Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-06-01T00:05:15.333Z Has data issue: false hasContentIssue false

Section 8 - Prevention, Detection, and Treatment of Relapse after Hematopoietic Cell Transplants

Published online by Cambridge University Press:  24 May 2017

Hillard M. Lazarus
Affiliation:
Case Western Reserve University, Ohio
Robert Peter Gale
Affiliation:
Imperial College London
Armand Keating
Affiliation:
University of Toronto
Andrea Bacigalupo
Affiliation:
Ospedale San Martino, Genoa
Reinhold Munker
Affiliation:
Louisiana State University, Shreveport
Kerry Atkinson
Affiliation:
University of Queensland
Syed Ali Abutalib
Affiliation:
Midwestern Regional Medical Center, Cancer Treatment Centers of America, Chicago
Get access
Type
Chapter
Information
Hematopoietic Cell Transplants
Concepts, Controversies and Future Directions
, pp. 201 - 246
Publisher: Cambridge University Press
Print publication year: 2000

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

William, BM, de Lima, M. Advances in conditioning regimens for older adults undergoing allogeneic stem cell transplantation to treat hematologic malignancies. Drugs & Aging. 2013;30(6):373–81.CrossRefGoogle ScholarPubMed
Hourigan, CS, McCarthy, P, de Lima, M. Back to the future! The evolving role of maintenance therapy after hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation: Journal of the American Society for Blood and Marrow Transplantation. 2014;20(2):154–63.Google Scholar
de Lima, M, Porter, DL, Battiwalla, M, Bishop, MR, Giralt, SA, Hardy, NM, et al. Proceedings from the National Cancer Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse After Hematopoietic Stem Cell Transplantation: part III. Prevention and treatment of relapse after allogeneic transplantation. Biology of Blood and Marrow Transplantation: Journal of the American Society for Blood and Marrow Transplantation. 2014;20(1):413.Google Scholar
Gisselbrecht, C, Schmitz, N, Mounier, N, Singh Gill, D, Linch, DC, Trneny, M, et al. Rituximab maintenance therapy after autologous stem-cell transplantation in patients with relapsed CD20(+) diffuse large B-cell lymphoma: final analysis of the collaborative trial in relapsed aggressive lymphoma. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2012;30(36):4462–9.CrossRefGoogle Scholar
Goldman, JM, Gale, RP. What does MRD in leukemia really mean? Leukemia: Official Journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2014;28(5):1131.CrossRefGoogle ScholarPubMed
Hourigan, CS. Next Generation MRD. Biology of Blood and Marrow Transplantation: Journal of the American Society for Blood and Marrow Transplantation. 2014;20(9):1259–60.CrossRefGoogle ScholarPubMed
Puig, N, Sarasquete, ME, Balanzategui, A, Martinez, J, Paiva, B, Garcia, H, et al. Critical evaluation of ASO RQ-PCR for minimal residual disease evaluation in multiple myeloma. A comparative analysis with flow cytometry. Leukemia. 2014;28(2):391–7.CrossRefGoogle ScholarPubMed
Bruggemann, M, Raff, T, Kneba, M. Has MRD monitoring superseded other prognostic factors in adult ALL? Blood. 2012;120(23):4470–81.Google Scholar
Richardson, SE, Khan, I, Rawstron, A, Sudak, J, Edwards, N, Verfuerth, S, et al. Risk-stratified adoptive cellular therapy following allogeneic hematopoietic stem cell transplantation for advanced chronic lymphocytic leukaemia. British Journal of Haematology. 2013;160(5):640–8.CrossRefGoogle ScholarPubMed
Pott, C, Bruggemann, M, Ritgen, M, van der Velden, VH, van Dongen, JJ, Kneba, M. MRD detection in B-cell non-Hodgkin lymphomas using Ig gene rearrangements and chromosomal translocations as targets for real-time quantitative PCR. Methods in Molecular Biology (Clifton, NJ). 2013;971:175200.CrossRefGoogle ScholarPubMed
Gimenez, E, Chauvet, M, Rabin, L, Puteaud, I, Duley, S, Hamaidia, S, et al. Cloned IGH VDJ targets as tools for personalized minimal residual disease monitoring in mature lymphoid malignancies; a feasibility study in mantle cell lymphoma by the Groupe Ouest Est d’Etude des Leucemies et Autres Maladies du Sang. British Journal of Haematology. 2012;158(2):186–97.Google Scholar
Radich, JP. Monitoring response to tyrosine kinase inhibitor therapy, mutational analysis, and new treatment options in chronic myelogenous leukemia. Journal of the National Comprehensive Cancer Network. 2013;11(5 Suppl):663–6.CrossRefGoogle ScholarPubMed
O’Donnell, MR, Tallman, MS, Abboud, CN, Altman, JK, Appelbaum, FR, Arber, DA, et al. Acute myeloid leukemia, version 2.2013. Journal of the National Comprehensive Cancer Network. 2013;11(9):1047–55.Google Scholar
Hourigan, CS, Karp, JE. Minimal residual disease in acute myeloid leukaemia. Nature Reviews Clinical Oncology. 2013;10(8):460–71.Google Scholar
Qin, YZ, Zhu, HH, Liu, YR, Wang, YZ, Shi, HX, Lai, YY, et al. PRAME and WT1 transcripts constitute a good molecular marker combination for monitoring minimal residual disease in myelodysplastic syndromes. Leukemia & Lymphoma. 2013;54(7):1442–9.Google Scholar
Grimwade, D, Freeman, SD. Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for “Prime Time”? Blood. 2014;124(23):3345–55.Google Scholar
Logan, AC, Vashi, N, Faham, M, Carlton, V, Kong, K, Buno, I, et al. Immunoglobulin and T cell receptor gene high-throughput sequencing quantifies minimal residual disease in acute lymphoblastic leukemia and predicts post-transplantation relapse and survival. Biology of Blood and Marrow Transplantation: Journal of the American Society for Blood and Marrow Transplantation. 2014;20(9):1307–13.Google Scholar
Brenner, MK, Rill, DR, Moen, RC, Krance, RA, Mirro, J Jr., Anderson, WF, et al. Gene-marking to trace origin of relapse after autologous bone-marrow transplantation. Lancet. 1993;341(8837):85–6.CrossRefGoogle ScholarPubMed
Stewart, AK, Vescio, R, Schiller, G, Ballester, O, Noga, S, Rugo, H, et al. Purging of autologous peripheral-blood stem cells using CD34 selection does not improve overall or progression-free survival after high-dose chemotherapy for multiple myeloma: results of a multicenter randomized controlled trial. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2001;19(17):3771–9.CrossRefGoogle ScholarPubMed
Schouten, HC, Qian, W, Kvaloy, S, Porcellini, A, Hagberg, H, Johnsen, HE, et al. High-dose therapy improves progression-free survival and survival in relapsed follicular non-Hodgkin’s lymphoma: results from the randomized European CUP trial. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2003;21(21):3918–27.CrossRefGoogle ScholarPubMed
Furman, RR, Grossbard, ML, Johnson, JL, Pecora, AL, Cassileth, PA, Jung, SH, et al. A phase III study of anti-B4-blocked ricin as adjuvant therapy post-autologous bone marrow transplant: CALGB 9254. Leukemia & Lymphoma. 2011; 52(4):587–96.Google Scholar
Pettengell, R, Schmitz, N, Gisselbrecht, C, Smith, G, Patton, WN, Metzner, B, et al. Rituximab purging and/or maintenance in patients undergoing autologous transplantation for relapsed follicular lymphoma: a prospective randomized trial from the lymphoma working party of the European group for blood and marrow transplantation. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2013;31(13):1624–30.CrossRefGoogle ScholarPubMed
Wetzler, M, Watson, D, Stock, W, Koval, G, Mulkey, FA, Hoke, EE, et al. Autologous transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia achieves outcomes similar to allogeneic transplantation: results of CALGB Study 10001 (Alliance). Haematologica. 2014;99(1):111–5.CrossRefGoogle ScholarPubMed
Younes, A, Bartlett, NL, Leonard, JP, Kennedy, DA, Lynch, CM, Sievers, EL, et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. The New England Journal of Medicine. 2010;363(19):1812–21.Google Scholar
Rytting, M, Triche, L, Thomas, D, O’Brien, S, Kantarjian, H. Initial experience with CMC-544 (inotuzumab ozogamicin) in pediatric patients with relapsed B-cell acute lymphoblastic leukemia. Pediatric Blood and Cancer. 2014;61(2):369–72.Google Scholar
O’Hear, C, Inaba, H, Pounds, S, Shi, L, Dahl, G, Bowman, WP, et al. Gemtuzumab ozogamicin can reduce minimal residual disease in patients with childhood acute myeloid leukemia. Cancer. 2013;119(22):4036–43.Google Scholar
de Lima, M, Giralt, S, Thall, PF, de Padua, Silva L, Jones, RB, Komanduri, K, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116(23):5420–31.CrossRefGoogle ScholarPubMed
Choi, J, Ritchey, J, Prior, JL, Holt, M, Shannon, WD, Deych, E, et al. In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood. 2010;116(1):129–39.Google Scholar
Sanchez-Abarca, LI, Gutierrez-Cosio, S, Santamaria, C, Caballero-Velazquez, T, Blanco, B, Herrero-Sanchez, C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115(1):107–21.CrossRefGoogle ScholarPubMed
Goodyear, OC, Dennis, M, Jilani, NY, Loke, J, Siddique, S, Ryan, G, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood. 2012;119(14):3361–9.Google Scholar
Garcia-Manero, G, Gore, SD, Cogle, C, Ward, R, Shi, T, Macbeth, KJ, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2011;29(18):2521–7.Google Scholar
