Reduced Intensity Bone Marrow Transplantation with Post-Transplant Cyclophosphamide for Pediatric Inherited Immune Deficiencies and Bone Marrow Failure Syndromes

Orly R Klein, Samantha Bapty, Howard M Lederman, M Elizabeth M Younger, Elias T Zambidis, Richard J Jones, Kenneth R Cooke, Heather J Symons, Orly R Klein, Samantha Bapty, Howard M Lederman, M Elizabeth M Younger, Elias T Zambidis, Richard J Jones, Kenneth R Cooke, Heather J Symons

Abstract

Purpose: Allogeneic bone marrow transplantation (alloBMT) is the only cure for many primary immune deficiency disorders (PIDD), primary immune regulatory disorders (PIRD), and inherited bone marrow failure syndromes (IBMFS).

Methods: We report the results of 25 patients who underwent alloBMT using reduced intensity conditioning (RIC), alternative donors, and post-transplantation cyclophosphamide (PTCy). In an attempt to reduce regimen-related toxicities, we removed low-dose TBI from the prep and added mycophenolate mofetil and tacrolimus for graft-versus-host disease (GVHD) prophylaxis for all donor types in the latter 14 patients. Donors were haploidentical related (n = 14), matched unrelated (n = 9), or mismatched unrelated (n = 2). The median age was 9 years (range 5 months-21 years).

Results: With a median follow-up of 26 months (range 7 months-9 years), the 2-year overall survival is 92%. There were two deaths, one from infection, and one from complications after a second myeloablative BMT. Three patients developed secondary graft failure, one at 2 years and two at >3 years, successfully treated with CD34 cell boost in one or second BMT in two. The remaining 20 patients have full or stable mixed donor chimerism and are disease-free. The incidence of mixed chimerism is increased since removing TBI from the prep. The 6-month cumulative incidence of grade II acute GVHD is 17%, with no grade III-IV. The 1-year cumulative incidence of chronic GVHD is 14%, with severe of 5%.

Conclusion: This alloBMT platform using alternative donors, RIC, and PTCy is associated with excellent rates of engraftment and low rates of GVHD and non-relapse mortality, and offers a curative option for patients with PIDD, PIRD, and IBMFS.

Trial registration: ClinicalTrials.gov Identifier: NCT04232085.

Keywords: Blood and marrow transplantation; alternative donor; inherited bone marrow failure disorders; post-transplant cyclophosphamide; primary immune deficiency disorders; primary immune regulatory disorders.

Conflict of interest statement

K.R.C. serves on the Speaker Bureau and Advisory Board for Jazz pharmaceuticals and received a research grant from Jazz pharmaceuticals; he is on an investigator-initiated trial of post-transplant therapy for solid tumors that is supported in part by Bristol Meyers Squibb. H.J.S. is on the Speaker Bureau for Jazz pharmaceuticals and is on an investigator-initiated trial of post-transplant therapy for solid tumors that is supported in part by Bristol Meyers Squibb. None of these are in direct conflict with the manuscript.

Figures

Fig. 1
Fig. 1
Preparatory regimen and GVHD prophylaxis. Prep includes alemtuzumab, fludarabine, and melphalan. GVHD prophylaxis includes cyclophosphamide, tacrolimus, and mycophenolate mofetil. Cy, cyclophosphamide; GCSF, granulocyte colony stimulating factor; MMF, mycophenolate mofetil
Fig. 2
Fig. 2
Absolute lymphocyte count (ALC) values and median. The individual values for absolute lymphocyte count (ALC) are shown for days 30, 60, 180, and 365. The median ALC at each time point were 0.38 × 109/L (range 0.02–1.14 × 109/L), 0.60 (range 0.18–2.22 × 109/L), 0.95 (range 0.14–2.10) × 109/L, and 1.76 × 109/L (range 0.37–4.35 × 109/L), respectively. The total number of patients at each time point is n = 26, n = 25, n = 24, and n = 18, respectively
Fig. 3
Fig. 3
Chimerism in whole blood and CD3+ compartment. Chimerism for all patients, n = 25 (A), chemotherapy + total body irradiation, n = 8 (B), Chemotherapy-only, n = 18 (C), IBMFS patients, n = 5 (D), PIDD/PIRD, n = 20 patients (E), and CGD patients, n = 11 (F). Graphs show percent of patients with full chimerism (defined as > 95%), high-level mixed (defined as 50–95%), low-level mixed (defined as < 50% but detectable), none detected, or not measured
Fig. 4
Fig. 4
Disease-free survival (DFS)/overall survival (OS) and event-free survival (EFS). Disease-free survival/overall survival and event-free survival for all patients. OS and DFS are the same at 2 years; three patients who developed secondary graft failure were successfully treated

