• Children with AL given haplo-HSCT after αβ T- and B-cell depletion are exposed to a low risk of acute and chronic GVHD and NRM.

  • The leukemia-free, GVHD-free survival of patients given this type of allograft is comparable to that of HLA-matched donor HSCT recipients.

Allogeneic hematopoietic stem cell transplantation (HSCT) from an HLA-haploidentical relative (haplo-HSCT) is a suitable option for children with acute leukemia (AL) either relapsed or at high-risk of treatment failure. We developed a novel method of graft manipulation based on negative depletion of αβ T and B cells and conducted a prospective trial evaluating the outcome of children with AL transplanted with this approach. Eighty AL children, transplanted between September 2011 and September 2014, were enrolled in the trial. All children were given a fully myeloablative preparative regimen. Anti–T-lymphocyte globulin from day −5 to −3 was used for preventing graft rejection and graft-versus-host disease (GVHD); no patient received any posttransplantation GVHD prophylaxis. Two children experienced primary graft failure. The cumulative incidence of skin-only, grade 1-2 acute GVHD was 30%; no patient developed extensive chronic GVHD. Four patients died, the cumulative incidence of nonrelapse mortality being 5%, whereas 19 relapsed, resulting in a 24% cumulative incidence of relapse. With a median follow-up of 46 months for surviving patients, the 5-year probability of chronic GVHD-free, relapse-free survival (GRFS) is 71%. Total body irradiation–containing preparative regimen was the only variable favorably influencing relapse incidence and GRFS. The outcomes of these 80 patients are comparable to those of 41 and 51 children given transplantation from an HLA-identical sibling or a 10/10 allelic-matched unrelated donor in the same period. These data indicate that haplo-HSCT after αβ T- and B-cell depletion represents a competitive alternative for children with AL in need of urgent allograft. This trial was registered at www.clinicaltrials.gov as #NCT01810120.

Allogeneic hematopoietic stem cell transplantation (HSCT) from an HLA-haploidentical relative (haplo-HSCT) offers the option of immediate transplantation virtually to any patient in need of an allograft and lacking a suitable HLA-matched donor.1-3  In order to remove T cells, responsible for graft-versus-host disease (GVHD), and B cells, from which posttransplant lymphoproliferative disease (PTLD) can arise,4  positive selection of CD34+ hematopoietic stem cells (HSCs) has been employed for many years in haplo-HSCT.1-3  Although the administration of CD34+ cell “megadoses” demonstrated to be a suitable approach for preventing both graft failure and severe GVHD in haplo-HSCT recipients,5  removal of lymphoid cells and committed hematopoietic progenitors from the graft entailed prolonged lymphopenia and delayed immune reconstitution, resulting in an increased risk of nonrelapse mortality (NRM), mainly from opportunistic infections.2,3,6 

A promising approach to circumvent this delay in immune recovery is represented by a more sophisticated method of graft manipulation that we and other groups recently developed, based on selective depletion of αβ T lymphocytes, and of B cells.7-9  Through this approach, it is possible to transfer to the recipient not only donor HSCs but also committed hematopoietic progenitors, as well as mature natural killer (NK) and γδ T cells, both these lymphocyte subsets being capable of exerting a protective effect against leukemia cell regrowth and life-threatening infections.10  Initial clinical results in children with nonmalignant disorders who received haplo-HSCT using this new method of selective T-cell depletion11  and in small series of pediatric patients with malignancies, some of which were given additional post-transplantation immune suppression,12,13  were promising, as the reported incidence of GVHD was low, whereas immune reconstitution was improved.

Herein, we report the long-term outcome of a cohort of 80 children with acute leukemia (AL) recruited into a prospective, single-center phase 2 trial, aimed at testing the safety and efficacy of αβ T- and B-cell–depleted haplo-HSCT. Moreover, to better define the role of this approach, we compared the outcomes of these children with those of 41 and 51 patients transplanted in our center in the same period from either an HLA-identical sibling or a 10/10 allelic-matched unrelated donor (UD), respectively.