Burchert, A, Nicolaus, K, Huenecke, S, Duenzinger, U, Wolf, A, Bader, P, et al. Post-transplant maintenance with the deacetylase inhibitor panobinostat in patients with high-risk AML or MDS: results of the phase I part of the panobest trial. Blood. 2013;122(21):3315.Google Scholar
Dhedin, N, Huynh, A, Maury, S, Tabrizi, R, Beldjord, K, Asnafi, V, et al. Role of allogeneic stem cell transplantation in adult patients with Ph-negative acute lymphoblastic leukemia. Blood. 2015;125(16):2486–96; quiz 586.Google Scholar
Walter, RB, Buckley, SA, Pagel, JM, Wood, BL, Storer, BE, Sandmaier, BM, et al. Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood. 2013;122(10):1813–21.Google Scholar
Schroeder, T, Czibere, A, Platzbecker, U, Bug, G, Uharek, L, Luft, T, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013;27(6):1229–35.Google Scholar
Thompson, JA, Fisher, RI, Leblanc, M, Forman, SJ, Press, OW, Unger, JM, et al. Total body irradiation, etoposide, cyclophosphamide, and autologous peripheral blood stem-cell transplantation followed by randomization to therapy with interleukin-2 versus observation for patients with non-Hodgkin lymphoma: results of a phase 3 randomized trial by the Southwest Oncology Group (SWOG 9438). Blood. 2008;111(8):4048–54.Google Scholar
Bolanos-Meade, J, Garrett-Mayer, E, Luznik, L, Anders, V, Webb, J, Fuchs, EJ, et al. Induction of autologous graft-versus-host disease: results of a randomized prospective clinical trial in patients with poor risk lymphoma. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2007;13(10):1185–91.Google Scholar
Simonsson, B, Totterman, T, Hokland, P, Lauria, F, Carella, AM, Fernandez, MN, et al. Roquinimex (Linomide) vs placebo in AML after autologous bone marrow transplantation. Bone Marrow Transplant. 2000;25(11):1121–7.Google Scholar
Attal, M, Harousseau, JL, Leyvraz, S, Doyen, C, Hulin, C, Benboubker, L, et al. Maintenance therapy with thalidomide improves survival in patients with multiple myeloma. Blood. 2006;108(10):3289–94.CrossRefGoogle ScholarPubMed
Barlogie, B, Pineda-Roman, M, van Rhee, F, Haessler, J, Anaissie, E, Hollmig, K, et al. Thalidomide arm of Total Therapy 2 improves complete remission duration and survival in myeloma patients with metaphase cytogenetic abnormalities. Blood. 2008;112(8):3115–21.Google Scholar
Lokhorst, HM, van der Holt, B, Zweegman, S, Vellenga, E, Croockewit, S, van Oers, MH, et al. A randomized phase 3 study on the effect of thalidomide combined with adriamycin, dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma. Blood. 2010;115(6):1113–20.CrossRefGoogle ScholarPubMed
Morgan, GJ, Davies, FE, Gregory, WM, Bell, SE, Szubert, AJ, Cook, G, et al. Long-term follow-up of MRC Myeloma IX trial: Survival outcomes with bisphosphonate and thalidomide treatment. Clin Cancer Res. 2013;19(21):6030–8.CrossRefGoogle ScholarPubMed
Spencer, A, Prince, HM, Roberts, AW, Prosser, IW, Bradstock, KF, Coyle, L, et al. Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2009;27(11):1788–93.Google Scholar
Krishnan, A, Pasquini, MC, Logan, B, Stadtmauer, EA, Vesole, DH, Alyea, E 3rd, et al. Autologous haemopoietic stem-cell transplantation followed by allogeneic or autologous haemopoietic stem-cell transplantation in patients with multiple myeloma (BMT CTN 0102): a phase 3 biological assignment trial. The Lancet Oncology. 2011;12(13):1195–203.Google Scholar
Maiolino, A, Hungria, VT, Garnica, M, Oliveira-Duarte, G, Oliveira, LC, Mercante, DR, et al. Thalidomide plus dexamethasone as a maintenance therapy after autologous hematopoietic stem cell transplantation improves progression-free survival in multiple myeloma. Am J Hematol. 2012;87(10):948–52.Google Scholar
Stewart, AK, Trudel, S, Bahlis, NJ, White, D, Sabry, W, Belch, A, et al. A randomized phase 3 trial of thalidomide and prednisone as maintenance therapy after ASCT in patients with MM with a quality-of-life assessment: the National Cancer Institute of Canada Clinicals Trials Group Myeloma 10 Trial. Blood. 2013;121(9):1517–23.CrossRefGoogle ScholarPubMed
McCarthy, PL, Owzar, K, Hofmeister, CC, Hurd, DD, Hassoun, H, Richardson, PG, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. The New England Journal of Medicine. 2012;366(19):1770–81.Google Scholar
Attal, M, Lauwers-Cances, V, Marit, G, Caillot, D, Moreau, P, Facon, T, et al. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. The New England Journal of Medicine. 2012;366(19):1782–91.CrossRefGoogle ScholarPubMed
Boccadoro, M, Cavallo, F., Gay, F. Melphalan/prednisone/lenalidomide (MPR) versus high-dose melphalan and autologous transplantation MEL200) plus lenalidomide maintenance or no maintenance in newly diagnosed multiple myeloma (MM) patients. Journal of Clinical Oncology. 2013;(Suppl): Abstract 8509.Google Scholar
Sonneveld, P, Schmidt-Wolf, IG, van der Holt, B, El Jarari, L, Bertsch, U, Salwender, H, et al. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/ GMMG-HD4 trial. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2012;30(24):2946–55.CrossRefGoogle ScholarPubMed
Cavo, M, Pantani, L, Petrucci, MT, Patriarca, F, Zamagni, E, Donnarumma, D, et al. Bortezomib-thalidomide-dexamethasone is superior to thalidomide-dexamethasone as consolidation therapy after autologous hematopoietic stem cell transplantation in patients with newly diagnosed multiple myeloma. Blood. 2012;120(1):919.Google Scholar
Mellqvist, UH, Gimsing, P, Hjertner, O, Lenhoff, S, Laane, E, Remes, K, et al. Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial. Blood. 2013;121(23):4647–54.CrossRefGoogle Scholar
Palumbo, A, Cavallo, F, Gay, F, Di Raimondo, F, Ben Yehuda, D, Petrucci, MT, et al. Autologous transplantation and maintenance therapy in multiple myeloma. The New England Journal of Medicine. 2014;371(10):895905.Google Scholar

References

Cheson, BD, Bennett, JM, Kopecky, KJ, et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol 2003;21:4642–9.Google Scholar
Freireich, EJ, Gehan, EA, Sulman, D, et al. The effect of chemotherapy on acute leukemia in the human. J Chronic Dis 1961;14:593608.Google Scholar
Bradstock, KF, Janossy, G, Tidman, N, et al. Immunological monitoring of residual disease in treated thymic acute lymphoblastic leukemia. Leuk Res 1981;5:301–9.Google Scholar
Walter, RB, Kantarjian, HM, Huang, X, et al. Effect of complete remission and responses less than complete remission on survival in acute myeloid leukemia: a combined Eastern Cooperative Oncology Group, Southwest Oncology Group, and M. D. Anderson Cancer Center Study. J Clin Oncol 2010;28:1766–71.Google Scholar
Chen, X, Xie, H, Bohm, C, et al. The relation of clinical response and minimal residual disease and their prognostic impact on outcome in acute myeloid leukemia. J Clin Oncol 2015;33:1258-64.Google Scholar
Freeman, SD, Virgo, P, Couzens, S, et al. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patient with acute myeloid leukemia. J Clin Oncol 2013;31:4123–31.Google Scholar
Kang, H, Chen, I-M, Wilson, CS, et al. Gene expression classifiers for relapse-free survival and minimal residual disease improve risk-classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia. Blood 2010;115:1394–405.Google Scholar
Yang, JJ, Cheng, C, Devidas, M, et al. Genome-wide association study identifies Germline polymorphisms associated with relapse of childhood acute lymphoblastic leukemia. Blood 2012;120:4197–204.Google Scholar
Beldjord, K, Chevret, S, Asnafi, V, et al. Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia. Blood 2014;123:3739–49.Google Scholar
Vora, A, Frost, L, Goodeve, A, et al. Late relapsing childhood lymphoblastic leukemia. Blood 1998;92:2334–7.Google Scholar
Ommen, HB, Schnittger, S, Jovanovic, JV, et al. Strikingly different molecular relapse Grimwadekinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood 2010;115:198205.Google Scholar
Parker, C, Waters, R, Leighton, C, et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukemia (ALL R3): an open-label randomized trial. The Lancet 2010;376:2009–17.Google Scholar
Butturini, A, Klein, J, Gale, RP. Modeling minimal residual disease (MRD)-testing. Leuk Res 2003;27:293300.Google Scholar
Mullighan, CG, Su, X, Zhang, J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med 2009;360:470–80.Google Scholar
van der Veer, A, Waanders, E, Pieters, R, et al. Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL. Blood 2013;122:2622–9.CrossRefGoogle Scholar
Roberts, KG, Li, Y, Payne-Turner, D, et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med 2014;371:1005–15.CrossRefGoogle ScholarPubMed
Lee, S, Kim, D-W, Cho, B-S, et al. Impact of minimal residual disease kinetics during imatinib-based treatment on transplantation outcome in Philadelphia chromosome-positive acute lymphoblastic leukemia. Leukemia 2012;26:2367–74.Google Scholar
Ravandi, F, Jorgensen, JL, Thomas, DA, et al. Detection of MRD may predict the outcome of patients with Philadelphia chromosome-positive ALL treated with tyrosine kinase inhibitors plus chemotherapy. Blood 2013;122:1214–21.Google Scholar
