• This first multicenter trial of MAC haplo-BMT with PTCy in pediatric patients with acute leukemias ahowed 0% TRM and 0% grade 3 or 4 aGVHD.

  • Our study shows low CuIs of grade 2 aGVHD and moderate-to-severe cGVHD and similar relapse rates as those of matched-donor BMT.

Promising results have been reported for adult patients with high-risk hematologic malignancies undergoing haploidentical bone marrow transplant (haploBMT) with posttransplant cyclophosphamide (PTCy). To our knowledge, we report results from the first multicenter trial for pediatric and young adult patients with high-risk acute leukemias and myelodysplastic syndrome (MDS) in the Pediatric Transplantation and Cellular Therapy Consortium. Nine centers performed transplants in 32 patients having acute leukemias or MDS, with myeloablative conditioning (MAC), haploBMT with PTCy, mycophenolate mofetil, and tacrolimus. The median patient age was 12 years. Diagnoses included AML (15), ALL (11), mixed-lineage leukemia (1), and MDS (5). Transplant-related mortality (TRM) at 180 days was 0%. The cumulative incidence (CuI) of grade 2 acute graft-versus-host disease (aGVHD) on day 100 was 13%. No patients developed grades 3-4 aGVHD. The CuI of moderate-to-severe chronic GVHD (cGVHD) at 1 year was 4%. Donor engraftment occurred in 27 patients (84%). Primary graft failures included 3 patients who received suboptimal bone marrow grafts; all successfully engrafted after second transplants. The CuI of relapse at 1 year was 32%, with more relapse among patients MRD positive pre-BMT vs MRD negative. Overall survival rates at 1 and 2 years were 77% and 73%, and event-free survival rate at 1 and 2 years were 68% and 64%. There was no TRM or severe aGVHD, low cGVHD, and favorable relapse and survival rates. This successful pilot trial has led to a phase 3 trial comparing MAC haploBMT vs HLA-matched unrelated donor BMT in the Children’s Oncology Group. This trial was registered at www.clinicaltrials.gov as #NCT02120157.

HLA-haploidentical bone marrow transplant (haplo-BMT) has increased significantly in the past decade with the development of novel methods to control powerful allogeneic reactions generated in the HLA-mismatched setting. With the success of posttransplant cyclophosphamide1-11 (PTCy) and other novel graft-versus-host disease (GVHD) prophylaxis strategies12-16 in allowing for safe and effective mismatched donor transplants, almost everyone in need now has a donor option. Worldwide, studies primarily in adult patients with hematologic malignancy have shown that related haplo-BMT with PTCy provides results similar to those seen with both matched related17-20 and matched unrelated donors21-25 (MUDs). Haplo-BMT with PTCy is an affordable option readily adaptable to clinical settings around the globe and has helped fill the donor availability gap for racial and ethnic minorities. In addition, the incidence of donor-derived malignancies is not higher with PTCy than without it,26 and there is no increased risk of Epstein-Barr virus (EBV) posttransplant lymphoproliferative disease.26,27 Although haplo-BMT with PTCy has gained traction for pediatric patients with leukemia, particularly in single-center studies,28-37 there is a paucity of prospective, multicenter trial data. As such, enthusiasm for using haploBMT with PTCy in younger patients lags behind our adult colleagues.

Much of the available literature on using haplo-BMT with PTCy for adult hematologic malignancies is in the context of reduced-intensity conditioning or nonmyeloablative (NMA) conditioning regimens. In contrast, those performing pediatric BMTs generally use myeloablative conditioning (MAC), with full-dose total-body irradiation (TBI)-based conditioning for acute lymphoblastic leukemia (ALL) and chemotherapy-based conditioning for acute myeloid leukemia (AML). Potential differences in disease biology and graft source (peripheral blood vs bone marrow) add additional concern that data of adult haplo-BMT with PTCy are not directly translatable to pediatric hematologic malignancies.

Herein, to our knowledge, we report the results of the first prospective international multicenter consortium pilot trial of haplo-BMT with PTCy, using MAC in children, and adolescents and young adults (AYAs) with high-risk hematologic malignancies.

Study design

This was a Pediatric Transplantation and Cellular Therapy Consortium international, phase 2, single-arm, prospective clinical trial (#NCT02120157) using MAC T-cell–replete haplo-BMT with PTCy in children and AYA with high-risk leukemias. The study protocol was approved by the institutional review board at each participating center and by the Johns Hopkins Medicine Institutional Review Board. Written informed consent for all patients was obtained in accordance with the Declaration of Helsinki before the initiation of conditioning therapy. The primary objective was transplant-related mortality (TRM) at 180 days (TRM180), with the hypothesis that TRM180 would be <17%. Secondary end points included donor engraftment, time to platelet and neutrophil recovery, acute GVHD (aGVHD), chronic GVHD (cGVHD), event-free survival (EFS), overall survival (OS), and GVHD relapse–free survival (GRFS).

A total of 32 patients were enrolled in this pilot trial between July 2015 and November 2017. Patients aged between 6 months and 25 years with high-risk leukemia, as previously defined within the Children’s Oncology Group trials, in complete remission (defined as morphology with <5% blasts and no morphological characteristics of acute leukemia in a bone marrow with >20% cellularity), or with myelodysplastic syndrome (MDS) and without an HLA-matched sibling donor were eligible. Additional inclusion criteria included left ventricular ejection fraction >50%; for patients receiving a nonTBI regimen, a forced expiratory volume in 1 second and forced vital capacity ≥50% of predicted and for patients receiving a TBI-based preparative regimen, ≥60%; total bilirubin level <2 mg/dL, alanine aminotransferase or aspartate aminotransferase levels <3 × the laboratory upper limits of normal, and creatinine clearance >60 mL/min; and no evidence of anti–donor HLA antibodies. Grade 3 to 5 adverse events (AEs) possibly, probably, or definitely related to PTCy were collected, as were grade 3 to 5 infections, serious AEs, and unanticipated grade 3 to 5 events.