Patients

Children (aged <21 years) with acute lymphoblastic or myeloid leukemia (ALL and AML, respectively) who received a first allograft between September 2011 and September 2014 were enrolled in this prospective clinical trial, approved by the local Ethical Committee and registered at ClinicalTrial.gov Web site (#NCT01810120). Written informed consent was obtained from either patients or their legal guardians in accordance with the Helsinki Declaration.

We offered αβ T- and B-cell–depleted haplo-HSCT to all children with AL in morphological complete remission (CR) with an indication to receive an allograft and who lacked either an HLA-identical sibling or a fully allelic-matched (at the HLA A, B, C, DRB1, and DQB1 loci) UD or who needed an urgent procedure (ie, within 2 months from recognition of the indication) according to physician’s judgment. Details on patient and donor characteristics are reported in Table 1. All patients received a pretransplant, myeloablative conditioning regimen; total body irradiation (TBI) was employed in patients older than 3 years affected by either ALL or very high-risk AML (ie, those with cytogenetic/molecular features predicting high risk of relapse; see Table 1 for further details). Anti–T-lymphocyte globulin (Grafalon; Neovii Biotech) was administered at a dose of 12 mg/kg over 3 days (ie, from day −5 to −3) for prevention of both graft rejection and GVHD through in vivo depletion/modulation of bidirectional alloreactivity. Moreover, to reduce as much as possible the risk of Epstein-Barr virus (EBV) –related PTLD, on day −1, patients were also given rituximab (200 mg/m2) for in vivo depletion of both donor and recipient B cells. No patient received posttransplantation pharmacological GVHD prophylaxis.

Table 1.