Jeha, S, Coustan-Smith, E, Pei, D, et al. Impact of tyrosine kinase inhibitors on minimal residual disease and outcome in childhood Philadelphia chromosome-positive acute lymphoblastic leukemia. Cancer 2014;120:1514–9.Google Scholar
Helgestad, J, Rosthøj, S, Johansen, P, et al. Bone marrow aspiration technique may have an impact on therapy stratification in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2011;57:224–6.Google Scholar
Paietta, E. Minimal residual disease in AML: coming of age. Hematol Am Soc Hematol Educ Program 2012;2012:3542.Google Scholar
Paietta, E. Minimal residual disease in acute leukaemia: a guide to precision medicine ready for prime time? In Treatment Strategies Haematology, Vol 4, issue 2, 2014, pp 45-48.Google Scholar
Grimwade, D, Freeman, SD. Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for “prime time”? Blood 2014;124:3345–55.Google Scholar
Loken, MR, Chu, S-C, Fritschle, W, et al. Normalization of bone marrow aspirates for hemodilution in flow cytometric analyses. Cytometry Part B (Clin Cytometry) 2009;76B:2736.Google Scholar
Buccisano, F, Maurillo, L, Del Principe, MI, et al. Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia. Blood 2012;119:332–41.Google Scholar
Loken, MR, Alonzo, TA, Pardo, L, et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children’s Oncology Group. Blood 2012;120:1581–8.Google Scholar
Inaba, H, Coustan-Smith, E, Cao, X, et al. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J Clin Oncol 2012;30:3625–32.Google Scholar
Campana, D, Coustan-Smith, E. Measurements of treatment response in childhood acute leukemia. Korean J Hematol 2012;47:245–54.CrossRefGoogle ScholarPubMed
McKenna, RW, Washington, LT, Aquino, DB, et al. Immunophenotypic analysis of hematogones (B-lymphocyte precursor) in 662 consecutive bone marrow specimens by 4-color flow cytometry. Blood 2001;98:2498–507.CrossRefGoogle Scholar
Sevilla, DW, Colovai, AI, Emmons, FN, et al. Hematogones: a review and update. Leuk Lymphoma 2010;51:1019.Google Scholar
Sedek, L, Bulsa, J, Sonsala, A, et al. The immunophenotypes of blast cells in B-cell precursor acute lymphoblastic leukemia: how different are they from their normal counterparts? Cytometry B Clin Cytom 2014;86:329–39.Google Scholar
Loken, MR, Alonzo, TA, Pardo, L, et al. Multidimensional flow cytometry significantly improves upon the morphologic assessment of post-induction marrow remission status-comparison of morphology and multidimensional flow cytometry: a report from the Children’s Oncology Group AML Protocol AAML0531. Blood 2012;120(8):1581–8.Google Scholar
Grimwade, D, Tallman, M. Should minimal residual disease monitoring be the standard of care for all patients with acute promyelocytic leukemia? Leuk Research 2011;35:37.Google Scholar
Wang, F, Travis, J, DeLaBarre, B, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 2013;340:622–6.Google Scholar
Gaipa, G, Cazzaniga, G, Valsecchi, MG, et al. Time point-dependent concordance of flow cytometry and real-time quantitative polymerase chain reaction for minimal residual disease detection in childhood acute lymphoblastic leukemia. Haematologica 2012;97:1582–93.CrossRefGoogle ScholarPubMed
Van der Velden, VHJ, Cazzaniga, G, Schrauder, A, et al. Analysis of minimal residual disease by Ig/TCR gene rearrangements: guidelines for interpretation of real-time quantitative PCR data. Leukemia 2007;21:604–11.CrossRefGoogle ScholarPubMed
van Dongen, JJM. EuroFlow Educational Book 2012; Rotterdam: Macmillan.Google Scholar
Boeckx, N, Willemse, MJ, Szczepanski, T, et al. Fusion gene transcripts and Ig/TCR gene rearrangements are complementary but infrequent targets for PCR-based detection of minimal residual disease in acute myeloid leukemia. Leukemia 2002;16:368–75.Google Scholar
Warren, EH, Matsen, FA IV, Chou, J. High-throughput sequencing of B- and T-lymphocyte antigen receptors in hematology. Blood 2013;122:1922.Google Scholar
Faham, M, Zheng, J, Moorhead, M, et al. Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia. Blood 2012;120:5173–80.Google Scholar
Martinez-Lopez, J, Lahuerta, JJ, Pepin, F, et al. Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood 2014;123:3073–9.Google Scholar
Wood, B. Flow cytometric assessment of MRD. Presented at the FDA Public Workshop on Minimal Residual Disease, FDA White Oak Campus, Silver Spring, MD, April 18, 2012.Google Scholar
Wood, BL. Flow cytometric monitoring of residual disease in acute leukemia. In Czader, M. (ed) Hematological Malignancies: Methods in Molecular Biology, vol 999. 2013; New York: Springer, pp. 123–36.Google Scholar
Chevallier, P, Robillard, N, Ayari, S, et al. Persistence of CD33 expression at relapse in CD33(+) acute myeloid leukaemia patients after receiving Gemtuzumab in the course of the disease. Br J Haematol 2008;143:744–6.Google Scholar
Chu, PG, Chen, YY, Molina, A, et al. Recurrent B-cell neoplasms after Rituximab therapy: an immunophenotypic and genotypic study. Leuk Lymphoma 2002;43:2335–41.Google Scholar
Topp, MS, Kufer, P, Gökbuget, N, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 2011;29:2493–8.Google Scholar
Campana, D. Should minimal residual disease monitoring in acute lymphoblastic leukemia be standard of care? Curr Hematol Malig Rep 2012;7:170–7.CrossRefGoogle ScholarPubMed
van Dongen, JJM, Lhermitte, L, Böttcher, S, et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia 2012;26:1908–75.Google Scholar
Zeijlemaker, W, Gratama, JW, Schuurhuis, GJ. Tumor heterogeneity makes AML a “moving target” for detection of residual disease. Cytometry B Clin Cytom 2014;86:314.Google Scholar
Coustan-Smith, E, Song, G, Clark, C, et al. New markers for minimal residual disease detection in acute lymphoblastic leukemia. Blood 2011;117:6267–76.Google Scholar
Langebrake, C, Brinkmann, I, Teigler-Schlegel, A, et al. Immunophenotypic differences between diagnosis and relapse in childhood AML: implications for MRD monitoring. Cytometry B Clin Cytom 2005;63B:19.Google Scholar
Macedo, A, San Miguel, JF, Vidriales, MB, et al. Phenotypic changes in acute myeloid leukemia: implications in the detection of minimal residual disease. J Clin Pathol 1996;49:1518.Google Scholar
Brϋggemann, M, Raff, T, Kneba, M. Has MRD monitoring superseded other prognostic factors in adult ALL? Blood 2012;120:4470–81.Google Scholar
Wu, D, Emerson, RO, Sherwood, A, et al. Detection of minimal residual disease in B lymphoblastic leukemia by high-throughput sequencing of IGH. Clin Cancer Res 2014;20:4540–8.Google Scholar
Thol, F, Kölking, B, Damm, F, et al. Next-generation sequencing for minimal residual disease monitoring in acute myeloid leukemia patients with FLT3-ITD or NPM1 mutations. Genes Chromosom Cancer 2012;51:689–95.Google Scholar
Bachas, C, Schuurhuis, GJ, Assaraf, YG, et al. The role of minor subpopulations within the leukemic blast compartment of AML patients at initial diagnosis in the development of relapse. Leukemia 2012;26:1313–20.Google Scholar
Hou, H-A, Kuo, Y-Y, Liu, C-Y, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood 2012;119:559–68.Google Scholar
Im, AP, Sehgal, AR, Carroll, MP, et al. DNMT3A and IDH mutations in acute myeloid leukemia and other myeloid malignancies: associations with prognosis and potential treatment strategies. Leukemia 2014;28:1774–83.Google Scholar
Salipante, SJ, Fromm, JR, Shendure, J, et al. Detection of minimal residual disease in NPM1-mutated acute myeloid leukemia by next-generation sequencing. Mod Pathol 2014;27:1438–46.CrossRefGoogle ScholarPubMed
Stow, P, Key, L, Chen, X, et al. Clinical significance of low levels of minimal residual disease at the end of remission induction therapy in childhood acute lymphoblastic leukemia. Blood 2010;115:4657–63.Google Scholar
Maurillo, L, Buccisano, F, Del Principe, MI, et al. Toward optimization of postremission therapy for residual disease-positive patients with acute myeloid leukemia. J Clin Oncol 2008;26:4944–51.Google Scholar
Rubnitz, JE, Inaba, H, Dahl, G, et al. Minimal residual disease-directed therapy for childhood acute myeloid leukemia: results of the AML02 multicentre trial. The Lancet 2010;11:543–52.Google Scholar
Terwijn, M, van Putten, WLJ, Kelder, A, et al. High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: Data from the HOVON/SAKK AML 42A study. J Clin Oncol 2013;31:3889–97.Google Scholar
Ganzel, C, Sun, Z, Gönen, M, et al. Minimal residual disease assessment by flow cytometry in AML is an independent prognostic factor even after adjusting for cytogenetic/molecular abnormalities. Blood 2014;124:1016.Google Scholar
Fernandez, HF, Sun, Z, Yao, X, et al. Anthracycline dose intensification in acute myeloid leukemia: Results of ECOG Study E1900. N Engl J Med 2009;361:1249–59.Google Scholar
Perea, G, Lasa, A, Aventin, A, et al. Prognostic value of minimal residual disease (MRD) in acute myeloid leukemia (AML) with favorable cytogenetics [t(8;21) and inv(16)]. Leukemia 2006;20:8794.Google Scholar
Miyamoto, T, Weissman, IL, Akashi, K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci USA 2000;97:7521–6.Google Scholar