Transplant platforms

All patients received a MAC regimen with busulfan/cyclophosphamide (Bu/Cy), except those with ALL who received a Cy/TBI preparative regimen. For the patients receiving Bu/Cy (Figure 1A), Bu was given every 6 hours for 4 consecutive days, followed by Cy at 50 mg/kg per day for 2 successive days. Bu dosing was started at either 0.8 mg/kg per dose, 32 mg/m2 per dose, or 1.1 mg/kg per dose IV, in accordance with the age and weight guidelines. Dosing was adjusted for the fifth and subsequent doses based on measured pharmacokinetic variables to achieve a targeted area under the concentration curve from 800 to 1400 mmol × min/L (∼53-92 mg × h/L). Seizure prophylaxis with levetiracetam was administered for all patients receiving Bu aged >10 years. For the patients receiving Cy/TBI (Figure 1B), Cy 50 mg/kg per day was given for 2 consecutive days followed by TBI, with a total dose of 1200 cGy (either 200 cGy twice a day for 3 days beginning on day −3 or 150 cGy twice a day for 4 days starting on day −4). On day 0, patients received unmanipulated bone marrow at least 24 hours after the last dose of Cy. Allografts comprised unmanipulated bone marrow from an HLA-haploidentical (matched for at least 1 allele each of HLA-A, HLA-B, HLA-C, HLA-DRB1, and HLA-DQB1) first-degree relative (full or half sibling, parent, or child), with a targeted dose of 4 × 108 marrow total nucleated cells per kilogram (TNC/kg) of recipient ideal body weight (IBW) and a recommended minimum yield of 2.5 × 108 marrow TNC/kg of recipient IBW. All patients received Cy 50 mg/kg per day on posttransplant days 3 and 4 (on day 3, Cy was given 60-72 hours after the start of bone marrow infusion). Mesna was administered with high-dose Cy at >80% of Cy dosing, per institutional standards. Additional GVHD prophylaxis consisted of mycophenolate mofetil 15 mg/kg orally, 3 times per day (maximum total daily dose, 3 g) from days 5 to 35, and tacrolimus 0.015 mg/kg per dose 2 times daily, adjusted to maintain a serum trough level of 5 to 15 ng/mL (or institutional equivalent), initially given from days 5 to 180 but subsequently ending as early as day 60 after the data with early cessation of tacrolimus were published.38 Tacrolimus was given IV until the patient tolerated oral medications. With this protocol, filgrastim was not routinely given before engraftment. Centers were instructed to notify the principal investigator if filgrastim was initiated and the reason for its start. An example of a reason to start filgrastim provided in the protocol was neutropenia with severe infection.

Figure 1.

Conditioning regimens. (A) Chemotherapy-based; (B) TBI-based.

Figure 1.

Conditioning regimens. (A) Chemotherapy-based; (B) TBI-based.

Close modal

Supportive care was provided per standard institutional practice. Post-BMT central nervous system prophylaxis was per institutional practice.

Definitions and end points

Neutrophil engraftment was defined as the number of days from BMT to the first of 3 consecutive days with an absolute neutrophil count >0.5 × 109/L. Platelet recovery time was defined as the number of days from BMT until the platelet count was >20 × 109/L without platelet transfusion in the preceding 7 days. Primary graft failure was defined as <5% donor chimerism in the blood or bone marrow by ∼day 60 and upon all subsequent measurements. Secondary graft failure was determined based on an initial achievement of >5% donor chimerism, followed by its sustained loss with <5% donor chimerism in the blood or bone marrow. The definition and grading of aGVHD and cGVHD were in accordance with the Blood and Marrow Transplant Clinical Transplant Network’s Manual of Procedures.39 Pretransplant disease evaluation included, at minimum, bone marrow aspirate and flow cytometry. Minimal residual disease (MRD) was defined as any evidence of malignant cells via flow cytometry, fluorescence in-situ hybridization, polymerase chain reaction, or other techniques without morphological or cytogenetic evidence of disease in the blood or bone marrow.

Per the protocol, posttransplant disease evaluation was first performed on day 60. Relapse was defined as the reappearance of leukemic blast cells in the peripheral blood; or >5% blasts in the bone marrow, not attributable to another cause (eg, bone marrow regeneration); the appearance of previous new dysplastic changes (MDS-specific) within the bone marrow; the development of extramedullary leukemia or the presence of leukemic cells in the cerebral spinal fluid, or the reappearance of cytogenetic or molecular abnormalities present before transplant. Standard relapse-prevention therapies after BMT, such as tyrosine kinase inhibitors for Ph+ leukemias, and sorafenib for FLT3–internal tandem duplication leukemias, were allowed.

EFS was defined as the time from transplant to the first occurrence of any disease progression or relapse, an unplanned therapeutic maneuver for disease persistence, or death from any cause. Data of patients without any event were censored at the last clinic visit at which they were known to be disease free. OS was defined as the time from transplant to death from any cause. TRM was defined as death without evidence of disease progression or relapse after transplant. TRM was considered a competing risk for relapse and vice versa. Competing risks for GVHD included graft failure, relapse, or death. GRFS was defined as the time from BMT to the onset of grade 3 or 4 aGVHD, cGVHD requiring systemic immunosuppression, disease relapse, or death.

Statistical analysis

The proportion of patients with TRM180 was reported with an exact 95% confidence interval (CI). The cumulative incidence (CuI) of relapse and GVHD were estimated via subdistribution functions with 95% CIs via the Aalen method.40,41 Specifically, relapse was further compared using disease (myeloid vs lymphoid), disease risk index (DRI; low, intermediate, high), and pre-BMT MRD (positive vs negative). EFS and OS were estimated with the Kaplan and Meier method, and patient subgroups were compared using the log-rank statistic or Cox proportional hazards regression models. Outcomes with competing risks were assessed with the proportional subdistribution hazard regression model for competing risks.42 Statistical analyses were performed using SAS version 9.2 (SAS Institute Inc, Cary, NC) and R version 4.2.2 (R Foundation for Statistical Computing, Vienna, Austria).