Patient, donor, and transplantation characteristics

Patients(n = 80)
Sex  
 Male 55 (69%) 
 Female 25 (31%) 
Median (range) age at diagnosis, y 6.6 (0.4-16.8) 
Median (range) age at transplantation, y 9.7 (0.9-20.9) 
Disease  
 ALL 56 (70%) 
 AML 24 (30%) 
ALL phenotype  
 BCP 41 (73%) 
 T 15 (27%) 
ALL recurrent molecular lesions  
 t(4;11)(AF4/MLL) 
 t(9;22)(BCR/ABL) 
 SIL-TAL 
 t(12;21)(TEL/AML1) 
 Hypodiploid 
AML recurrent molecular/cytogenetic lesions  
 MLL/FLT3-ITD 
 7- 
 Complex karyotype 
 inv(16) (MYH11-CBFB) 
 Other 
Disease status at transplantation  
 ALL  
  CR1* 15 (19%) 
  CR2 37 (46%) 
  ≥CR3 4 (5%) 
AML  
  CR1 16 (20%) 
  CR2 8 (10%) 
CMV serology (donor/recipient)  
 Neg/Neg 5 (6%) 
 Neg/Pos 7 (9%) 
 Pos/Neg 11 (14%) 
 Pos/Pos 57 (71%) 
Conditioning regimens§  
 TBI+TT+Flu 40 (50%) 
 TBI+TT+L-PAM 20 (25%) 
 TT+Bu+Flu 13 (16%) 
 Bu+Cy+L-PAM 7 (9%) 
Donor characteristics  
 Age, y 41.5 (27-55) 
 Type of donor  
  Mother 46 (58%) 
  Father 34 (42%) 
 Sex mismatch 49 (61%) 
  Female donor → Male recipient 35/49 (71%) 
 NK alloreactivity (KIR/KIR-L model) yes/no 36 (45%)/44 (55%) 
 KIR genotype A/A vs B/X 16 (20%)/64 (80%) 
 Donor B content value 0-1 vs ≥2 44 (55%)/36 (45%) 
 Donor KIR2DS1 “educated and useful” yes/no 28 (35%)/52 (65%) 
Cell dose infused, median (range)  
 CD34+ cells × 106/kg 13.93 (6-40.44) 
 αβ+ T cells × 106/kg 0.047 (0.002-0.099) 
 γδ+ T cells × 106/kg 8.1 (0.86-56.7) 
 NK cells × 106/kg 34.6 (3.84-146.1) 
 CD20+ B cells × 106/kg 0.09 (0.05-0.48) 
Patients(n = 80)
Sex  
 Male 55 (69%) 
 Female 25 (31%) 
Median (range) age at diagnosis, y 6.6 (0.4-16.8) 
Median (range) age at transplantation, y 9.7 (0.9-20.9) 
Disease  
 ALL 56 (70%) 
 AML 24 (30%) 
ALL phenotype  
 BCP 41 (73%) 
 T 15 (27%) 
ALL recurrent molecular lesions  
 t(4;11)(AF4/MLL) 
 t(9;22)(BCR/ABL) 
 SIL-TAL 
 t(12;21)(TEL/AML1) 
 Hypodiploid 
AML recurrent molecular/cytogenetic lesions  
 MLL/FLT3-ITD 
 7- 
 Complex karyotype 
 inv(16) (MYH11-CBFB) 
 Other 
Disease status at transplantation  
 ALL  
  CR1* 15 (19%) 
  CR2 37 (46%) 
  ≥CR3 4 (5%) 
AML  
  CR1 16 (20%) 
  CR2 8 (10%) 
CMV serology (donor/recipient)  
 Neg/Neg 5 (6%) 
 Neg/Pos 7 (9%) 
 Pos/Neg 11 (14%) 
 Pos/Pos 57 (71%) 
Conditioning regimens§  
 TBI+TT+Flu 40 (50%) 
 TBI+TT+L-PAM 20 (25%) 
 TT+Bu+Flu 13 (16%) 
 Bu+Cy+L-PAM 7 (9%) 
Donor characteristics  
 Age, y 41.5 (27-55) 
 Type of donor  
  Mother 46 (58%) 
  Father 34 (42%) 
 Sex mismatch 49 (61%) 
  Female donor → Male recipient 35/49 (71%) 
 NK alloreactivity (KIR/KIR-L model) yes/no 36 (45%)/44 (55%) 
 KIR genotype A/A vs B/X 16 (20%)/64 (80%) 
 Donor B content value 0-1 vs ≥2 44 (55%)/36 (45%) 
 Donor KIR2DS1 “educated and useful” yes/no 28 (35%)/52 (65%) 
Cell dose infused, median (range)  
 CD34+ cells × 106/kg 13.93 (6-40.44) 
 αβ+ T cells × 106/kg 0.047 (0.002-0.099) 
 γδ+ T cells × 106/kg 8.1 (0.86-56.7) 
 NK cells × 106/kg 34.6 (3.84-146.1) 
 CD20+ B cells × 106/kg 0.09 (0.05-0.48) 

BCP, B-cell precursors; BU, busulfan; CMV, cytomegalovirus; CY, cyclophosphamide; Flu, fludarabine; L-PAM, melphalan; Neg, negative; Pos, positive; TT, thiotepa.

*

Of the 15 patients with ALL transplanted in CR1, 9 had high level of minimal residual disease at the end of induction therapy (ie, >1 × 10−3 at day +78 after beginning of treatment), 2 had high-risk infant ALL, 1 had t(4;11), and 3 had hyperleukocytosis T ALL with poor response to the steroid prephase.

Of the 16 patients with AML transplanted in CR1, 3 had t(10;11), 2 a complex karyotype, 3 had FLT3-ITD with high allelic ratio, 2 had M7 AML, and 6 were not in morphological CR after the first of the 2 induction courses.

Of the 37 patients with ALL transplanted in CR2, 21 (57%) and 16 (43%) patients belonged to the S2 and S3/S4 Berlin-Frankfurt-Münster classification of first relapse ALL, respectively.59 

§

TBI (12 Gy over 3 d in 6 fractions of 200 cGy each) was employed in 50 children with ALL and in 10 children with AML; all these patients were older than 3 y.