Corces-Zimmerman, MR, Hong, W-J, Weissman, IL, et al. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci USA 2014;111:2548–53.Google Scholar
Shlush, LI, Zandi, S, Mitchell, A, et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 2014;506:328–33.Google Scholar
Bodini, M, Ronchini, C, Giacò, L, et al. The hidden genomic landscape of acute myeloid leukemia: subclonal structure revealed by undetected mutations. Blood 2015;125:600–5.Google Scholar
Will, B, Steidl, U. Multi-parameter fluorescence-activated cell sorting and analysis of stem and progenitor cells in myeloid malignancies. Best Pract Res Clin Haematol 2010;23:391401.Google Scholar
Ebinger, M, Witte, K-E, Ahlers, J, et al. High frequency of immature cells at diagnosis predicts high minimal residual disease level in childhood acute lymphoblastic leukemia. Leuk Res 2010;34:1139–42.Google Scholar
Terwijn, M, Kelder, A, Snel, AN, et al. Minimal residual disease detection defined as the malignant fraction of the total primitive stem cell compartment offers additional prognostic information in acute myeloid leukemia. Int J Lab Hem 2012;34:432–41.Google Scholar
Gerber, JM, Smith, BD, Ngwang, B, et al. A clinically relevant population of leukemic CD34+CD38- cells in acute myeloid leukemia. Blood 2012;119:3571–7.Google Scholar
Roug, AS, Larsen, , Nederby, L, et al. hMICL and CD123 in combination with a CD45/CD34/CD117 backbone – a universal marker combination for the detection of minimal residual disease in acute myeloid leukemia. Br J Haematol 2014;164:212–22.Google Scholar
DiGiuseppe, JA. CD34+/CD38- cells and minimal residual disease in childhood lymphoblastic leukemia. Leuk Res 2010;34:1125–6.Google Scholar
Aoki, Y, Watanabe, T, Saito, Y, et al. Identification of CD34+ and CD34- leukemia-initiating cells in MLL-rearranged human acute lymphoblastic leukemia. Blood 2014;125:967–80.Google Scholar
Mullighan, CG, Phillips, LA, Su, X, et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science 2008;322:1377–80.Google Scholar
Notta, F, Mullighan, CG, Wang, JC, et al. Evolution of human BCR-ABL1 lymphoblastic leukaemia-initiating cells. Nature 2011;469:362–7.Google Scholar
Clappier, E, Gerby, B, Sigaux, F, et al. Clonal selection in xenografted human T cell acute lymphoblastic leukemia recapitulates gain of malignancy at relapse. J Exp Med 2011;208:653–61.Google Scholar
Klco, JM, Spencer, DH, Miller, CA, et al. Functional heterogeneity of genetically defined subclones in acute myeloid leukemia. Cancer Cell 2014;25:379–92.Google Scholar
Inaba, H, Greaves, M, Mullighan, CG. Acute lymphoblastic leukemia. Lancet 2013;381:1943–55.Google Scholar
Chan, SM, Majeti, R. Role of DNMT3A, TET2, and IDH1/2 mutations in pre-leukemic stem cells in acute myeloid leukemia. Int J Hematol 2013;98:648–57.Google Scholar
Peloquin, GL, Chen, YB, Fathi, AT. The evolving landscape in the therapy of acute myeloid leukemia. Protein Cell 2013;4:735–46.Google Scholar
Li, KK, Luo, LF, Shen, Y, et al. DNA methyltransferases in hematologic malignancies. Semin Hematol 2013;50:4860.Google Scholar
Barreyro, L, Will, B, Bartholdy, B, et al. Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS. Blood 2012;120:1290–8.Google Scholar
Askmyr, M, Ågerstam, H, Hansen, N, et al. Selective killing of candidate AML stem cells by antibody targeting of IL1RAP. Blood 2013;121:3709–13.Google Scholar
Schultz, KR, Bowman, WP, Aledo, A, et al. Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a Children’s Oncology Group study. J Clin Oncol 2009;27:5175–81.CrossRefGoogle ScholarPubMed
Manabe, A, Kawasaki, H, Shimada, H, et al. Imatinib use immediately before stem cell transplantation in children with Philadelphia chromosome-positive acute lymphoblastic leukemia: Results from Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) Study Ph+ALL04. Cancer Med 2015; Jan 31. doi: 10.1002/cam4.383. [Epub ahead of print]Google Scholar
Lee, HJ, Thompson, JE, Wang, ES, Wetzler, M. Philadelphia chromosome-positive acute lymphoblastic leukemia. Cancer 2011;117:1583–94.Google Scholar
Bachanova, V, Marks, DI, Zhang, M-J, et al. Ph+ ALL patients in first complete remission have similar survival after reduced intensity and myeloablative allogeneic transplantation: Impact of tyrosine kinase inhibitor and minimal residual disease. Leukemia 2014;28:658–65.Google Scholar
Wetzler, M, Watson, D, Stock, W, et al. Autologous transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia achieves outcomes similar to allogeneic transplantation: results of CALGB Study 10001 (Alliance). Haematologica 2014;99:111–5.Google Scholar
Schenk, TM, Keyhani, A, Böttcher, S, et al. Multilineage involvement of Philadelphia chromosome positive acute lymphoblastic leukemia. Leukemia 1998;12:666–74.Google Scholar
Rovida, E, Peppicelli, S, Bono, S, et al. The metabolically-modulated stem cell niche: a dynamic scenario regulating cancer cell phenotype and resistance to therapy. Cell Cycle 2014;13:3169–75.Google Scholar
Ribera, J-M. Optimal approach to treatment of patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: how to best use all the available tools. Leuk Lymphoma 2013;54:21–7.Google Scholar
Herrmann, H, Sadovnik, I, Cerny-Reiterer, S, et al. Dipeptidylpeptidase IV (CD26) defines leukemic stem cells in chronic myeloid leukemia. Blood 2014;123:3951–62.Google Scholar
Pfeifer, H, Wassmann, B, Bethge, W, et al. Randomized comparison of prophylactic and minimal residual disease-triggered imatinib after allogeneic stem cell transplantation for BCR-ABL1-positive acute lymphoblastic leukemia. Leukemia 2013;27:1254–62.Google Scholar
Orgel, E, Tucci, J, Alhushki, W, et al. Obesity is associated with residual leukemia following induction therapy for childhood B-precursor acute lymphoblastic leukemia. Blood 2014;124:3932–8.Google Scholar
Ehsanipour, EA, Sheng, X, Behan, JW, et al. Adipocytes cause leukemia cell resistance to L-asparaginase via release of glutamine. Cancer Res 2013;73:29983006.Google Scholar
Pramanik, R, Sheng, X, Ichihara, B, et al. Adipose tissue attracts and protects acute lymphoblastic leukemia cells from chemotherapy. Leuk Res 2013;37:503–9.Google Scholar
Borowitz, MJ, Devidas, M, Hunger, SP, et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children’s Oncology Group study. Blood 2008;111:5477–85.Google Scholar
Möricke, A, Reiter, A, Zimmermann, M, et al. Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial ALL-BFM 95. Blood 2008;111:4477–89.Google Scholar
Conter, V, Bartram, CR, Valsecchi, MG, et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010;115:3206-14.Google Scholar
Corbacioglu, A, Scholl, C, Schlenk, RF, et al. Prognostic impact of miminal residual disease in CBFB-MYH11-positive acute myeloid leukemia. J Clin Oncol 2010;28:3724–9.Google Scholar
Zhu, H-H, Zhang, X-H, Qin, Y-Z, et al. MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial. Blood 2013;121:4056–62.Google Scholar
Jourdan, E, Boissel, N, Chevret, S, et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood 2013;121:2213–23.Google Scholar
Liu Yin, JA, O’Brien, MA, Hills, RK, et al. Minimal residual disease monitoring by RT-qPCR in core-binding factor AML allows risk-stratification and predicts relapse: results of the UK MRC AML-15 trial. Blood 2012;120:2826–35.Google Scholar
Buccisano, F, Maurillo, L, Spagnoli, A, et al. Cytogenetic and molecular diagnostic characterization combined to post-consolidation minimal residual disease assessment by flow-cytometry improves risk stratification in adult acute myeloid leukemia. Blood 2010;116:2295–303.Google Scholar
Gönen, M, Sun, Z, Figueroa, ME, et al. CD25 expression status improves prognostic risk classification in AML independent of established biomarkers: ECOG phase III trial, E1900. Blood 2012;120:2297–306.Google Scholar
Waanders, E, van der Velden, VHJ, van der Schoot, CE, et al. Integrated use of minimal residual disease classification and IKZF1 alteration status accurately predicts 79% of relapses in pediatric acute lymphoblastic leukemia. Leukemia 2011;25:254–8.Google Scholar
Geng, H, Brennan, S, Milne, TA, et al. Integrative epigenomic analysis of adult B-acute lymphoblastic leukemia identifies biomarkers and therapeutic targets. Cancer Discovery 2012;2:1004–23.Google Scholar
Kobayashi, CI, Takubo, K, Kobayashi, H, et al. The IL-2/CD25 axis maintains distinct subsets of chronic myeloid leukemia-initiating cells. Blood 2014;123:2540–9.Google Scholar
Hagedorn, N, Acquaviva, C, Fronkova, E, et al. Submicroscopic bone marrow involvement in isolated extramedullary relapses in childhood acute lymphoblastic leukemia: a more precise definition of “isolated” and its possible clinical implications, a collaborative study of the Resistant Disease Committee of the International BFM study group. Blood 2007;110:4022–9.Google Scholar
Patel, B, Rai, L, Buck, G, et al. Minimal residual disease is a significant predictor of treatment failure in non T-lineage adult acute lymphoblastic leukaemia: final results of the international trial UKALL XII/ECOG2993. Br J Haematol 2009;148:80–9.Google Scholar