Patient, donor, and allograft characteristics

Patient characteristics are summarized in Table 1. In total, 32 patients were enrolled in the study. The median recipient age was 12 years (range, 1-23 years), and the median donor age was 35 years (range, 4-53 years). Of the patients, 18 (59%) were male. The median HLA mismatch was 5 of 10 alleles. A total of 24 patients (75%) received their graft from parents, and 8 (25%) from siblings. The median (interquartile range [IQR]) TNC/kg of recipient IBW was 5.2 × 108 (2.8 × 108-6.3 × 108), and the median (IQR) of CD34+ cells per kilogram of recipient IBW was 4 × 106 (2.8 × 106-6.3 × 106). All patients received T-cell–replete, haploidentical bone marrow. The conditioning regimen for all patients consisted of MAC therapy. Twenty patients (62%) received a Bu-based preparative regimen, whereas 12 received a TBI-based regimen. The Center for International Blood and Marrow Transplant Research Pediatric DRI assignments43 included 1 patient (3%) at the low risk, 16 patients (50%) at intermediate risk, and 15 patients (47%) at high risk. Four patients with AML and 1 with ALL had evidence of MRD before BMT. Five patients with MDS had evidence of disease, including 3 with multilineage dysplasia and 2 with excess blasts.

Table 1.

Patient, donor, and graft characteristics

VariableEntire cohort (n = 32)
Sex, n (%)  
Male 18 (59) 
Female 14 (41) 
Median age, Y (range)  
Recipient 12 (1-23) 
Donor 35 (4-53) 
MRD results, n (%)  
Negative 22 (69) 
Positive 10 (31) 
DRI, n (%)  
Low/intermediate 17 (53) 
High 15 (47) 
Diagnosis, n (%)  
AML 15 (47) 
MDS 5 (16) 
ALL 11 (34) 
Mixed lineage 1 (3) 
Donor relation, n (%)  
Sibling 8 (25) 
Parent 24 (75) 
HLA alleles, n (range)  
Median HLA match 5/10 (5-8/10) 
No. of HLA alleles match n (%) 
8/10 2 (6) 
7/10 2 (6) 
6/10 6 (19) 
5/10 22 (69) 
Cell dose infused, n (range)  
TNC/kg BM infused 4.8 × 108 (1.2 × 108-6.8 × 108
CD34+/kg BM infused 4.3 × 106 (0.9 × 106-9.3 × 106
Conditioning regimen, n (%)  
Bu/Cy 20 (62) 
Cy/TBI 12 (38) 
VariableEntire cohort (n = 32)
Sex, n (%)  
Male 18 (59) 
Female 14 (41) 
Median age, Y (range)  
Recipient 12 (1-23) 
Donor 35 (4-53) 
MRD results, n (%)  
Negative 22 (69) 
Positive 10 (31) 
DRI, n (%)  
Low/intermediate 17 (53) 
High 15 (47) 
Diagnosis, n (%)  
AML 15 (47) 
MDS 5 (16) 
ALL 11 (34) 
Mixed lineage 1 (3) 
Donor relation, n (%)  
Sibling 8 (25) 
Parent 24 (75) 
HLA alleles, n (range)  
Median HLA match 5/10 (5-8/10) 
No. of HLA alleles match n (%) 
8/10 2 (6) 
7/10 2 (6) 
6/10 6 (19) 
5/10 22 (69) 
Cell dose infused, n (range)  
TNC/kg BM infused 4.8 × 108 (1.2 × 108-6.8 × 108
CD34+/kg BM infused 4.3 × 106 (0.9 × 106-9.3 × 106
Conditioning regimen, n (%)  
Bu/Cy 20 (62) 
Cy/TBI 12 (38) 

NRM

The primary objective of our study was TRM180. The incidence of TRM at 6 and 12 months was 0% (exact 95% CI, 0-10.9).

Engraftment/chimerism

The median day of neutrophil engraftment was 22 days (range, 14-58 days) and 21 days (range, 12-44 days) for platelets. Engraftment of donor cells with donor chimerism of >95% at 60 days was achieved in 27 patients (84%). Of these patients, 2 received filgrastim (both for slow count recovery at the discretion of the treating physicians).

Patients who did not achieve >95% donor chimerism included 3 patients with MDS, who did not undergo pretransplant disease reduction, and 2 patients with AML. The median (IQR) TNC/kg of recipient IBW for those patients who did not achieve engraftment was statistically significantly lower at 2.6 × 108 (2.0 × 108-2.9 × 108) than those who achieved engraftment, at 5.2 × 108 (3.9 × 108-6.7 × 108; P = .0147). All patients who failed to achieve engraftment received a salvage haplo-BMT, which engrafted, and at the time of analysis, all 5 patients were alive, 4 of them with no evidence of disease.

