566-72
  • Eapen M, Horowitz MM, Klein JP, et al. Higher mortality after allogeneic peripheral-blood transplantation compared with bone marrow in children and adolescents: the Histocompatibility and Alternate Stem Cell Source Working Committee of the International Bone Marrow Transplant Registry. J Clin Oncol 2004;22(24):4872-80
  • Ringden O, Labopin M, Gorin NC, et al. Treatment with granulocyte colony-stimulating factor after allogeneic bone marrow transplantation for acute leukemia increases the risk of graft-versus-host disease and death: a study from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation. J Clin Oncol 2004;22(3):416-23
  • Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature 2013;493(7432):338-45
  • Haxhinasto S, Mathis D, Benoist C. The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. J Exp Med 2008;205(3):565-74
  • Huang J, Nguyen-McCarty M, Hexner EO, et al. Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nat Med 2012;18(12):1778-85
  • Delaney C, Heimfeld S, Brashem-Stein C, et al. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med 2010;16(2):232-6
  • Ciceri F, Bregni M, Peccatori J. Innovative platforms for haploidentical stem cell transplantation: the role of unmanipulated donor graft. J Cancer 2011;2:339-40
  • Peccatori J, Clerici D, Forcina A, et al. In-vivo T-regs generation by rapamycin-mycophenolate-ATG as a new platform for GvHD prophylaxis in T-cell repleted unmanipulated haploidentical peripheral stem cell transplantation: results in 59 patients. Bone Marrow Transplant 2010;45:S3-4
  • von Reyn Cream L, Ehmann WC, Rybka WB, Claxton DF. Sirolimus in unmanipulated haploidentical cell transplantation. Bone Marrow Transplant 2008;42(11):765-6
  • Campbell TB, Basu S, Hangoc G, et al. Overexpression of Rheb2 enhances mouse hematopoietic progenitor cell growth while impairing stem cell repopulation. Blood 2009;114(16):3392-401
  • Rohrabaugh SL, Campbell TB, Hangoc G, Broxmeyer HE. Ex vivo rapamycin treatment of human cord blood CD34+ cells enhances their engraftment of NSG mice. Blood Cells Mol Dis 2011;46(4):318-20
  • Baron F, Beguin Y. Adoptive immunotherapy with donor lymphocyte infusions after allogeneic HPC transplantation. Transfusion 2000;40(4):468-76
  • Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood 1990;76(12):2462-5
  • Hoelle W, Beck JF, Dueckers G, et al. Clinical relevance of serial quantitative analysis of hematopoietic chimerism after allogeneic stem cell transplantation in children for severe aplastic anemia. Bone Marrow Transplant 2004;33(2):219-23
  • Frugnoli I, Cappelli B, Chiesa R, et al. Escalating doses of donor lymphocytes for incipient graft rejection following SCT for thalassemia. Bone Marrow Transplant 2010;45(6):1047-51
  • Guardiola P, Kuentz M, Garban F, et al. Second early allogeneic stem cell transplantations for graft failure in acute leukaemia, chronic myeloid leukaemia and aplastic anaemia. Br J Haematol 2000;111(1):292-302
  • McCann SR, Bacigalupo A, Gluckman E, et al. Graft rejection and second bone marrow transplants for acquired aplastic anaemia: a report from the Aplastic Anaemia Working Party of the European Bone Marrow Transplant Group. Bone Marrow Transplant 1994;13(3):233-7
  • Zecca M, Perotti C, Marradi P, et al. Recombinant human G-CSF-mobilized peripheral blood stem cells for second allogeneic transplant after bone marrow graft rejection in children. Br J Haematol 1996;92(2):432-4
  • Dreger P, Suttorp M, Haferlach T, et al. Allogeneic granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells for treatment of engraftment failure after bone marrow transplantation. Blood 1993;81(5):1404-7