Grimwade, D, Jovanovic, JV, Hills, RK, et al. Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy. J Clin Oncol 2009;27:3650–8.Google Scholar
Sockel, K, Wermke, M, Radke, J, et al. Minimal residual disease-directed preemptive treatment with azacitidine in patients with NPM1-mutant acute myeloid leukemia and molecular relapse. Haematologica 2011;96:1568–70.Google Scholar
Platzbecker, U, Wermke, M, Radke, J, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia 2012;26:381–9.Google Scholar
Rubnitz, J, Inaba, H, Dahl, G, et al. Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial. Lancet Oncol 2010;11:543–52.Google Scholar
O’Hear, C, Inaba, H, Punds, S, et al. Gemtuzumab ozogamicin can reduce minimal residual disease in patients with childhood acute myeloid leukemia. Cancer 2013;119:4036–43.Google Scholar
Annesley, CE, Brown, P. Novel agents for the treatment of childhood acute leukemia. Ther Adv Hematol 2015;6:6179.Google Scholar
Topp, M, Gökbuget, N, Zugmaier, G, et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 2012;120:5185–7.Google Scholar

References

Goldman, JM, Gale, RP, Horowitz, MM, Biggs, JC, Champlin, RE, Gluckman, E, et al. Bone marrow transplantation for chronic myelogenous leukemia in chronic phase. Increased risk for relapse associated with T-cell depletion. Ann Intern Med. 1988;108(6):806–14. PubMed PMID: 3285744.Google Scholar
Kolb, HJ, Mittermuller, J, Clemm, C, Holler, E, Ledderose, G, Brehm, G, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76(12):2462–5. PubMed PMID: 2265242.Google Scholar
Dazzi, F, Szydlo, RM, Cross, NC, Craddock, C, Kaeda, J, Kanfer, E, et al. Durability of responses following donor lymphocyte infusions for patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Blood. 2000;96(8):2712–6. PubMed PMID: 11023502.Google Scholar
Craddock, C, Szydlo, RM, Klein, JP, Dazzi, F, Olavarria, E, van Rhee, F, et al. Estimating leukemia-free survival after allografting for chronic myeloid leukemia: a new method that takes into account patients who relapse and are restored to complete remission. Blood. 2000;96(1):8690. PubMed PMID: 10891435.Google Scholar
Mackinnon, S, Papadopoulos, EB, Carabasi, MH, Reich, L, Collins, NH, Boulad, F, et al. Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease. Blood. 1995;86(4):1261–8. PubMed PMID: 7632930.Google Scholar
van Rhee, F, Savage, D, Blackwell, J, Orchard, K, Dazzi, F, Lin, F, et al. Adoptive immunotherapy for relapse of chronic myeloid leukemia after allogeneic bone marrow transplant: equal efficacy of lymphocytes from sibling and matched unrelated donors. Bone Marrow Transplant. 1998;21(10):1055–61. PubMed PMID: 9632281.Google Scholar
Simula, MP, Marktel, S, Fozza, C, Kaeda, J, Szydlo, RM, Nadal, E, et al. Response to donor lymphocyte infusions for chronic myeloid leukemia is dose-dependent: the importance of escalating the cell dose to maximize therapeutic efficacy. Leukemia. 2007;21(5):943–8. PubMed PMID: 17361226. Epub 2007/03/16. eng.Google Scholar
Dazzi, F, Szydlo, RM, Craddock, C, Cross, NC, Kaeda, J, Chase, A, et al. Comparison of single-dose and escalating-dose regimens of donor lymphocyte infusion for relapse after allografting for chronic myeloid leukemia. Blood. 2000;95(1):6771. PubMed PMID: 10607686.Google Scholar
Fozza, C, Szydlo, RM, Abdel-Rehim, MM, Nadal, E, Goldman, JM, Apperley, JF, et al. Factors for graft-versus-host disease after donor lymphocyte infusions with an escalating dose regimen: lack of association with cell dose. Bri J Haematol. 2007;136(6):833–6. PubMed PMID: 17341269. Epub 2007/03/08. eng.Google Scholar
Ferrara, JL, Levine, JE, Reddy, P, Holler, E. Graft-versus-host disease. Lancet. 2009;373(9674):1550–61. PubMed PMID: 19282026. Pubmed Central PMCID: 2735047. Epub 2009/03/14. eng.Google Scholar
Vianello, F, Cannella, L, Coe, D, Chai, JG, Golshayan, D, Marelli-Berg, FM, et al. Enhanced and aberrant T cell trafficking following total body irradiation: a gateway to graft-versus-host disease? Br J Haematol. 2013;162(6):808–18. PubMed PMID: 23855835.Google Scholar
Bar, M, Sandmaier, BM, Inamoto, Y, Bruno, B, Hari, P, Chauncey, T, et al. Donor lymphocyte infusion for relapsed hematological malignancies after allogeneic hematopoietic cell transplantation prognostic relevance of the initial CD3+ T cell dose. Biol Blood Marrow Transplant. 2013 19(6):949–57.Google Scholar
Schmid, C, Schleuning, M, Hentrich, M, Markl, GE, Gerbitz, A, Tischer, J, et al. High antileukemic efficacy of an intermediate intensity conditioning regimen for allogeneic stem cell transplantation in patients with high-risk acute myeloid leukemia in first complete remission. Bone Marrow Transplant. 2008;41(8):721–7.Google Scholar
Schmid, C, Labopin, M, Nagler, A, Bornhauser, M, Finke, J, Fassas, A et al. Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. J Clin Oncol. 2007;25:4938–45.CrossRefGoogle ScholarPubMed
Campregher, PV, Gooley, T, Scott, BL, Moravec, C, Sandmaier, B, Martin, PJ, et al. Results of donor lymphocyte infusions for relapsed myelodysplastic syndrome after hematopoietic cell transplantation. Bone Marrow Transplant. 2007;40(10):965–71.Google Scholar
Elliott, MA, Tefferi, A, Hogan, WJ, Letendre, L, Gastineau, DA, Ansell, SM, et al. Allogeneic stem cell transplantation and donor lymphocyte infusions for chronic myelomonocytic leukemia. Bone Marrow Transplant. 2006; 37(11):1003–8.Google Scholar
Krishnamurthy, P, Potter, VT, Barber, LD, Kulasekararaj, AG, Lim, ZY, Pearce, RM, de Lavallade, H, et al. Outcome of donor lymphocyte infusion after T cell-depleted allogeneic hematopoietic stem cell transplantation for acute myelogenous leukemia and myelodysplastic syndromes. Biol Blood Marrow Transplant. 2013;19(4):562–8.Google Scholar
Liga, M, Triantafyllou, E, Tiniakou, M, Lambropoulou, P, Karakantza, M, Zoumbos, NC, et al. High alloreactivity of low-dose prophylactic donor lymphocyte infusion in patients with acute leukemia undergoing allogeneic hematopoietic cell transplantation with an alemtuzumab-containing conditioning regimen. Biol Blood Marrow Transplant. 2013;19(1):7581.Google Scholar
Schroeder, T, Fröbel, J, Cadeddu, RP, Czibere, A, Dienst, A, Platzbecker, U, et al. Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation. Leukemia. 2013;27(9):1910–3.Google Scholar
McLornan, DP, Mead, AJ, Jackson, G, Harrison, CN. Allogeneic stem cell transplantation for myelofibrosis in 2012. Br J Haematol. 2012;157(4):413–25.Google Scholar
Kröger, N, Alchalby, H, Klyuchnikov, E, Badbaran, A, Hildebrandt, Y, Ayuk, F, et al. JAK2-V617F-triggered preemptive and salvage adoptive immunotherapy with donor-lymphocyte infusion in patients with myelofibrosis after allogeneic stem cell transplantation. Blood. 2009;113(8):1866–8.Google Scholar
Yoshimi, A, Bader, P, Matthes-Martin, S, Stary, J, Sedlacek, P, Duffner, U et al. Donor leukocyte infusion after hematopoietic stem cell transplantation in patients with juvenile myelomonocytic leukemia. Leukemia 2005;19:971–7.Google Scholar
El-Jurdi, N, Reljic, T, Kumar, A, Pidala, J, Bazarbachi, A, Djulbegovic, B, et al. Efficacy of adoptive immunotherapy with donor lymphocyte infusion in relapsed lymphoid malignancies. Immunotherapy. 2013;5(5):457–66.Google Scholar
Bloor, AJ, Thomson, K, Chowdhry, N, Verfuerth, S, Ings, SJ, Chakraverty, R, et al. High response rate to donor lymphocyte infusion after allogeneic stem cell transplantation for indolent non-Hodgkin lymphoma. Biol Blood Marrow Transplant. 2008;14(1):50–8.Google Scholar
Thomson, KJ, Morris, EC, Milligan, D, Parker, AN, Hunter, AE, Cook, G, 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.Google Scholar
Peggs, KS, Kayani, I, Edwards, N, Kottaridis, P, Goldstone, AH, Linch, DC et al. Donor lymphocyte infusions modulate relapse risk in mixed chimeras and induce durable salvage in relapsed patients after T-cell-depleted allogeneic transplantation for Hodgkin’s lymphoma. J Clin Oncol. 2011;29(8):971–8.Google Scholar
Theurich, S, Malcher, J, Wennhold, K, Shimabukuro-Vornhagen, A, Chemnitz, J, Holtick, U, et al. Brentuximab vedotin combined with donor lymphocyte infusions for early relapse of Hodgkin lymphoma after allogeneic stem-cell transplantation induces tumor-specific immunity and sustained clinical remission. J Clin Oncol. 2013;31(5):e59-63.Google Scholar
Sala, E, Crocchiolo, R, Gandolfi, S, Bruno-Ventre, M, Bramanti, S, Peccatori, J, et al. Bendamustine combined with donor lymphocytes infusion in Hodgkin’s lymphoma relapsing after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2014;20(9):1444–7.Google Scholar
Richardson, SE, Khan, I, Rawstron, A, Sudak, J, Edwards, N, Verfuerth, S, et al. Risk-stratified adoptive cellular therapy following allogeneic hematopoietic stem cell transplantation for advanced chronic lymphocytic leukaemia. Br J Haematol. 2013;160(5):640–8.Google Scholar
Slavin, S, Naparstek, E, Nagler, A, Ackerstein, A, Samuel, S, Kapelushnik, J et al. Allogeneic cell therapy with donor peripheral blood cells and recombinant human interleukin-2 to treat leukemia relapse after allogeneic bone marrow transplantation. Blood 1996; 87:2195–204.Google Scholar