HLA-C C1pos donor and HLA-C C2pos patient.

Chimerism analysis was performed biweekly for the first 3 months and monthly thereafter. Immune recovery (count of T-cell receptor [TCR] αβ CD3+, TCR γδ CD3+, CD4+, CD8+, NK, and CD19+ cells) was evaluated at 1, 3, 6, and 12 months after transplantation. Immunoglobulin G (IgG) replacement therapy was continued until achievement of sustained serum levels comparable to those normal for patient’s age.

The donor was mainly chosen according to immunological criteria, giving priority to NK alloreactivity, evaluated according to the killer immunoglobulin-like receptor (KIR)/KIR-Ligand model, KIR B haplotype, higher B-content score, and size of NK alloreactive subset.14-19  A detailed description of the methods used for NK-cell genotype/phenotype characterization, of the criteria used for selecting the more appropriate donor, and of the KIR genotype asset of the 80 donors is reported in supplemental Figure 1 and supplemental Table 1, available on the Blood Web site.

The donor was the mother in 46 patients (57%) and the father in the remaining 34 patients (43%). Since June 2012, all recipients were tested for the presence of donor-specific antibodies,20  and 2 out of 65 resulted to be positive.

Donor mobilization and graft manipulation procedures have been previously reported.8,9  Briefly, donors received granulocyte-colony stimulating factor for 4 days at 12 µg/kg body weight in 2 divided doses to induce peripheral mobilization of CD34+ hematopoietic progenitors. Apheresis was performed on day 5 after start of mobilization. When on day 4 the CD34+ cell count was <40/µL and/or the predicted apheresis yields was ≤12.0 × 106 CD34+ HSC/kg recipient’s body weight, according to a previously reported formula,8,9  Plerixafor (Mozobil) was given at 0.24 mg/kg with the aim of boosting mobilization of hematopoietic stem/progenitor cells. Plerixafor was usually given at midnight, 9 hours prior to collection on day 5. Large-volume apheresis was performed with the Spectra Optia Cell Separator (Terumo BCT, Leuven, Belgium). Manipulations were performed in a closed system. Clinical grade reagents, disposable kits, and instrumentation were from Miltenyi Biotec (Bergisch Gladbach, Germany). Procedures were performed with the fully automated CliniMACS device in a laminar-flow hood, located in a clean room certified for sterile manipulations.

Definitions and statistical analysis

Graft failure was defined as either lack of initial engraftment of donor cells (primary graft failure) or loss of donor cells after initial engraftment (secondary graft failure). Time to neutrophil engraftment was defined as time from haplo-HSCT to the first of 3 consecutive days with an absolute neutrophil count ≥0.5 × 109/L, whereas time to platelet engraftment was defined as time from transplantation to the first of 7 consecutive days with an unsupported platelet count ≥20 × 109/L.

Patients surviving >14 and 100 days after transplantation were evaluated for acute and chronic GVHD, respectively, which were diagnosed and graded according to previously published criteria.21,22 

Overall survival (OS) was defined as the probability of survival, regardless of disease status, from the time of haplo-HSCT to time of death or of last follow-up (surviving patients were censored at last follow-up, whereas only death was considered an event). NRM was defined as the probability of death from any cause other than malignancy recurrence. Leukemia-free survival (LFS) was defined as the probability of survival, without evidence of disease at any time after transplantation. In estimating LFS, death and relapse were considered events, whereas patients who were alive with sustained donor engraftment and disease free were censored at last follow-up. We also evaluated event-free survival (EFS), considering graft failure as an additional event to those considered for estimating LFS and the composite endpoint of chronic GVHD-free and relapse-free survival (GRFS).23 

Quantitative variables were reported as median value and range, whereas categorical variables were expressed as absolute value and percentage. Demographic and clinical characteristics of patients were compared using the χ2 test or Fisher’s exact test for categorical variables, whereas the Mann-Whitney rank sum test or the Student t test was used for continuous variables, as appropriate.