References

    1. Gragert L, Eapen M, Williams E, Freeman J, Spellman S, Baitty R, Hartzman R, Rizzo JD, Horowitz M, Confer D, Maiers M. HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. Registry. N Engl J Med. 2014;371(4):339–348. doi: 10.1056/NEJMsa1311707.
    1. Filipovich AH, Stone JV, Tomany SC, Ireland M, Kollman C, Pelz CJ, Casper JT, Cowan MJ, Edwards JR, Fasth A, Gale RP, Junker A, Kamani NR, Loechelt BJ, Pietryga DW, Ringdén O, Vowels M, Hegland J, Williams AV, Klein JP, Sobocinski KA, Rowlings PA, Horowitz MM. Impact of donor type on outcome of bone marrow transplantation for Wiskott-Aldrich syndrome: collaborative study of the International Bone Marrow Transplant Registry and the National Marrow Donor Program. Blood. 2001;97(6):1598–1603. doi: 10.1182/blood.V97.6.1598.
    1. Antoine C, Muller S, Cant A, Cavazzana-Calvo M, Veys P, Vossen J, et al. Long-term survival and transplantation of haemopoietic stem cells for immunodeficiencies: report of the European experience 1968-99. Lancet. 2003;361(9357):553–560. doi: 10.1016/S0140-6736(03)12513-5.
    1. Marsh RA, Rao MB, Gefen A, Bellman D, Mehta PA, Khandelwal P, Chandra S, Jodele S, Myers KC, Grimley M, Dandoy C, el-Bietar J, Kumar AR, Leemhuis T, Zhang K, Bleesing JJ, Jordan MB, Filipovich AH, Davies SM. Experience with alemtuzumab, fludarabine, and melphalan reduced-intensity conditioning hematopoietic cell transplantation in patients with nonmalignant diseases reveals good outcomes and that the risk of mixed chimerism depends on underlying disease, stem cell source, and alemtuzumab regimen. Biol Blood Marrow Transplant. 2015;21(8):1460–1470. doi: 10.1016/j.bbmt.2015.04.009.
    1. Hagin D, Burroughs L, Torgerson TR. Hematopoietic stem cell transplant for immune deficiency and immune dysregulation disorders. Immunol Allergy Clin N Am. 2015;35(4):695–711. doi: 10.1016/j.iac.2015.07.010.
    1. Dinardo L, Brown V, Perez E, Bunin N, Sullivan KE. A single-center study of hematopoietic stem cell transplantation for primary immune deficiencies (PIDD) Pediatr Transplant. 2012;16(1):63–72. doi: 10.1111/j.1399-3046.2011.01606.x.
    1. Allen CE, Marsh R, Dawson P, Bollard CM, Shenoy S, Roehrs P, Hanna R, Burroughs L, Kean L, Talano JA, Schultz KR, Pai SY, Baker KS, Andolina JR, Stenger EO, Connelly J, Ramirez A, Bryant C, Eapen M, Pulsipher MA. Reduced-intensity conditioning for hematopoietic cell transplant for HLH and primary immune deficiencies. Blood. 2018;132(13):1438–1451. doi: 10.1182/blood-2018-01-828277.
    1. Klein OR, Chen AR, Gamper C, Loeb D, Zambidis E, Llosa N, Huo J, Dezern AE, Steppan D, Robey N, Holuba MJ, Cooke KR, Symons HJ. Alternative-donor hematopoietic stem cell transplantation with post-transplantation cyclophosphamide for nonmalignant disorders. Biol Blood Marrow Transplant. 2016;22(5):895–901. doi: 10.1016/j.bbmt.2016.02.001.
    1. Mallhi KK, Srikanthan MA, Baker KK, Frangoul HA, Torgerson TR, Petrovic A, et al. HLA-haploidentical hematopoietic cell transplantation for treatment of nonmalignant diseases using nonmyeloablative conditioning and post-transplant cyclophosphamide. Biol Blood Marrow Transplant. 2020;26(7):1332–41.