  • Larocca A, Piaggio G, Podesta M, et al. Boost of CD34+-selected peripheral blood cells without further conditioning in patients with poor graft function following allogeneic stem cell transplantation. Haematologica 2006;91(7):935-40
  • Stachel D, Schmid I, Straka C, et al. Allogenic peripheral blood stem cell (PBSC) transplantation in two patients with graft failure. Bone Marrow Transplant 1995;16(6):839-42
  • Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284(5411):143-7
  • Di Nicola M, Carlo-Stella C, Magni M, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002;99(10):3838-43
  • Locatelli F, Maccario R, Frassoni F. Mesenchymal stromal cells, from indifferent spectators to principal actors. Are we going to witness a revolution in the scenario of allograft and immune-mediated disorders? Haematologica 2007;92(7):872-7
  • Le Blanc K, Frassoni F, Ball L, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 2008;371(9624):1579-86
  • Muguruma Y, Yahata T, Miyatake H, et al. Reconstitution of the functional human hematopoietic microenvironment derived from human mesenchymal stem cells in the murine bone marrow compartment. Blood 2006;107(5):1878-87
  • Cheng L, Qasba P, Vanguri P, Thiede MA. Human mesenchymal stem cells support megakaryocyte and pro-platelet formation from CD34(+) hematopoietic progenitor cells. J Cell Physiol 2000;184(1):58-69
  • de Lima M, McNiece I, Robinson SN, et al. Cord-blood engraftment with ex vivo mesenchymal-cell coculture. N Engl J Med 2012;367(24):2305-15
  • in 'tAnker PS, Noort WA, Kruisselbrink AB, et al. Nonexpanded primary lung and bone marrow-derived mesenchymal cells promote the engraftment of umbilical cord blood-derived CD34(+) cells in NOD/SCID mice. Exp Hematol 2003;31(10):881-9
  • Maitra B, Szekely E, Gjini K, et al. Human mesenchymal stem cells support unrelated donor hematopoietic stem cells and suppress T-cell activation. Bone Marrow Transplant 2004;33(6):597-604
  • Ball LM, Bernardo ME, Roelofs H, et al. Cotransplantation of ex vivo expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood 2007;110(7):2764-7
  • Bernardo ME, Ball LM, Cometa AM, et al. Co-infusion of ex vivo-expanded, parental MSCs prevents life-threatening acute GVHD, but does not reduce the risk of graft failure in pediatric patients undergoing allogeneic umbilical cord blood transplantation. Bone Marrow Transplant 2011;46(2):200-7
  • Meuleman N, Tondreau T, Ahmad I, et al. Infusion of mesenchymal stromal cells can aid hematopoietic recovery following allogeneic hematopoietic stem cell myeloablative transplant: a pilot study. Stem Cells Dev 2009;18(9):1247-52
  • Ringden O. Mesenchymal stromal cells as first-line treatment of graft failure after hematopoietic stem cell transplantation. Stem Cells Dev 2009;18(9):1243-6
  • Uhlin M, Sairafi D, Berglund S, et al. Mesenchymal Stem Cells Inhibit Thymic Reconstitution After Allogeneic Cord Blood Transplantation. Stem Cells Dev 2012;21(9):1409-17
  • Reisner Y, Hagin D, Martelli MF. Haploidentical hematopoietic transplantation: current status and future perspectives. Blood 2011;118(23):6006-17
  • Miller RG. An immunological suppressor cell inactivating cytotoxic T-lymphocyte precursor cells recognizing it. Nature 1980;287(5782):544-6
  • Rachamim N, Gan J, Segall H, et al. Tolerance induction by "megadose" hematopoietic transplants: donor-type human CD34 stem cells induce potent specific reduction of host anti-donor cytotoxic T lymphocyte precursors in mixed lymphocyte culture. Transplantation 1998;65(10):1386-93
  • Gur H, Krauthgamer R, Bachar-Lustig E, et al. Immune regulatory activity of CD34+ progenitor cells: evidence for a deletion-based mechanism mediated by TNF-alpha. Blood 2005;105(6):2585-93