Rejection, engraftment, acute and chronic GVHD, OS, LFS, NRM, and relapse incidence were estimated from the date of transplantation to the date of an event or last follow-up.

Probabilities of OS, LFS, and EFS were calculated according to the Kaplan and Meier method.24  Engraftment, acute GVHD and chronic GVHD, NRM, and relapse were calculated as cumulative incidence curves in order to adjust the estimates for competing risks.25  All results were expressed as probability or cumulative incidence (%) and 95% confidence interval (95% CI).26,27 

The significance of differences between survival probabilities was estimated by the log-rank test (Mantel-Cox), whereas Gray’s test was used to assess, in univariable analyses, differences between cumulative incidences.28  Multivariable analysis was performed using the Cox proportional hazard regression model.29  Several patient-, donor-, and transplantation-related factors, detailed in Table 2, were evaluated for their impact on relapse incidence and LFS.

Table 2.

Cumulative incidence of relapse and LFS: univariable analysis

Cumulative incidence of relapseLFS
OutcomeNo. of patientsEventsProbability (%)95% CIP valueEventsProbability (%)95% CIP value
Recipient sex          
 Male 55 12 22.8 14-38 .60 12 77.2 66-89 .031 
 Female 25 28.6 15-53  11 56.0 37-75  
Recipient age at haplo-HSCT          
 <9.7 y 41 13 31.7 20-50 .057 16 61.0 46-76 .023 
 >9.7 y 39 16.0 8-33  81.5 69-94  
Donor sex          
 Male 34 14.7 7-33 .13 73.5 59-88 .88 
 Female 46 14 31.2 20-48  14 68.7 55-82  
Donor age, y          
 <41.5 40 12 31.5 19-51 .17 12 68.5 53-84 .86 
 >41.5 40 17.5 9-34  11 72.5 59-86  
Donor-recipient sex combination          
 Female donor–male recipient 49 12 25.5 16-42 .90 16 66.4 53-80 .34 
 Other combinations 31 22.6 12-43  77.4 63-92  
Type of leukemia          
 ALL 56 13 23.2 14-37 .87 16 71.4 60-83 .98 
 AML 24 28.3 14-58  67.5 47-88  
Disease phase          
 ALL CR1 15 13.3 4-48 .61 80.0 60-100 .62 
 ALL CR2 37 24.3 14-43  10 73.0 59-87  
 AML CR1 16 36.5 17-77  53.7 29-85  
 AML CR2 12.5 2-78  87.5 65-100  
Berlin-Frankfurt-Münster classification of CR2 ALL patients          
 S2 21 14.3 3-33 .08 81.0 57-92 .18 
 S3-S4 16 37.5 15-61  62.5 35-81  
Donor-recipient CMV serology          
 Neg/Neg 40.0 14-100 .43 60.0 17-100 .75 
 Pos/Neg 42.9 18-100  57.1 20-94  
 Neg/Pos 11 18.2 5-64  72.7 46-99  
 Pos/Pos 57 12 21.9 13-36  15 72.8 61-85  
Conditioning regimen          
 TBI-based 60 10 16.7 9-29 .014 11 81.7 72-91 .0002 
 Chemo-based 20 47.5 29-77  12 37.5 15-60  
Graft composition          
 CD34+ cells × 106/kg
 ≥m.v.
 <m.v. 
         
41 15.1 7-32 .045 82.4 71-94 .016 
39 13 33.3 21-52  16 59.0 44-74  
 αβ+ T cells × 106/kg          
  ≥m.v.
<m.v. 
41 12 29.3 18-47 .23 15 63.4 49-78 .11 
39 18.9 10-37  78.5 65-92  
 γδ+ T cells × 106/kg
 ≥m.v.
 <m.v. 
         
40 11 28.2 17-47 .42 12 69.2 55-84 .82 
40 21.1 11-39  11 71.4 57-86  
 NK cells × 106/kg
 ≥m.v.
 <m.v. 
         