GVHD, relapse, and transplant-related complications

The CuI of grade 2 aGVHD at 1 year was 13% (95% CI, 1-25; Figure 2A). No patients experienced grade 3 to 4 aGVHD. Two patients developed moderate-to-severe cGVHD with a CuI at 1 and 2 years of 3% (95% CI, 0-10) and 11% (95% CI, 0-28), respectively (Figure 2B). The CuI of relapse at 1 and 2 years was 32% (95% CI, 15-48) and 36% (95% CI, 18-54), respectively (Figure 3A). Although not statistically significant, patients with pre-BMT positive MRD results and high DRI had higher relapse incidences than patients with MRD-negative results and those with a low/intermediate DRI (Figure 3B-C). The CuI of relapse at 1 year in patients with pre-BMT MRD positive results was 50% (95% CI, 17-83) compared with 23% (95% CI, 5-42) in patients with MRD-negative results. Patients with a high DRI had a CuI of relapse at 1 year of 41% (95% CI, 15-68) vs 24% (95% CI, 3-44) in patients with a low/Intermediate DRI. The estimated CuI of relapse at 1 year in ALL was 18% (95% CI, 6-42), in AML it was 35% (95% CI, 9-60), and in AML+ MDS it was 41% (95% CI, 18-64) (Figure 3D). Infections were the most common transplant-related complication and included catheter-associated grade 3–related infections (n = 5), enterocolitis (n = 2), Clostridium difficile (n = 2), bacteremia (n = 3), sepsis (n = 1), cytomegalovirus reactivation (n = 2), EBV reactivation (n = 1), parainfluenza (n = 1), and varicella-zoster virus infection (n = 1). Sinusoidal obstruction syndrome (SOS) occurred in 4 patients (common terminology criteria for AEs, grade 3 = 3 and grade 4 = 1), which fully resolved. Two patients developed grade 3 hemorrhagic cystitis, both cases BK virus-positive that presented on days 27 and 40 after transplantation, respectively. Grade 3 mucositis was reported in 13 patients. Most importantly, none of these complications resulted in mortality. No patients with transplant-associated thrombotic microangiopathy (TA-TMA) were reported.

Figure 2.

CuI of GVHD.

Figure 3.

Relapse. (A) CuI of relapse; (B) CuI of relapse based on the MRD results; (C) CuI of relapse based on the DRI; and (D) CuI of based on the by disease type.

Figure 3.

Relapse. (A) CuI of relapse; (B) CuI of relapse based on the MRD results; (C) CuI of relapse based on the DRI; and (D) CuI of based on the by disease type.

Close modal

OS, EFS, and GRFS

The median follow-up was 15 months, ranging from 5 months to 3.1 years. The OS probabilities at 1 and 2 years were 77% (95% CI, 64-94) and 73% (95% CI, 59-91), respectively. The estimated 1-year and 2-year EFSs were 68% (95% CI, 54-87) and 64% (95% CI, 49-84), respectively, and GRFSs were 65% (95% CI, 50-84) and 52% (95% CI, 34-79), respectively (Figure 4).

Figure 4.

Probabilities of OS, EFS, and GRFS.

Figure 4.

Probabilities of OS, EFS, and GRFS.

Close modal

We report the results from the first international, multicenter phase 2 Pediatric Transplantation and Cellular Therapy Consortium trial using MAC, T-cell–replete haplo-BMT, and PTCy in children and AYA patients with high-risk hematologic malignancies. Impressively, for our primary end point, we had no TRM, supporting the safety and tolerability of this regimen. Albeit with small numbers, having a CuI of 0% TRM compares very favorably with other published regimens.15,44,45 Moreover, the CuIs of severe aGVHD and moderate-to-severe cGVHD requiring systemic immunosuppression are lower than those of other successful BMT platforms and donor sources.15,45-48 This provides an opportunity to continue to shorten the duration of posttransplant tacrolimus for all patients in future trials using MAC regimens, as has already been safely accomplished in the NMA setting.38 This is especially important given that relapse remains challenging after BMT regardless of donor type, graft source, or conditioning regimen intensity. Incorporating novel agents early after the transplant to prevent or treat relapse is optimized when patients discontinue immunosuppression and are without GVHD.49 We did not collect data on the use of posttransplant relapse–prevention therapy in this trial.

Several aspects of our data warrant further discussion. Firstly, graft failure was higher than expected in this trial: 3 patients with untreated MDS and 2 with AML, all receiving chemotherapy-based conditioning with Bu/Cy, failed to engraft. These patients underscore 2 key factors when using haploidentical donors: (1) the importance of graft cell dose and (2) the challenge of engraftment for patients who are naive to any cytoreductive therapy, even after MAC. All 5 patients received a TNC/kg cell dose lower than the recommended target yield; in fact, the 2 patients with AML received a TNC/kg lower than the minimum recommended target yield (2.5 × 108 TNC/kg of recipient IBW). The necessity of adequate cell dose to facilitate engraftment is evident in this small data set, as in other trials.50 Continuing to provide harvest training and education regarding the importance of graft cell dose will be important for future trials. Three patients with MDS also had not received any pretransplant disease reduction therapy before BMT. Strategies to improve engraftment for these patients could also include using peripheral blood stem cells (PBSCs) or pretransplant therapy with agents such as azacytidine, as has been used in the NMA setting, or both.38,51 Ultimately, all 5 patients achieved engraftment after salvage haplo-BMT. Four of them received PBSCs, 2 from the same donor, 2 from a second donor, and one of the patients received bone marrow stem cells from the same donor. Salvage conditioning regimens included alemtuzumab (0.2 mg/kg per dose), fludarabine (flu; 30 mg/m2 per dose), Cy (2 grams/m2 per dose), and 200 cGy TBI (n = 2), all on day −1; antithymocyte globulin (3.5 mg/kg divided over 2 doses) and flu (200 mg/m2 divided over 5 doses; n = 1); flu (150 mg/m2 divided over 5 doses), Cy (28 mg/kg divided over 2 doses), and 200 cGy TBI (n = 1); and alemtuzumab, flu, and Cy (n = 1, doses not provided).

Secondly, the incidences of transplant-related complications in this study are similar to those in other published allogeneic BMT studies in the HLA-matched and haploBMT with PTCy settings.35,52-54 Infections were the most common transplant-related complication. They included bacterial infections; BK virus–associated hemorrhagic cystitis and viral reactivations of EBV, cytomegalovirus, and varicella-zoster virus with no end-organ disease. SOS was reported in 4 patients. In this pilot trial, we did not collect data of which SOS diagnostic criteria were used. Most importantly, none of these complications resulted in mortality. There were no reports of TA-TMA; it is possible that the collection of our AEs/serious AEs, as stated in “Methods,” was not conducive to capturing these patients, and we did not specifically ask centers to report TA-TMA in this trial. Thus, we cannot conclusively report its incidence. A prior publication using haplo-BMT with PTCy reported an extremely low TA-TMA incidence (1.4%).55 Therefore, it is also possible that there were no patients with TA-TMA.