    1. Rao K, Adams S, Qasim W, Allwood Z, Worth A, Silva J, Lucchini G, Chiesa R, Veys P, Amrolia P. Effect of stem cell source on long-term chimerism and event-free survival in children with primary immunodeficiency disorders after fludarabine and melphalan conditioning regimen. J Allergy Clin Immunol. 2016;138(4):1152–1160. doi: 10.1016/j.jaci.2016.01.053.
    1. Przepiorka D, Weisdorf D, Martin P, Klingemann H, Beatty P, Hows J, et al. 1994 Consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;6(15):825–828.
    1. Jagasia MH, Greinix HT, Arora M, Williams KM, Wolff D, Cowen EW, et al. National Institutes of Health Consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21(3):389–401.e1. doi: 10.1016/j.bbmt.2014.12.001.
    1. Filipovich AH, Weisdorf D, Pavletic S, Socie G, Wingard JR, Lee SJ, Martin P, Chien J, Przepiorka D, Couriel D, Cowen EW, Dinndorf P, Farrell A, Hartzman R, Henslee-Downey J, Jacobsohn D, McDonald G, Mittleman B, Rizzo JD, Robinson M, Schubert M, Schultz K, Shulman H, Turner M, Vogelsang G, Flowers MED. National Institutes of Health Consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2005;11(12):945–956. doi: 10.1016/j.bbmt.2005.09.004.
    1. De Pauw B, Walsh TJ, Donnelly JP, Stevens DA, Edwards JE, Calandra T, et al. Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin Infect Dis. 2008;46(12):1813–1821. doi: 10.1086/588660.
    1. Kaplan E, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53:457–480. doi: 10.1080/01621459.1958.10501452.
    1. Gooley TA, Leisenring W, Crowley J, Storer BE. Estimation of failure probabilities in the presence of competing risks: new representations of old estimators. Stat Med. 1999;18(6):695–706. doi: 10.1002/(SICI)1097-0258(19990330)18:6<695::AID-SIM60>;2-O.
    1. Jones R, Lee K, Beschorner W, Vogel V, Grochow L, Braine H, et al. Venoocclusive disease of the liver following bone marrow transplantation. Transplantation. 1987;6(44):778–783. doi: 10.1097/00007890-198712000-00011.
    1. Corbacioglu S, Cesaro S, Faraci M, Valteau-Couanet D, Gruhn B, Rovelli A, Boelens JJ, Hewitt A, Schrum J, Schulz AS, Müller I, Stein J, Wynn R, Greil J, Sykora KW, Matthes-Martin S, Führer M, O'Meara A, Toporski J, Sedlacek P, Schlegel PG, Ehlert K, Fasth A, Winiarski J, Arvidson J, Mauz-Körholz C, Ozsahin H, Schrauder A, Bader P, Massaro J, D'Agostino R, Hoyle M, Iacobelli M, Debatin KM, Peters C, Dini G. Defibrotide for prophylaxis of hepatic veno-occlusive disease in paediatric haemopoietic stem-cell transplantation: an open-label, phase 3, randomised controlled trial. Lancet. 2012;379(9823):1301–1309. doi: 10.1016/S0140-6736(11)61938-7.
    1. Imus PH, Blackford AL, Bettinotti M, Luznik L, Fuchs EJ, Huff CA, et al. Severe cytokine release syndrome after haploidentical peripheral blood stem cell transplantation. Biol Blood Marrow Transplant. 2019;25(12):2431–37
    1. McCurdy SR, Muth ST, Tsai HL, Symons HJ, Huff CA, Matsui WH, et al. Early fever after haploidentical bone marrow transplantation correlates with class II HLA-mismatching and myeloablation but not outcomes. Biol Blood Marrow Transplant. 2018;24(10):2056–2064. doi: 10.1016/j.bbmt.2018.06.004.