40 23.8 13-42 .73 12 68.5 54-83 .88 
40 10 25.0 15-43  11 72.5 59-86  
NK cell alloreactivity          
 Yes 36 22.2 12-41 .84 75.0 61-89 .53 
 No 44 11 26.4 16-44  14 66.7 52-81  
KIR genotype          
 B/X 64 18 28.7 19-43 .11 22 65.0 53-77 .045 
 A/A 16 6.7 1-44  93.3 81-100  
B-content score          
 ≥2 36 25.0 14-44 .89 10 72.2 58-87 .72 
 0-1 44 10 23.9 14-41  13 69.1 55-83  
Donor KIR2DS1 “educated and useful”          
 Yes 28 28.6 13-46 .48 10 64.3 44-79 .32 
 No 52 11 22.2 12-35  13 74.0 59-84  
Grade 1-2 acute GVHD          
 Yes 24 12.5 4-36 .12 18 67.2 55-80 .34 
 No 56 16 29.2 19-44  79.2 63-95  
Chronic GVHD          
 Yes 25.0 5-100 .94 75.0 33-100 1.00 
 No 69 15 22.0 14-35  16 76.6 66-87  
Cumulative incidence of relapseLFS
OutcomeNo. of patientsEventsProbability (%)95% CIP valueEventsProbability (%)95% CIP value
Recipient sex          
 Male 55 12 22.8 14-38 .60 12 77.2 66-89 .031 
 Female 25 28.6 15-53  11 56.0 37-75  
Recipient age at haplo-HSCT          
 <9.7 y 41 13 31.7 20-50 .057 16 61.0 46-76 .023 
 >9.7 y 39 16.0 8-33  81.5 69-94  
Donor sex          
 Male 34 14.7 7-33 .13 73.5 59-88 .88 
 Female 46 14 31.2 20-48  14 68.7 55-82  
Donor age, y          
 <41.5 40 12 31.5 19-51 .17 12 68.5 53-84 .86 
 >41.5 40 17.5 9-34  11 72.5 59-86  
Donor-recipient sex combination          
 Female donor–male recipient 49 12 25.5 16-42 .90 16 66.4 53-80 .34 
 Other combinations 31 22.6 12-43  77.4 63-92  
Type of leukemia          
 ALL 56 13 23.2 14-37 .87 16 71.4 60-83 .98 
 AML 24 28.3 14-58  67.5 47-88  
Disease phase          
 ALL CR1 15 13.3 4-48 .61 80.0 60-100 .62 
 ALL CR2 37 24.3 14-43  10 73.0 59-87  
 AML CR1 16 36.5 17-77  53.7 29-85  
 AML CR2 12.5 2-78  87.5 65-100  
Berlin-Frankfurt-Münster classification of CR2 ALL patients          
 S2 21 14.3 3-33 .08 81.0 57-92 .18 
 S3-S4 16 37.5 15-61  62.5 35-81  
Donor-recipient CMV serology          
 Neg/Neg 40.0 14-100 .43 60.0 17-100 .75 
 Pos/Neg 42.9 18-100  57.1 20-94  
 Neg/Pos 11 18.2 5-64  72.7 46-99  
 Pos/Pos 57 12 21.9 13-36  15 72.8 61-85  
Conditioning regimen          
 TBI-based 60 10 16.7 9-29 .014 11 81.7 72-91 .0002 
 Chemo-based 20 47.5 29-77  12 37.5 15-60  
Graft composition          
 CD34+ cells × 106/kg
 ≥m.v.
 <m.v. 
         
41 15.1 7-32 .045 82.4 71-94 .016 
39 13 33.3 21-52  16 59.0 44-74  
 αβ+ T cells × 106/kg          
  ≥m.v.
<m.v. 
41 12 29.3 18-47 .23 15 63.4 49-78 .11 
39 18.9 10-37  78.5 65-92  
 γδ+ T cells × 106/kg
 ≥m.v.
 <m.v. 
         