The pediatric BMT community has expressed general concern regarding the CuI of relapse using PTCy. The CuI of relapse reported in published studies using haploidentical donors, MUDs, cord, and matched sibling donors has ranged from 12% to 70%15,44-46,51,56-60 with pre-BMT MRD positivity and a high DRI associated with a higher incidence of relapse. The CuI of relapse of 32% and 36% at 1 and 2 years, respectively, in our small, multicenter study, with lower but not statistically significant CuI of relapse in MRD negative and low-intermediate DRI patients, is directly comparable with that reported in the pediatric BMT published literature and does not support that haplo-BMT with PTCy is inferior. It is important to remember that more than one-third of our patients MRD positive before transplant, almost one-half had a high DRI, and patients with MDS with >10% blasts were eligible and enrolled. Other single-center haplo-BMT trials suggest similar supporting evidence.28,29,32-37 The current standard of discontinuing tacrolimus at day 60 after haplo-BMT was implemented toward the end of this trial, and consideration of PBSC grafts for patients with high-risk disease may provide additional relapse prevention.

Limitations of this study include patient heterogeneity and small numbers. It is difficult to directly compare our results with that of other haplo-BMT platforms, such as the megadose CD34+ selection61 and α-β T-cell–depleted haplo-BMT13,15 without randomized controlled trials. Nevertheless, the absolute number of longitudinal experiences with pediatric patients who have undergone haplo-BMT after MAC is low compared with the number of longitudinal experiences among those who have received MAC allogeneic BMT from HLA-matched donors and cord blood. This underscores the importance of continuing to study MAC haplo-BMT with PTCy, laying the groundwork for prospective randomized controlled BMT trials using MAC, different donor sources, and other haploidentical graft methods, especially for pediatric and AYA populations.

In conclusion, this first (to our knowledge) multicenter, international consortium trial demonstrates that myeloablative HLA–haplo-BMT with PTCy for pediatric and AYA high-risk acute leukemias offers a widely available platform that is safe and feasible. Our findings are an essential contribution to the literature, serving as background for the recently opened Children’s Oncology Group randomized controlled trial of haplo (using both PTCy and α-β T-cell–depleted platforms) vs MUD hematopoietic cell transplants in pediatric patients with high-risk hematologic malignancies.

Funding for this trial was provided by National Heart, Lung, and Blood Institute/National Cancer Institute grant UG1HL069254 and a Johnny Crisstopher Children’s Charitable Foundation St. Baldrick’s Consortium grant. M.A.P. was supported by National Cancer Institute grant P30CA040214.

Contribution: J.C.F.-P. interpreted data and wrote the manuscript; H.-L.T. and A.B. designed the trial, analyzed and interpreted data, performed the statistical analysis, and reviewed the manuscript; J. Dalal, M.E., M.A.P., S.S.K., D.L., L.L., and L.M. designed the trial, enrolled patients, and reviewed the manuscript; A.C., E.C., J. Davis, J.H., A.K., J.K., and B.O. enrolled patients and reviewed the manuscript; T.F. and M.A.P. designed the trial, interpreted data, and reviewed the manuscript; M.H. designed the trial, enrolled patients, and reviewed the manuscript; and H.J.S. designed the trial, enrolled patients, collected data, analyzed and interpreted data, and wrote the manuscript.

Conflict-of-interest disclosure: M.H. reports advisory board activities for Novartis, Mesoblast, Jazz Pharmaceuticals, and Kadmon Pharmaceuticals. D.L. reports research funding, consulting, and licensing fees from Kiadis Pharma. B.O. is a full-time employee of ImmunoGen Inc. M.A.P. reports advisory board activities for Novartis, Gentibio, bluebird bio, Vertex, Medexus, and Equillium; educational honoraria from Novartis; and study support from Miltenyi and Adaptive. H.J.S. reports speaker bureau activity for Jazz Pharmaceuticals. The remaining authors declare no competing financial interests.

Correspondence: Heather Symons, Pediatric Blood and Marrow Transplantation, Johns Hopkins University, 1650 Orleans St, CRB I 2M-52, Baltimore, MD 21287; e-mail: hsymons2@jhmi.edu.