  • Gur H, Krauthgamer R, Berrebi A, et al. Tolerance induction by megadose hematopoietic progenitor cells: expansion of veto cells by short-term culture of purified human CD34(+) cells. Blood 2002;99(11):4174-81
  • Zangi L, Klionsky YZ, Yarimi L, et al. Deletion of cognate CD8 T cells by immature dendritic cells: a novel role for perforin, granzyme A, TREM-1, and TLR7. Blood 2012;120(8):1647-57
  • Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment of T-cell-depleted haploidentical “three-loci” incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells to bone marrow inoculum. Blood 1994;84(11):3948-55
  • Aversa F, Tabilio A, Velardi A, et al. Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype. N Engl J Med 1998;339(17):1186-93
  • Handgretinger R, Klingebiel T, Lang P, et al. Megadose transplantation of purified peripheral blood CD34(+) progenitor cells from HLA-mismatched parental donors in children. Bone Marrow Transplant 2001;27(8):777-83
  • Klingebiel T, Cornish J, Labopin M, et al. Results and factors influencing outcome after fully haploidentical hematopoietic stem cell transplantation in children with very high-risk acute lymphoblastic leukemia: impact of center size: an analysis on behalf of the Acute Leukemia and Pediatric Disease Working Parties of the European Blood and Marrow Transplant group. Blood 2010;115(17):3437-46
  • Locatelli F, Pende D, Maccario R, et al. Haploidentical hemopoietic stem cell transplantation for the treatment of high-risk leukemias: how NK cells make the difference. Clin Immunol 2009;133(2):171-8
  • Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002;295(5562):2097-100
  • Locatelli F, Pende D, Mingari MC, et al. Cellular and molecular basis of haploidentical hematopoietic stem cell transplantation in the successful treatment of high-risk leukemias: role of alloreactive NK cells. Front Immunol 2013;4:15
  • Handgretinger R. Negative depletion of CD3(+) and. TcRalphabeta(+) T cells. Curr Opin Hematol 2012;19(6):434-9
  • Gordon PR, Leimig T, Mueller I, et al. A large-scale method for T cell depletion: towards graft engineering of mobilized peripheral blood stem cells. Bone Marrow Transplant 2002;30(2):69-74
  • Barfield RC, Otto M, Houston J, et al. A one-step large-scale method for T- and B-cell depletion of mobilized PBSC for allogeneic transplantation. Cytotherapy 2004;6(1):1-6
  • Urban C, Benesch M, Sovinz P, et al. Alternative donor HSCT in refractory acquired aplastic anemia - prevention of graft rejection and graft versus host disease by immunoablative conditioning and graft manipulation. Pediatr Transplant 2012;16(6):577-81
  • Chaleff S, Otto M, Barfield RC, et al. A large-scale method for the selective depletion of alphabeta T lymphocytes from PBSC for allogeneic transplantation. Cytotherapy 2007;9(8):746-54
  • Bonneville M, O'Brien RL, Born WK. Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol 2010;10(7):467-78
  • Vantourout P, Hayday A. Six-of-the-best: unique contributions of gammadelta T cells to immunology. Nat Rev Immunol 2013;13(2):88-100
  • Godder KT, Henslee-Downey PJ, Mehta J, et al. Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation. Bone Marrow Transplant 2007;39(12):751-7
  • Blazar BR, Taylor PA, Bluestone JA, Vallera DA. Murine gamma/delta-expressing T cells affect alloengraftment via the recognition of nonclassical major histocompatibility complex class Ib antigens. Blood 1996;87(10):4463-72
  • Bertaina A, Romano M, Rutella S, et al. HLA haploidentical stem cell transplantation after removal of alpha beta(+) T Lymphocytes and B Lymphocytes is an effective treatment for children with life-threatening, non-malignant disorders. Blood 2012;120:21