    1. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA, Mackall CL. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–195. doi: 10.1182/blood-2014-05-552729.
    1. Kim HT, Armand P, Frederick D, Andler E, Cutler C, Koreth J, Alyea EP, III, Antin JH, Soiffer RJ, Ritz J, Ho VT. Absolute lymphocyte count recovery after allogeneic hematopoietic stem cell transplantation predicts clinical outcome. Biol Blood Marrow Transplant. 2015;21(5):873–880. doi: 10.1016/j.bbmt.2015.01.019.
    1. Bayraktar UD, Milton DR, Guindani M, Rondon G, Chen J, Al-Atrash G, et al. Optimal threshold and time of absolute lymphocyte count assessment for outcome prediction after bone marrow transplantation. Biol Blood Marrow Transplant. 2016;22(3):505–513. doi: 10.1016/j.bbmt.2015.10.020.
    1. Jacoby E, Chen A, Loeb DM, Gamper CJ, Zambidis E, Llosa NJ, Huo J, Cooke KR, Jones R, Fuchs E, Luznik L, Symons HJ. Single-agent post-transplantation cyclophosphamide as graft-versus-host disease prophylaxis after human leukocyte antigen-matched related bone marrow transplantation for pediatric and young adult patients with hematologic malignancies. Biol Blood Marrow Transplant. 2016;22(1):112–118. doi: 10.1016/j.bbmt.2015.08.034.
    1. McCurdy SR, Kanakry CG, Tsai HL, Gojo I, Smith BD, Gladstone DE, et al. Development of grade II acute graft-versus-host disease is associated with improved survival after myeloablative HLA-matched bone marrow transplantation using single-agent post-transplant cyclophosphamide. Biol Blood Marrow Transplant. 2019;25(6):1128–1135. doi: 10.1016/j.bbmt.2018.12.767.
    1. Luznik L, Bolaños-Meade J, Zahurak M, Chen AR, Smith BD, Brodsky R, Huff CA, Borrello I, Matsui W, Powell JD, Kasamon Y, Goodman SN, Hess A, Levitsky HI, Ambinder RF, Jones RJ, Fuchs EJ. High-dose cyclophosphamide as single-agent, short-course prophylaxis of graft-versus-host disease. Blood. 2010;115(16):3224–3230. doi: 10.1182/blood-2009-11-251595.
    1. Bolaños-Meade J, Reshef R, Fraser R, Fei M, Abhyankar S, Al-Kadhimi Z, et al. Three prophylaxis regimens (tacrolimus, mycophenolate mofetil, and cyclophosphamide; tacrolimus, methotrexate, and bortezomib; or tacrolimus, methotrexate, and maraviroc) versus tacrolimus and methotrexate for prevention of graft-versus-host disease with haemopoietic cell transplantation with reduced-intensity conditioning: a randomised phase 2 trial with a non-randomised contemporaneous control group (BMT CTN 1203) Lancet Haematol. 2019;6(3):e132–ee43. doi: 10.1016/S2352-3026(18)30221-7.
    1. Bolanos-Meade J, Fuchs EJ, Luznik L, Lanzkron SM, Gamper CJ, Jones RJ, et al. HLA-haploidentical bone marrow transplantation with posttransplant cyclophosphamide expands the donor pool for patients with sickle cell disease. Blood. 2012;120(22):4285–4291. doi: 10.1182/blood-2012-07-438408.
    1. Bonfim C, Ribeiro L, Nichele S, Loth G, Bitencourt M, Koliski A, Kuwahara C, Fabro AL, Pereira NF, Pilonetto D, Thakar M, Kiem HP, Page K, Fuchs EJ, Eapen M, Pasquini R. Haploidentical bone marrow transplantation with post-transplant cyclophosphamide for children and adolescents with Fanconi anemia. Biol Blood Marrow Transplant. 2017;23(2):310–317. doi: 10.1016/j.bbmt.2016.11.006.