1.
O'Donnell
PV
,
Luznik
L
,
Jones
RJ
, et al
.
Nonmyeloablative bone marrow transplantation from partially HLA-mismatched related donors using posttransplantation cyclophosphamide
.
Biol Blood Marrow Transplant
.
2002
. ;
8
(
7
):
377
-
386
.
2.
Paul
S
,
Zahurak
M
,
Luznik
L
, et al
.
Non-myeloablative allogeneic transplantation with post-transplant cyclophosphamide after immune checkpoint inhibition for classic Hodgkin lymphoma: a retrospective cohort study
.
Biol Blood Marrow Transplant
.
2020
. ;
26
(
9
):
1679
-
1688
.
3.
Luznik
L
,
O'Donnell
PV
,
Symons
HJ
, et al
.
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
.
4.
Brunstein
CG
,
Fuchs
EJ
,
Carter
SL
, et al
.
Alternative donor transplantation after reduced intensity conditioning: results of parallel phase 2 trials using partially HLA-mismatched related bone marrow or unrelated double umbilical cord blood grafts
.
Blood
.
2011
. ;
118
(
2
):
282
-
288
.
5.
Ciurea
SO
,
Mulanovich
V
,
Saliba
RM
, et al
.
Improved early outcomes using a t cell replete graft compared with t cell depleted haploidentical hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2012
. ;
18
(
12
):
1835
-
1844
.
6.
Raiola
AM
,
Dominietto
A
,
Ghiso
A
, et al
.
Unmanipulated haploidentical bone marrow transplantation and posttransplantation cyclophosphamide for hematologic malignancies after myeloablative conditioning
.
Biol Blood Marrow Transplant
.
2013
. ;
19
(
1
):
117
-
122
.
7.
Solomon
SR
,
Sizemore
CA
,
Sanacore
M
, et al
.
Haploidentical transplantation using t cell replete peripheral blood stem cells and myeloablative conditioning in patients with high-risk hematologic malignancies who lack conventional donors is well tolerated and produces excellent relapse-free survival: results of a prospective phase II trial
.
Biol Blood Marrow Transplant
.
2012
. ;
18
(
12
):
1859
-
1866
.
8.
Castagna
L
,
Crocchiolo
R
,
Furst
S
, et al
.
Bone marrow compared with peripheral blood stem cells for haploidentical transplantation with a nonmyeloablative conditioning regimen and post-transplantation cyclophosphamide
.
Biol Blood Marrow Transplant
.
2014
. ;
20
(
5
):
724
-
729
.
9.
Fuchs
EJ
,
O’Donnell
PV
,
Eapen
M
, et al
.
Double unrelated umbilical cord blood vs HLA-haploidentical bone marrow transplantation: the BMT CTN 1101 trial
.
Blood
.
2021
. ;
137
(
3
):
420
-
428
.
10.
O'Donnell
PV
,
Brunstein
CG
,
Fuchs
EJ
, et al
.
Umbilical cord blood or HLA-haploidentical transplantation: real-world outcomes versus randomized trial outcomes
.
Transplant Cell Ther
.
2022
. ;
28
(
2
):
109.e1
-
109.e8
.
11.
Bashey
A
,
Zhang
MJ
,
McCurdy
SR
, et al
.
Mobilized peripheral blood stem cells versus unstimulated bone marrow as a graft source for t-cell-replete haploidentical donor transplantation using post-transplant cyclophosphamide
.
J Clin Oncol
.
2017
. ;
35
(
26
):
3002
-
3009
.
12.
Bethge
WA
,
Eyrich
M
,
Mielke
S
, et al
.
Results of a multicenter phase I/II trial of TCRαβ and CD19-depleted haploidentical hematopoietic stem cell transplantation for adult and pediatric patients
.
Bone Marrow Transplant
.
2022
. ;
57
(
3
):
423
-
430
.
13.
Locatelli
F
,
Merli
P
,
Pagliara
D
, et al
.
Outcome of children with acute leukemia given HLA-haploidentical HSCT after αβ T-cell and B-cell depletion
.
Blood
.
2017
. ;
130
(
5
):
677
-
685
.
14.
Pulsipher
MA
,
Ahn
KW
,
Bunin
NJ
, et al
.
KIR-favorable TCR-αβ/CD19-depleted haploidentical HCT in children with ALL/AML/MDS: primary analysis of the PTCTC ONC1401 trial
.
Blood
.
2022
. ;
140
(
24
):
2556
-
2572
.
15.
Bertaina
A
,
Zecca
M
,
Buldini
B
, et al
.
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
.
16.
Shook
DR
,
Triplett
BM
,
Eldridge
PW
,
Kang
G
,
Srinivasan
A
,
Leung
W
.
Haploidentical stem cell transplantation augmented by CD45RA negative lymphocytes provides rapid engraftment and excellent tolerability
.
Pediatr Blood Cancer
.
2015
. ;
62
(
4
):
666
-
673
.
17.
Dreger
P
,
Sureda
A
,
Ahn
KW
, et al
.
PTCy-based haploidentical vs matched related or unrelated donor reduced-intensity conditioning transplant for DLBCL
.
Blood Adv
.
2019
. ;
3
(
3
):
360
-
369
.
18.
Ghosh
N
,
Karmali
R
,
Rocha
V
, et al
.
Reduced-intensity transplantation for lymphomas using haploidentical related donors versus HLA-matched sibling donors: a center for international blood and marrow transplant research analysis
.
J Clin Oncol
.
2016
. ;
34
(
26
):
3141
-
3149
.
19.
Martínez
C
,
Gayoso
J
,
Canals
C
, et al
.
Post-transplantation cyclophosphamide-based haploidentical transplantation as alternative to matched sibling or unrelated donor transplantation for Hodgkin lymphoma: a registry study of the lymphoma working party of the European society for blood and marrow transplantation
.
J Clin Oncol
.
2017
. ;
35
(
30
):
3425
-
3432
.
20.
Rashidi
A
,
Hamadani
M
,
Zhang
MJ
, et al
.
Outcomes of haploidentical vs matched sibling transplantation for acute myeloid leukemia in first complete remission
.
Blood Adv
.
2019
. ;
3
(
12
):
1826
-
1836
.
21.
Ciurea
SO
,
Zhang
MJ
,
Bacigalupo
AA
, et al
.
Haploidentical transplant with posttransplant cyclophosphamide vs matched unrelated donor transplant for acute myeloid leukemia