  • Roelofs AJ, Jauhiainen M, Monkkonen H, et al. Peripheral blood monocytes are responsible for gammadelta T cell activation induced by zoledronic acid through accumulation of IPP/DMAPP. Br J Haematol 2009;144(2):245-50
  • Coomes SM, Moore BB. Pleiotropic effects of transforming growth factor-beta in hematopoietic stem-cell transplantation. Transplantation 2010;90(11):1139-44
  • Barao I, Hanash AM, Hallett W, et al. Suppression of natural killer cell-mediated bone marrow cell rejection by CD4+CD25+ regulatory T cells. Proc Natl Acad Sci USA 2006;103(14):5460-5
  • Greenwood J, Steinman L, Zamvil SS. Statin therapy and autoimmune disease: from protein prenylation to immunomodulation. Nat Rev Immunol 2006;6(5):358-70
  • Dunn SE, Youssef S, Goldstein MJ, et al. Isoprenoids determine Th1/Th2 fate in pathogenic T cells, providing a mechanism of modulation of autoimmunity by atorvastatin. J Exp Med 2006;203(2):401-12
  • Mausner-Fainberg K, Luboshits G, Mor A, et al. The effect of HMG-CoA reductase inhibitors on naturally occurring CD4+CD25+ T cells. Atherosclerosis 2008;197(2):829-39
  • Hamadani M, Craig MD, Gibson LF, Remick SC. The evolving role of statins in hematopoietic stem and progenitor cell transplantation. Am J Blood Res 2011;1(1):57-64
  • Shimabukuro-Vornhagen A, Schlosser H, Kisner T, et al. The use of statins in hematopoietic stem cell transplantation and solid organ transplantation. Curr Stem Cell Res Ther 2013;
  • Bittencourt MC, Perruche S, Contassot E, et al. Intravenous injection of apoptotic leukocytes enhances bone marrow engraftment across major histocompatibility barriers. Blood 2001;98(1):224-30
  • Saas P, Gaugler B, Perruche S. Intravenous apoptotic cell infusion as a cell-based therapy toward improving hematopoietic cell transplantation outcome. Ann NY Acad Sci 2010;1209:118-26
  • Kleinclauss F, Perruche S, Masson E, et al. Intravenous apoptotic spleen cell infusion induces a TGF-beta-dependent regulatory T-cell expansion. Cell Death Differ 2006;13(1):41-52
  • Perruche S, Kleinclauss F, Bittencourt Mde C, et al. Intravenous infusion of apoptotic cells simultaneously with allogeneic hematopoietic grafts alters anti-donor humoral immune responses. Am J Transplant 2004;4(8):1361-5
  • Bonnefoy F, Masson E, Perruche S, et al. Sirolimus enhances the effect of apoptotic cell infusion on hematopoietic engraftment and tolerance induction. Leukemia 2008;22(7):1430-4
  • Bonnefoy F, Perruche S, Couturier M, et al. Plasmacytoid dendritic cells play a major role in apoptotic leukocyte-induced immune modulation. J Immunol 2011;186(10):5696-705
  • Nouri-Shirazi M, Guinet E. Direct and indirect cross-tolerance of alloreactive T cells by dendritic cells retained in the immature stage. Transplantation 2002;74(7):1035-44
  • Pereira MI, Paiva A. Dendritic cells in cord blood transplantation: a review. Stem Cells Int 2011;2011:539896
  • Fugier-Vivier IJ, Rezzoug F, Huang Y, et al. Plasmacytoid precursor dendritic cells facilitate allogeneic hematopoietic stem cell engraftment. J Exp Med 2005;201(3):373-83
  • Huang Y, Bozulic LD, Miller T, et al. CD8α+ plasmacytoid precursor DCs induce antigen-specific regulatory T cells that enhance HSC engraftment in vivo. Blood 2011;117(8):2494-505
  • Ophir E, Eidelstein Y, Bachar-Lustig E, et al. Induction of transplantation tolerance in haploidenical transplantation under reduced intensity conditioning: the role of ex-vivo generated donor CD8+ T cells with central memory phenotype. Best Pract Res Clin Haematol 2011;24(3):393-401

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.