Collins, RH Jr., Goldstein, S, Giralt, S, Levine, J, Porter, D, Drobyski, W et al. Donor leukocyte infusions in acute lymphocytic leukemia. Bone Marrow Transplant. 2000;26:511–16.Google Scholar
Medd, PG, Peniket, AJ, Littlewood, TJ, Pearce, R, Perry, J, Kirkland, KE et al; British Society of Blood and Marrow Transplantation. Evidence for a GVL effect following reduced-intensity allo-SCT in ALL: a British Society of Blood and Marrow Transplantation study. Bone Marrow Transplant. 2013;48(7):982–7.Google Scholar
van de Donk, NW, Kröger, N, Hegenbart, U, Corradini, P, San Miguel, JF, Goldschmidt, H, et al. Prognostic factors for donor lymphocyte infusions following non-myeloablative allogeneic stem cell transplantation in multiple myeloma. Bone Marrow Transplant. 2006;37(12):1135–41.Google Scholar
Stern, M, de Wreede, LC, Brand, R, van Biezen, A, Dreger, P, Mohty, M, et al. Sensitivity of hematological malignancies to graft-versus-host effects: an EBMT megafile analysis. Leukemia. 2014; Apr 30. [Epub ahead of print]Google Scholar
Tyler, EM, Jungbluth, AA, O’Reilly, RJ, Koehne, G. WT1-specific T-cell responses in high-risk multiple myeloma patients undergoing allogeneic T cell-depleted hematopoietic stem cell transplantation and donor lymphocyte infusions. Blood. 2013;121(2):308–17.Google Scholar
Cooper, N., Rao, K., Goulden, N., Amrolia, P. and Veys, P. Alpha interferon augments the graft-versus-leukaemia effect of second stem cell transplants and donor lymphocyte infusions in high-risk paediatric leukaemias. Br J Haematol. 2012;156:550–2.Google Scholar
El-Cheikh, J, Crocchiolo, R, Furst, S, Ladaique, P, Castagna, L, Faucher, C, et al. Lenalidomide plus donor-lymphocytes infusion after allogeneic stem-cell transplantation with reduced-intensity conditioning in patients with high-risk multiple myeloma. Exp Hematol. 2012;40(7):521–7.Google Scholar
Rizzieri, DA, Storms, R, Chen, DF, Long, G, Yang, Y, Nikcevich, DA et al. Natural killer cell-enriched donor lymphocyte infusions from A 3–6/6 HLA-matched family member following nonmyeloablative allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2010;16(8):1107–14.Google Scholar
Ho, VT, Kim, HT, Kao, G, Cutler, C, Levine, J, Rosenblatt, J, et al. Sequential infusion of donor-derived dendritic cells with donor lymphocyte infusion for relapsed hematologic cancers after allogeneic hematopoietic stem cell transplantation. Am J Hematol. 2014; Aug 13. [Epub ahead of print]Google Scholar
Wagner, E, Wehler, D, Kolbe, K, Theobald, M, Herr, W and Meyer, R. Impact of Prophylactic CD8-Depleted Donor-Lymphocyte Infusions After Allogeneic Hematopoietic Stem Cell Transplantation and Alemtuzumab Mediated T-Cell Depletion. Abstract 4109. American Society of Hematology Annual Meeting 2013.Google Scholar
Samarasinghe, S, Mancao, C, Pule, M, Nawroly, N, Karlsson, H, Brewin, J, et al. Functional characterization of alloreactive T cells identifies CD25 and CD71 as optimal targets for a clinically applicable allodepletion strategy. Blood. 2010;115(2):396407.Google Scholar
Solomon, SR, Mielke, S, Savani, BN, Montero, A, Wisch, L, Childs, R, et al. Selective depletion of alloreactive donor lymphocytes: a novel method to reduce the severity of graft-versus-host disease in older patients undergoing matched sibling donor stem cell transplantation. Blood. 2005;106(3):1123–9. Epub 2005 Apr 7.Google Scholar
Ciceri, F, Bonini, C, Marktel, S, Zappone, E, Servida, P, Bernadi, M et al. Anti-tumor effect of HSV-TK engineered donor lymphocytes after allogeneic stem cell transplantation. Blood. 2007;109:4698–707.Google Scholar
Ciceri, F, Lupo-Stanghellini, M, Oliveira, G, Greco, R et al. Long-term safety and survival outcomes after TK-expressing donor lymphocyte infusion (TK-DLI) in allogeneic hematopoietic stem cell transplantation (HSCT).J Clin Oncol. 2013;31(Suppl): abstr 7007.Google Scholar
Di Stasi, A, Tey, SK, Dotti, G, Fujita, Y, Kennedy-Nasser, A, Martinez, C, Vago, L, Bondanzo, A et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365(18):1673–83.Google Scholar

References

Gooley, TA, Chien, JW, Pergam, SA, et al. Reduced mortality after allogeneic hematopoietic-cell transplantation. N Engl J Med. 2010;363(22):2091–101.Google Scholar
Horan, JT, Logan, BR, Agovi-Johnson, MA, et al. Reducing the risk for transplantation-related mortality after allogeneic hematopoietic cell transplantation: how much progress has been made? J Clin Oncol. 2011;29(7):805–13.Google Scholar
Anasetti, C, Logan, BR, Lee, SJ, et al. Blood and Marrow Transplant Clinical Trials Network. Peripheral-blood stem cells versus bone marrow from unrelated donors. N Engl J Med. 2012;367(16):1487–96.Google Scholar
Rocha, V, Labopin, M, Sanz, G, et al. Acute Leukemia Working Party of European Blood and Marrow Transplant Group; Eurocord-Netcord Registry. Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia. N Engl J Med. 2004;351(22):2276–85.Google Scholar
Locatelli, F, Kabbara, N, Ruggeri, A, et al. Outcome of patients with hemoglobinopathies given either cord blood or bone marrow transplantation from an HLA-identical sibling. Blood. 2013;122(6):1072–8.Google Scholar
Bernaudin, F, Socie, G, Kuentz, M, et al. SFGM-T: .Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease. Blood. 2007;110(7):2749–56.Google Scholar
Porter, DL, Alyea, EP, Antin, JH, et al. NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: Report from the Committee on Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2010;16(11):1467–503.Google Scholar
Schmid, C, Labopin, M, Nagler, A, et al. Acute Leukaemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT). Treatment, risk factors, and outcome of adults with relapsed AML after reduced intensity conditioning for allogeneic stem cell transplantation. Blood. 2012;119(6):1599–606.Google Scholar
Pollyea, DA, Artz, AS, Stock, W, et al. Outcomes of patients with AML and MDS who relapse or progress after reduced intensity allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2007;40(11):1027–32.Google Scholar
Kenkre, VP, Horowitz, S, Artz, AS, et al. T-cell-depleted allogeneic transplant without donor leukocyte infusions results in excellent long-term survival in patients with multiply relapsed Lymphoma. Predictors for survival after transplant relapse. Leuk Lymphoma. 2011;52(2):214–22.Google Scholar
Elmaagacli, AH, Beelen, DW, Schaefer, UW. A retrospective single centre study of the outcome of five different therapy approaches in 48 patients with relapse of chronic myelogenous leukemia after allogeneic bone marrow transplantation. Bone Marrow Transplant. 1997;20(12):1045–55.Google Scholar
Mehta, J, Powles, R, Kulkarni, S, et al. Induction of graft-versus-host disease as immunotherapy of leukemia relapsing after allogeneic transplantation: single-center experience of 32 adult patients. Bone Marrow Transplant. 1997;20(2):129–35.Google Scholar
Higano, CS, Brixey, M, Bryant, EM, et al. Durable complete remission of acute nonlymphocytic leukemia associated with discontinuation of immunosuppression following relapse after allogeneic bone marrow transplantation. A case report of a probable graft-versus-leukemia effect. Transplantation. 1990;50(1):175–7.Google Scholar
Giralt, S, Escudier, S, Kantarjian, H, et al. Preliminary results of treatment with filgrastim for relapse of leukemia and myelodysplasia after allogeneic bone marrow transplantation. N Engl J Med. 1993;329(11):757–61.Google Scholar
Worth, LL, Mullen, CA, Choroszy, M, et al. Treatment of leukemia relapse with recombinant granulocyte-macrophage colony stimulating factor (rhGM-CSF) following unrelated umbilical cord blood transplant: Induction of graft-vs.-leukemia. Pediatr Transplant. 2002;6(5):439–42.Google Scholar
Jabbour, E, Giralt, S, Kantarjian, H, et al. Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia. Cancer. 2009;115(9):1899–905.Google Scholar
Bolaños-Meade, J, Smith, BD, Gore, SD, et al. 5-azacytidine as salvage treatment in relapsed myeloid tumors after allogeneic bone marrow transplantation. Biol Blood Marrow Transplant. 2011;17(5):754–8.Google Scholar
Czibere, A, Bruns, I, Kröger, N, et al. 5-Azacytidine for the treatment of patients with acute myeloid leukemia or myelodysplastic syndrome who relapse after allo-SCT: a retrospective analysis. Bone Marrow Transplant. 2010;45(5):872–6.Google Scholar
Fathi, AT, Chen, YB. Treatment of relapse of acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Curr Hematol Malig Rep. 2014;9(2):186–92.Google Scholar
Giralt, SA, Champlin, RE. Leukemia relapse after allogeneic bone marrow transplantation: a review. Blood. 1994;84(11):3603–12.Google Scholar
Sharma, M, Ravandi, F, Bayraktar, UD, et al. Treatment of FLT3-ITD-positive acute myeloid leukemia relapsing after allogeneic stem cell transplantation with sorafenib. Biol Blood Marrow Transplant. 2011;17(12):1874–7.Google Scholar
Metzelder, SK, Schroeder, T, Finck, A, et al. High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses. Leukemia. 2012;26(11):2353–9.Google Scholar