.
Blood
.
2015
. ;
126
(
8
):
1033
-
1040
.
22.
Kanate
AS
,
Mussetti
A
,
Kharfan-Dabaja
MA
, et al
.
Reduced-intensity transplantation for lymphomas using haploidentical related donors vs HLA-matched unrelated donors
.
Blood
.
2016
. ;
127
(
7
):
938
-
947
.
23.
Rubio
MT
,
Savani
BN
,
Labopin
M
, et al
.
Impact of conditioning intensity in T-replete haplo-identical stem cell transplantation for acute leukemia: a report from the acute leukemia working party of the EBMT
.
J Hematol Oncol
.
2016
. ;
9
(
1
):
25
.
24.
Saglio
F
,
Berger
M
,
Spadea
M
, et al
.
Haploidentical HSCT with post transplantation cyclophosphamide versus unrelated donor HSCT in pediatric patients affected by acute leukemia
.
Bone Marrow Transplant
.
2021
. ;
56
(
3
):
586
-
595
.
25.
Hong
KT
,
Park
HJ
,
Kim
BK
,
An
HY
,
Choi
JY
,
Kang
HJ
.
Post-transplantation cyclophosphamide-based haploidentical versus matched unrelated donor peripheral blood hematopoietic stem cell transplantation using myeloablative targeted busulfan-based conditioning for pediatric acute leukemia
.
Transplant Cell Ther
.
2022
. ;
28
(
4
):
195.e1
-
195.e7
.
26.
Majzner
RG
,
Mogri
H
,
Varadhan
R
, et al
.
Post-transplantation cyclophosphamide after bone marrow transplantation is not associated with an increased risk of donor-derived malignancy
.
Biol Blood Marrow Transplant
.
2017
. ;
23
(
4
):
612
-
617
.
27.
Kanakry
JA
,
Kasamon
YL
,
Bolaños-Meade
J
, et al
.
Absence of post-transplantation lymphoproliferative disorder after allogeneic blood or marrow transplantation using post-transplantation cyclophosphamide as graft-versus-host disease prophylaxis
.
Biol Blood Marrow Transplant
.
2013
. ;
19
(
10
):
1514
-
1517
.
28.
Katsanis
E
,
Sapp
LN
,
Reid
SC
,
Reddivalla
N
,
Stea
B
.
T-cell replete myeloablative haploidentical bone marrow transplantation is an effective option for pediatric and young adult patients with high-risk hematologic malignancies
.
Front Pediatr
.
2020
. ;
8
:
282
-
292
.
29.
Sanz
J
,
Galimard
JE
,
Labopin
M
, et al
.
Post-transplant cyclophosphamide after matched sibling, unrelated and haploidentical donor transplants in patients with acute myeloid leukemia: a comparative study of the ALWP EBMT
.
J Hematol Oncol
.
2020
. ;
13
(
1
):
46
-
58
.
30.
Yesilipek
MA
,
Uygun
V
,
Karasu
G
,
Daloglu
H
,
Dincer
Z
.
Haploidentical hematopoietic stem cell transplantation with post-transplant high-dose cyclophosphamide in high-risk children: A single-center study
.
Pediatr Transplant
.
2016
. ;
20
(
3
):
417
-
423
.
31.
Klein
OR
,
Buddenbaum
J
,
Tucker
N
, et al
.
Nonmyeloablative haploidentical bone marrow transplantation with post-transplantation cyclophosphamide for pediatric and young adult patients with high-risk hematologic malignancies
.
Biol Blood Marrow Transplant
.
2017
. ;
23
(
2
):
325
-
332
.
32.
Jaiswal
SR
,
Chakrabarti
A
,
Chatterjee
S
, et al
.
Haploidentical peripheral blood stem cell transplantation with post-transplantation cyclophosphamide in children with advanced acute leukemia with fludarabine-busulfan-and melphalan-based conditioning
.
Biol Blood Marrow Transplant
.
2016
. ;
22
(
3
):
499
-
504
.
33.
Pérez-Martínez
A
,
Ferreras
C
,
Pascual
A
, et al
.
Haploidentical transplantation in high-risk pediatric leukemia: a retrospective comparative analysis on behalf of the Spanish working Group for bone marrow transplantation in children (GETMON) and the Spanish Grupo for hematopoietic transplantation (GETH)
.
Am J Hematol
.
2020
. ;
95
(
1
):
28
-
37
.
34.
Berger
M
,
Lanino
E
,
Cesaro
S
, et al
.
Feasibility and outcome of haploidentical hematopoietic stem cell transplantation with post-transplant high-dose cyclophosphamide for children and adolescents with hematologic malignancies: an AIEOP-GITMO retrospective multicenter study
.
Biol Blood Marrow Transplant
.
2016
. ;
22
(
5
):
902
-
909
.
35.
Symons
HJ
,
Zahurak
M
,
Cao
Y
, et al
.
Myeloablative haploidentical BMT with posttransplant cyclophosphamide for hematologic malignancies in children and adults
.
Blood Adv
.
2020
. ;
4
(
16
):
3913
-
3925
.
36.
Sharma
A
,
Rastogi
N
,
Chatterjee
G
,
Kapoor
R
,
Nivargi
S
,
Yadav
SP
.
Haploidentical stem cell transplantation with post-transplant cyclophosphamide for pediatric acute leukemia is safe and effective
.
J Pediatr Hematol Oncol
.
2021
. ;
43
(
7
):
e1033
-
e1036
.
37.
Katsanis
E
,
Sapp
LN
,
Varner
N
,
Koza
S
,
Stea
B
,
Zeng
Y
.
Haploidentical bone marrow transplantation with post-transplant cyclophosphamide/bendamustine in pediatric and young adult patients with hematologic malignancies
.
Biol Blood Marrow Transplant
.
2018
. ;
24
(
10
):
2034
-
2039
.
38.
Kasamon
YL
,
Fuchs
EJ
,
Zahurak
M
, et al
.
Shortened-duration tacrolimus after nonmyeloablative, HLA-haploidentical bone marrow transplantation
.
Biol Blood Marrow Transplant
.
2018
. ;
24
(
5
):
1022
-
1028
.
39.
BMT-CTN
.
Technical Manual of Procedures. Version 3.0
. Accessed 19 March 2013. https://web.emmes.com/study/bmt2/public/MOP/BMT%20CTN%20Technical%20MOP%20v3.pdf.
40.
Aalen
O
.
Nonparametric estimation of partial transition probabilities in multiple decrement models
.
Ann Stat
.
1978
. ;
6
(
3
):
534