    1. DeZern AE, Zahurak M, Symons H, Cooke K, Jones RJ, Brodsky RA. Alternative donor transplantation with high-dose post-transplantation cyclophosphamide for refractory severe aplastic anemia. Biol Blood Marrow Transplant. 2017;23(3):498–504. doi: 10.1016/j.bbmt.2016.12.628.
    1. Luznik L, O'Donnell PV, Symons HJ, Chen AR, Leffell MS, Zahurak M, Gooley TA, Piantadosi S, Kaup M, Ambinder RF, Huff CA, Matsui W, Bolaños-Meade J, Borrello I, Powell JD, Harrington E, Warnock S, Flowers M, Brodsky RA, Sandmaier BM, Storb RF, Jones RJ, Fuchs EJ. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2008;14(6):641–650. doi: 10.1016/j.bbmt.2008.03.005.
    1. Dimitrova D, Gea-Banacloche J, Steinberg SM, Sadler JL, Hicks SN, Carroll E, et al. Prospective study of a novel, radiation-free, reduced-intensity bone marrow transplantation platform for primary immunodeficiency diseases. Biol Blood Marrow Transplant. 2020;26(1):94–106.
    1. Gungor T, Teira P, Slatter M, Stussi G, Stepensky P, Moshous D, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet. 2014;383(9915):436–448. doi: 10.1016/S0140-6736(13)62069-3.
    1. Bertaina A, Zecca M, Buldini B, Sacchi N, Algeri M, Saglio F, Perotti C, Gallina AM, Bertaina V, Lanino E, Prete A, Barberi W, Tumino M, Favre C, Cesaro S, del Bufalo F, Ripaldi M, Boghen S, Casazza G, Rabusin M, Balduzzi A, Fagioli F, Pagliara D, Locatelli F. Unrelated donor vs HLA-haploidentical α/β T-cell- and B-cell-depleted HSCT in children with acute leukemia. Blood. 2018;132(24):2594–2607. doi: 10.1182/blood-2018-07-861575.
    1. Rubio MT, D'Aveni-Piney M, Labopin M, Hamladji R-M, Sanz MA, Blaise D, et al. Impact of in vivo T cell depletion in HLA-identical allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission conditioned with a fludarabine iv-busulfan myeloablative regimen: a report from the EBMT Acute Leukemia Working Party. J Hematol Oncol. 2017;10(1):31. doi: 10.1186/s13045-016-0389-4.
    1. Shah RM, Elfeky R, Nademi Z, Qasim W, Amrolia P, Chiesa R, et al. T-cell receptor αβ(+) and CD19(+) cell-depleted haploidentical and mismatched hematopoietic stem cell transplantation in primary immune deficiency. J Allergy Clin Immunol. 2018;141(4):1417–26.e1. doi: 10.1016/j.jaci.2017.07.008.
    1. Balashov D, Shcherbina A, Maschan M, Trakhtman P, Skvortsova Y, Shelikhova L, Laberko A, Livshits A, Novichkova G, Maschan A. Single-center experience of unrelated and haploidentical stem cell transplantation with TCRαβ and CD19 depletion in children with primary immunodeficiency syndromes. Biol Blood Marrow Transplant. 2015;21(11):1955–1962. doi: 10.1016/j.bbmt.2015.07.008.
    1. Patriarca F, Luznik L, Medeot M, Zecca M, Bacigalupo A, Di Bartolomeo P, et al. Experts’ considerations on HLA-haploidentical stem cell transplantation. Eur J Haematol. 2014;93(3):187–197. doi: 10.1111/ejh.12322.
    1. Parikh SH, Mendizabal A, Benjamin CL, Komanduri KV, Antony J, Petrovic A, Hale G, Driscoll TA, Martin PL, Page KM, Flickinger K, Moffet J, Niedzwiecki D, Kurtzberg J, Szabolcs P. A novel reduced-intensity conditioning regimen for unrelated umbilical cord blood transplantation in children with nonmalignant diseases. Biol Blood Marrow Transplant. 2014;20(3):326–336. doi: 10.1016/j.bbmt.2013.11.021.

Source: PubMed

3
Suscribir