Kolb, HJ, Mittermüller, 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.Google Scholar
Kolb, HJ, Schattenberg, A, Goldman, JM, et al. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia: Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood. 1995;86(5):2041–50.Google Scholar
Porter, DL, Collins, RH Jr, Hardy, C, et al. Treatment of relapsed leukemia after unrelated donor marrow transplantation with unrelated donor leukocyte infusions. Blood. 2000;95(4):1214–21.Google Scholar
Schmid, C, Labopin, M, Nagler, A, et al. EBMT Acute Leukemia Working Party. Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. J Clin Oncol. 2007;25(31):4938–45.Google Scholar
Schroeder, T, Czibere, A, Platzbecker, U, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013;27(6):1229–35.Google Scholar
Hasskarl, J, Zerweck, A, Wäsch, R, et al. Induction of graft versus malignancy effect after unrelated allogeneic PBSCT using donor lymphocyte infusions derived from frozen aliquots of the original graft. Bone Marrow Transplant. 2012;47(2):277–82.Google Scholar
Warlick, ED, DeFor, T, Blazar, BR, et al. Successful remission rates and survival after lymphodepleting chemotherapy and donor lymphocyte infusion for relapsed hematologic malignancies postallogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2012;18(3):480–6.Google Scholar
Porter, DL, Collins, RH Jr, Shpilberg, O, et al. Long-term follow-up of patients who achieved complete remission after donor leukocyte infusions. Biol Blood Marrow Transplant. 1999;5(4):253–61.Google Scholar
Fan, Y, Liu, H, Artz, A et al. The outcomes of second allogeneic stem cell transplantation for disease relapse after T cell depleted allogeneic stem cell transplantation: A single center experience, University of Chicago. Blood. 2014;124:2509.Google Scholar
Christopeit, M, Kuss, O, Finke, J, et al. Second allograft for hematologic relapse of acute leukemia after first allogeneic stem-cell transplantation from related and unrelated donors: the role of donor change. J Clin Oncol. 2013;31(26):3259–71.Google Scholar
Leung, AY, Tse, E, Hwang, YY, et al. Primary treatment of leukemia relapses after allogeneic hematopoietic stem cell transplantation with reduced-intensity conditioning second transplantation from the original donor. Am J Hematol. 2013;88(6):485–91.Google Scholar
Christopoulos, P, Schmoor, C, Waterhouse, M, et al. Reduced-intensity conditioning with fludarabine and thiotepa for second allogeneic transplantation of relapsed patients with AML. Bone Marrow Transplant. 2013;48(7):901–7.Google Scholar
Poon, LM, Bassett, R Jr, Rondon, G, et al. Outcomes of second allogeneic hematopoietic stem cell transplantation for patients with acute lymphoblastic leukemia. Bone Marrow Transplant. 2013;48(5):666–70.Google Scholar
Spyridonidis, A, Labopin, M, Schmid, C, et al. Immunotherapy Subcommittee of Acute Leukemia Working Party. Outcomes and prognostic factors of adults with acute lymphoblastic leukemia who relapse after allogeneic hematopoietic cell transplantation. An analysis on behalf of the Acute Leukemia Working Party of EBMT. Leukemia. 2012;26(6):1211–7.Google Scholar
Hartwig, M, Ocheni, S, Asenova, S, et al. Second allogeneic stem cell transplantation in myeloid malignancies. Acta Haematol. 2009;122(4):185–92.Google Scholar
Shaw, BE, Mufti, GJ, Mackinnon, S, et al. Outcome of second allogeneic transplants using reduced-intensity conditioning following relapse of haematological malignancy after an initial allogeneic transplant. Bone Marrow Transplant. 2008;42(12):783–9.Google Scholar
Eapen, M, Giralt, SA, Horowitz, MM, et al. Second transplant for acute and chronic leukemia relapsing after first HLA-identical sibling transplant. Bone Marrow Transplant. 2004;34(8):721–7.Google Scholar
Chueh, HW, Lee, SH, Sung, KW, et al. Second allogeneic stem cell transplantation in hematologic malignancies: a single-center experience. J Pediatr Hematol Oncol. 2013;35(6):424–9.CrossRefGoogle ScholarPubMed
Sorror, ML. Comorbidities and hematopoietic cell transplantation outcomes. Hematology Am Soc Hematol Educ Program. 2010;2010:237–47.Google Scholar
Artz, AS, Pollyea, DA, Kocherginsky, M, et al. Performance status and comorbidity predict transplant-related mortality after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2006;12(9):954–64.Google Scholar
Kishi, K, Takahashi, S, Gondo, H, et al. Second allogeneic bone marrow transplantation for post-transplant leukemia relapse: results of a survey of 66 cases in 24 Japanese institutes. Bone Marrow Transplant. 1997;19(5):461–6.Google Scholar
Hill, BT, Bolwell, BJ, Rybicki, L, et al. Nonmyeloablative second transplants are associated with lower nonrelapse mortality and superior survival than myeloablative second transplants. Biol Blood Marrow Transplant. 2010;16(12):1738–46.Google Scholar
Kedmi, M, Resnick, IB, Dray, L, et al. A retrospective review of the outcome after second or subsequent allogeneic transplantation. Biol Blood Marrow Transplant. 2009;15(4):483–9.Google Scholar
Wagner, JE, Vogelsang, GB, Zehnbauer, BA, et al. Relapse of leukemia after bone marrow transplantation: effect of second myeloablative therapy. Bone Marrow Transplant. 1992;9(3):205–9.Google Scholar
Hurley, CK, Raffoux, C; World Marrow Donor Association. World Marrow Donor Association: international standards for unrelated hematopoietic stem cell donor registries. Bone Marrow Transplant. 2004;34(2):103–10.Google Scholar
Barrett, AJ, Locatelli, F, Treleaven, JG, et al. Second transplants for leukaemic relapse after bone marrow transplantation: high early mortality but favourable effect of chronic GVHD on continued remission. A report by the EBMT Leukaemia Working Party. Br J Haematol. 1991;79(4):567–74.Google Scholar
Al-Qurashi, F, Ayas, M, Al Sharif, F, et al. Second allogeneic bone marrow transplantation after myeloablative conditioning analysis of 43 cases from single institution. Hematology. 2004;9(2):123–9.Google Scholar
Storb, R, Gyurkocza, B, Storer, BE, et al. Graft-versus-host disease and graft-versus-tumor effects after allogeneic hematopoietic cell transplantation. J Clin Oncol. 2013;31(12):1530–8Google Scholar
Bhatia, S, Francisco, L, Carter, A, et al. Late mortality after allogeneic hematopoietic cell transplantation and functional status of long-term survivors: report from the Bone Marrow Transplant Survivor Study. Blood. 2007;110(10):3784–92.Google Scholar
Bosi, A, Laszlo, D, Labopin, M, et al. Acute Leukemia Working Party of the European Blood and Marrow Transplant Group. Second allogeneic bone marrow transplantation in acute leukemia: results of a survey by the European Cooperative Group for Blood and Marrow Transplantation. J Clin Oncol. 2001;19(16):3675–84.Google Scholar
Russell, JA, Bowen, T, Brown, C, et al. Second allogeneic transplants for leukemia using blood instead of bone marrow as a source of hemopoietic cells. Bone Marrow Transplant. 1996;18(3):501–5.Google Scholar
Körbling, M, Huh, YO, Durett, A, et al. Allogeneic blood stem cell transplantation: peripheralization and yield of donor-derived primitive hematopoietic progenitor cells (CD34+ Thy-1dim) and lymphoid subsets, and possible predictors of engraftment and graft-versus-host disease. Blood. 1995;86(7):2842–8.Google Scholar
Stroncek, DF, Lopez, AM, Maharaj, D, et al. Acute toxicities of unrelated bone marrow versus peripheral blood stem cell donation: results of a prospective trial from the National Marrow Donor Program. Blood. 2013;121(1):197206.Google Scholar
Churpek, JE, Nickels, E, Marquez, R, et al. Identifying familial myelodysplastic/acute leukemia predisposition syndromes through hematopoietic stem cell transplantation donors with thrombocytopenia. Blood. 2012;120(26):5247–9.Google Scholar
Srinivasan, R, Takahashi, Y, McCoy, JP, et al. Overcoming graft rejection in heavily transfused and allo-immunised patients with bone marrow failure syndromes using fludarabine-based haematopoietic cell transplantation. Br J Haematol. 2006;133(3):305–14.Google Scholar
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.Google Scholar
Rondón, G, Saliba, RM, Khouri, I, et al. Long-term follow-up of patients who experienced graft failure post allogeneic progenitor cell transplantation. Results of a single institution analysis. Biol Blood Marrow Transplant. 2008;14(8):859–66.Google Scholar
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.Google Scholar
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.Google Scholar
Young, JW, Papadopoulos, EB, Cunningham, I, et al. T-cell-depleted allogeneic bone marrow transplantation in adults with acute nonlymphocytic leukemia in first remission. Blood. 1992;79(12):3380–7.Google Scholar
Anasetti, C, Amos, D, Beatty, PG, et al. Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma. N Engl J Med. 1989;320(4):197204.Google Scholar
Ruggeri, A, Labopin, M, Sormani, MP, et al. Engraftment kinetics and graft failure after single umbilical cord blood transplantation using myeloablative conditioning regimen. Haematologica. 2014;99:1509–15.Google Scholar