-
545
.
41.
Gray
RJ
.
A class of K-sample tests for comparing the cumulative incidence of a competing risk
.
Ann Stat
.
1988
. ;
16
(
3
):
1141
-
1154
.
42.
Fine
JP
,
Gray
RJ
.
A proportional hazards model for the subdistribution of a competing risk
.
J Am Stat Assoc
.
1999
. ;
94
(
446
):
496
-
509
.
43.
Qayed
M
,
Ahn
KW
,
Kitko
CL
, et al
.
A validated pediatric disease risk index for allogeneic hematopoietic cell transplantation
.
Blood
.
2021
. ;
137
(
7
):
983
-
993
.
44.
Wagner
JE
,
Eapen
M
,
Carter
S
, et al
.
One-unit versus two-unit cord-blood transplantation for hematologic cancers
.
N Engl J Med
.
2014
. ;
371
(
18
):
1685
-
1694
.
45.
Eapen
M
,
Logan
BR
,
Confer
DL
, et al
.
Peripheral blood grafts from unrelated donors are associated with increased acute and chronic graft-versus-host disease without improved survival
.
Biol Blood Marrow Transplant
.
2007
. ;
13
(
12
):
1461
-
1468
.
46.
Keating
AK
,
Langenhorst
J
,
Wagner
JE
, et al
.
The influence of stem cell source on transplant outcomes for pediatric patients with acute myeloid leukemia
.
Blood Adv
.
2019
. ;
3
(
7
):
1118
-
1128
.
47.
MacMillan
ML
,
Weisdorf
DJ
,
Brunstein
CG
, et al
.
Acute graft-versus-host disease after unrelated donor umbilical cord blood transplantation: analysis of risk factors
.
Blood
.
2009
. ;
113
(
11
):
2410
-
2415
.
48.
Ruggeri
A
,
Labopin
M
,
Bacigalupo
A
, et al
.
Post-transplant cyclophosphamide for graft-versus-host disease prophylaxis in HLA matched sibling or matched unrelated donor transplant for patients with acute leukemia, on behalf of ALWP-EBMT
.
J Hematol Oncol
.
2018
. ;
11
(
1
):
40
-
49
.
49.
Kreidieh
F
,
Abou Dalle
I
,
Moukalled
N
, et al
.
Relapse after allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia: an overview of prevention and treatment
.
Int J Hematol
.
2022
. ;
116
(
3
):
330
-
340
.
50.
DeZern
AE
,
Eapen
M
,
Wu
J
, et al
.
Haploidentical bone marrow transplantation in patients with relapsed or refractory severe aplastic anaemia in the USA (BMT CTN 1502): a multicentre, single-arm, phase 2 trial
.
Lancet Haematol
.
2022
. ;
9
(
9
):
e660
-
e669
.
51.
McCurdy
SR
,
Kanakry
JA
,
Showel
MM
, et al
.
Risk-stratified outcomes of nonmyeloablative HLA-haploidentical BMT with high-dose posttransplantation cyclophosphamide
.
Blood
.
2015
. ;
125
(
19
):
3024
-
3031
.
52.
Singh
A
,
Dandoy
CE
,
Chen
M
, et al
.
Post-transplantation cyclophosphamide is associated with an increase in non-cytomegalovirus herpesvirus infections in patients with acute leukemia and myelodysplastic syndrome
.
Transplant Cell Ther
.
2022
. ;
28
(
1
):
48.e1
. -48.e10.
53.
Esquirol
A
,
Pascual
MJ
,
Kwon
M
, et al
.
Severe infections and infection-related mortality in a large series of haploidentical hematopoietic stem cell transplantation with post-transplant cyclophosphamide
.
Bone Marrow Transplant
.
2021
. ;
56
(
10
):
2432
-
2444
.
54.
Mulroney
CM
,
Abid
MB
,
Bashey
A
, et al
.
Incidence and impact of community respiratory viral infections in post-transplant cyclophosphamide-based graft-versus-host disease prophylaxis and haploidentical stem cell transplantation
.
Br J Haematol
.
2021
. ;
194
(
1
):
145
-
157
.
55.
Imus
PH
,
Tsai
HL
,
DeZern
AE
, et al
.
thrombotic microangiopathy after post-transplantation cyclophosphamide-based graft-versus-host disease prophylaxis
.
Biol Blood Marrow Transplant
.
2020
. ;
26
(
12
):
2306
-
2310
.
56.
Pulsipher
MA
,
Chitphakdithai
P
,
Logan
BR
, et al
.
Lower risk for serious adverse events and no increased risk for cancer after PBSC vs BM donation
.
Blood
.
2014
. ;
123
(
23
):
3655
-
3663
.
57.
Jacobsohn
DA
,
Loken
MR
,
Fei
M
, et al
.
Outcomes of measurable residual disease in pediatric acute myeloid leukemia before and after hematopoietic stem cell transplant: validation of difference from normal flow cytometry with chimerism studies and wilms tumor 1 gene expression
.
Biol Blood Marrow Transplant
.
2018
. ;
24
(
10
):
2040
-
2046
.
58.
Bader
P
,
Kreyenberg
H
,
Henze
GH
, et al
.
Prognostic value of minimal residual disease quantification before allogeneic stem-cell transplantation in relapsed childhood acute lymphoblastic leukemia: the ALL-REZ BFM Study Group
.
J Clin Oncol
.
2009
. ;
27
(
3
):
377
-
384
.
59.
Chen
YB
,
Wang
T
,
Hemmer
MT
, et al
.
GvHD after umbilical cord blood transplantation for acute leukemia: an analysis of risk factors and effect on outcomes
.
Bone Marrow Transplant
.
2017
. ;
52
(
3
):
400
-
408
.
60.
Leahy
AB
,
Li
Y
,
Talano
JA
, et al
.
Unrelated donor α/β T cell- and B cell-depleted HSCT for the treatment of pediatric acute leukemia
.
Blood Adv
.
2022
. ;
6
(
4
):
1175
-
1185
.
61.
Martelli
MF
,
Reisner
Y
.
Haploidentical 'megadose' CD34+ cell transplants for patients with acute leukemia
.
Leukemia
.
2002
. ;
16
(
3
):
404
-
405
.

Author notes

Deidentified individual participant data that underlie the reported results will be made available 3 months after publication for a period of 5 years after the publication date on the Pediatric Transplantation and Cellular Therapy Consortium website (theptctc.org).

The study protocol is available at https://www.theptctc.org/pastprotocols.