Ciurea, SO, de Lima, M, Cano, P, et al. High risk of graft failure in patients with anti-HLA antibodies undergoing haploidentical stem-cell transplantation. Transplantation. 2009;88(8):1019–24.Google Scholar
Cutler, C, Kim, HT, Sun, L, et al. Donor-specific anti-HLA antibodies predict outcome in double umbilical cord blood transplantation. Blood. 2011;118(25):6691–7.Google Scholar
Laughlin, MJ, Eapen, M, Rubinstein, P, et al. Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia. N Engl J Med. 2004;351(22):2265–75.Google Scholar
Lazarus, HM, Kan, F, Tarima, S, et al. Rapid transport and infusion of hematopoietic cells is associated with improved outcome after myeloablative therapy and unrelated donor transplant. Biol Blood Marrow Transplant. 2009;15(5):589–96.Google Scholar
Lioznov, M, Dellbrügger, C, Sputtek, A, et al. Transportation and cryopreservation may impair haematopoietic stem cell function and engraftment of allogeneic PBSCs, but not BM. Bone Marrow Transplant. 2008;42(2):121–8.Google Scholar
Ramirez, P, Wagner, JE, DeFor, TE, et al. Factors predicting single-unit predominance after double umbilical cord blood transplantation. Bone Marrow Transplant. 2012;47(6):799803.Google Scholar
Storb, R, Epstein, RB, Rudolph, RH, Thomas, ED. The effect of prior transfusion on marrow grafts between histocompatible canine siblings. J Immunol. 1970;105(3):627–33.Google Scholar
Storb, R, Rudolph, RH, Graham, TC, Thomas, ED. The influence of transfusions from unrelated donors upon marrow grafts between histocompatible canine siblings. J Immunol. 1971;107(2):409–13.Google Scholar
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):3611–9.Google Scholar
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.Google Scholar
Takanashi, M, Atsuta, Y, Fujiwara, K, et al. The impact of anti-HLA antibodies on unrelated cord blood transplantations. Blood. 2010;116(15):2839–46.Google Scholar
Spellman, S, Bray, R, Rosen-Bronson, 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(13):2704–8.Google Scholar
Ruggeri, A, Rocha, V, Masson, E, et al. Impact of donor-specific anti-HLA antibodies on graft failure and survival after reduced intensity conditioning-unrelated cord blood transplantation: a Eurocord, Société Francophone d’Histocompatibilité et d’Immunogénétique (SFHI) and Société Française de Greffe de Moelle et de Thérapie Cellulaire (SFGM-TC) analysis. Haematologica. 2013;98(7):1154–60.Google Scholar
Storb, R. B-cells versus T cells as primary barrier to hematopoietic engraftment in allosensitized recipients. Blood. 2009;113(5):1205.Google Scholar
Brunstein, CG, Noreen, H, DeFor, TE, et al. Anti-HLA antibodies in double umbilical cord blood transplantation. Biol Blood Marrow Transplant. 2011;17(11):1704–8.Google Scholar
Ferrà, C, Sanz, J, Díaz-Pérez, MA, et al. Outcome of graft failure after allogeneic stem cell transplantation: Study of 89 patients. Leuk Lymphoma. 2014;10:124.Google Scholar
Fuji, S, Nakamura, F, Hatanaka, K, et al. Peripheral blood as a preferable source of stem cells for salvage transplantation in patients with graft failure after cord blood transplantation: a retrospective analysis of the registry data of the Japanese Society for Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant. 2012;18(9):1407–14.Google Scholar
Waki, F, Masuoka, K, Fukuda, T, et al. Feasibility of reduced-intensity cord blood transplantation as salvage therapy for graft failure: results of a nationwide survey of adult patients. Biol Blood Marrow Transplant. 2011;17(6):841–51.Google Scholar
Remberger, M, Mattsson, J, Olsson, R, Ringdén, O. Second allogeneic hematopoietic stem cell transplantation: a treatment for graft failure. Clin Transplant. 2011;25(1):E68–76.Google Scholar
Byrne, BJ, Horwitz, M, Long, GD, et al. Outcomes of a second non-myeloablative allogeneic stem cell transplantation following graft rejection. Bone Marrow Transplant. 2008;41(1):3943.Google Scholar
Jabbour, E, Rondon, G, Anderlini, P, et al. Treatment of donor graft failure with nonmyeloablative conditioning of fludarabine, antithymocyte globulin and a second allogeneic hematopoietic transplantation. Bone Marrow Transplant. 2007;40(5):431–5.Google Scholar
Chewning, JH, Castro-Malaspina, H, Jakubowski, A, et al. Fludarabine-based conditioning secures engraftment of second hematopoietic stem cell allografts (HSCT) in the treatment of initial graft failure. Biol Blood Marrow Transplant. 2007;13(11):1313–23.Google Scholar
Remberger, M, Ringdén, O, Ljungman, P, Hägglund, H, et al. Booster marrow or blood cells for graft failure after allogeneic bone marrow transplantation. Bone Marrow Transplant. 1998;22(1):73–8.Google Scholar
Appelbaum, FR, Nemunaitis, J, Singer, JW, et al. Use of recombinant human granulocyte macrophage colony-stimulating factor to speed engraftment and treat graft failure following marrow transplantation in man. Haematol Blood Transfus. 1990;33:736–40.Google Scholar
Nemunaitis, J, Singer, JW, Buckner, CD, et al. Use of recombinant human granulocyte-macrophage colony-stimulating factor in graft failure after bone marrow transplantation. Blood. 1990;76(1):245–53.Google Scholar
Poon, LM, Di Stasi, A, Popat, U, et al. Romiplostim for delayed platelet recovery and secondary thrombocytopenia following allogeneic stem cell transplantation.Am J Blood Res. 2013;3(3):260–4.Google Scholar
Calmettes, C, Vigouroux, S, Tabrizi, R, Milpied, N. Romiplostim (AMG531, Nplate) for secondary failure of platelet recovery after allo-SCT. Bone Marrow Transplant. 2011;46(12):1587–9.Google Scholar
Díez-Martín, JL, Gómez-Pineda, A, Serrano, D, et al. Successful treatment of incipient graft rejection with donor leukocyte infusions, further proof of a graft versus host lymphohaemopoietic effect. Bone Marrow Transplant. 2004;33(10):1037–41.Google Scholar
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.Google Scholar
Mehta, J, Powles, R, Singhal, S, et al. Outcome of autologous rescue after failed engraftment of allogeneic marrow. Bone Marrow Transplant. 1996;17(2):213–7.Google Scholar
Pottinger, B, Walker, M, Campbell, M, et al. The storage and re-infusion of autologous blood and BM as back-up following failed primary hematopoietic stem-cell transplantation: a survey of European practice. Cytotherapy. 2002;4(2):127–35.Google Scholar
Stelljes, M, van Biezen, A, Slavin, S, et al. The harvest and use of autologous back-up grafts for graft failure or severe GvHD after allogeneic hematopoietic stem cell transplantation: a survey of the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2008;42(11):739–42.Google Scholar
Min, CK, Kim, DW, Lee, JW, et al. Additional stem cell therapy for graft failure after allogeneic bone marrow transplantation. Acta Haematol. 2000;104(4):185–92.Google Scholar
Larocca, A, Piaggio, G, Podestà, 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.Google Scholar
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. French Society of Bone Marrow Transplantation. Br J Haematol. 2000;111(1):292302.Google Scholar
Mehta, J, Powles, R, Singhal, S, et al. Early identification of patients at risk of death due to infections, hemorrhage, or graft failure after allogeneic bone marrow transplantation on the basis of the leukocyte counts. Bone Marrow Transplant. 1997;19(4):349–55.Google Scholar
van Besien, K, Shore, T, Cushing, M. Peripheral-blood versus bone marrow stem cells. N Engl J Med. 2013;368(3):287–8.Google Scholar
Gergis, U, Mayer, S, Gordon, B, et al. A strategy to reduce donor-specific HLA Abs before allogeneic transplantation. Bone Marrow Transplant. 2014;49(5):722–4.Google Scholar
van Besien, K. Advances in umbilical cord blood transplant: an overview of the 12th International Cord Blood Symposium, San Francisco, 5–7 June 2014. Leuk Lymphoma. 2014;18:15.Google Scholar
Tachibana, T, Matsumoto, K, Tanaka, M et al. Outcome and prognostic factors among patients who underwent a second transplantation for disease relapse post the first allogeneic cell transplantation. Leuk Lymphoma.; in press.Google Scholar
Orti, G, Sanz, J, Bermudez, A, et al. Outcome of second allogeneic hematopoietic cell transplantation after relapse of myeloid malignancies following allogeneic hematopoietic cell transplantation: a retrospective cohort on behalf of the Grupo Español de Trasplante Hematopoyetico. Biol Blood Marrow Transplant. 2016;22(3):584–8.Google Scholar
Vrhovac, R, Labopin, M, Ciceri, F, et al. Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation (EBMT). Second reduced intensity conditioning allogeneic transplant as a rescue strategy for acute leukaemia patients who relapse after an initial RIC allogeneic transplantation: analysis of risk factors and treatment outcomes. Bone Marrow Transplant. 2016;51(2):186–93.Google Scholar
Lund, TC, Liegel, J, Bejanyan, N, et al. Second allogeneic hematopoietic cell transplantation for graft failure: poor outcomes for neutropenic graft failure. Am J Hematol. 2015;90(10): 892–6.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×