40 11 28.2 17-47 .42 12 69.2 55-84 .82 
40 21.1 11-39  11 71.4 57-86  
 NK cells × 106/kg
 ≥m.v.
 <m.v. 
         
40 23.8 13-42 .73 12 68.5 54-83 .88 
40 10 25.0 15-43  11 72.5 59-86  
NK cell alloreactivity          
 Yes 36 22.2 12-41 .84 75.0 61-89 .53 
 No 44 11 26.4 16-44  14 66.7 52-81  
KIR genotype          
 B/X 64 18 28.7 19-43 .11 22 65.0 53-77 .045 
 A/A 16 6.7 1-44  93.3 81-100  
B-content score          
 ≥2 36 25.0 14-44 .89 10 72.2 58-87 .72 
 0-1 44 10 23.9 14-41  13 69.1 55-83  
Donor KIR2DS1 “educated and useful”          
 Yes 28 28.6 13-46 .48 10 64.3 44-79 .32 
 No 52 11 22.2 12-35  13 74.0 59-84  
Grade 1-2 acute GVHD          
 Yes 24 12.5 4-36 .12 18 67.2 55-80 .34 
 No 56 16 29.2 19-44  79.2 63-95  
Chronic GVHD          
 Yes 25.0 5-100 .94 75.0 33-100 1.00 
 No 69 15 22.0 14-35  16 76.6 66-87  

Values of P < .05 are significant (bold).

m.v., Median value.

With the aim of better defining the role of haplo-HSCT, we compared the outcomes of children enrolled in this trial with those of patients with AL transplanted from either an HLA-identical sibling or a 10/10 allelic matched UD in the same period in our center. Details on clinical characteristics of patients belonging to the 3 groups and the correlated comparative analysis are reported in supplemental Table 2.

Statistical analysis was performed using NCSS (NCSS 10 Statistical Software [2015]; NCSS, LLC, Kaysville, UT, ncss.com/software/ncss) and R 2.5.0 software package (http://www.R-project.org). Data were analyzed as of 1 January 2017.

All children received at least 6 × 106 CD34+ cells/kg body weight and an αβ T-cell number lower than 1 × 105/kg body weight; details on the number of HSC and lymphocyte subsets infused are shown in Table 1. Sixteen donors (20%) were given plerixafor for optimizing mobilization of HSC.

Engraftment

Two patients did not engraft; 1 patient was successfully rescued through haplo-HSCT from the other parent, whereas the other died of disseminated adenovirus infection, despite receiving a second allograft from the same donor with engraftment and hematopoietic recovery. This patient was 1 of the 2 with donor-specific alloantibodies. The number of CD34+ cells/kg body weight infused in these 2 patients was 8.9 and 24.3 × 106, respectively. The median time to neutrophil engraftment in the whole study population was 13 days (range, 9-19); no variable influenced the kinetics of recovery. The median time to platelet engraftment was 11 days (range, 8-20). It was 10 days (range, 8-16) and 12 days (range, 10-20) in patients given a number of CD34+ cells either above or below the median value infused (P = .03).

Acute and chronic GVHD

Twenty-four patients (30%) developed grade 1-2, skin-only acute GVHD, whereas 56 children (70%) did not present any grade of acute GVHD. No single case of acute GVHD with visceral involvement or of grade 3-4 acute GVHD was recorded (supplemental Figure 2A). The overall 100-day cumulative incidence of grade 1-2 acute GVHD was 30% (95% CI, 21-41). No variable predicted the occurrence of acute GVHD. All patients with grade 1-2, skin-only, acute GVHD responded to treatment with either topical or systemic steroids.

Seventy-three patients surviving >100 days after haplo-HSCT were evaluated for chronic GVHD occurrence. Clinically limited, skin-only chronic GVHD was diagnosed in 4 patients (5%). In these children, acute GVHD preceded chronic GVHD. The overall cumulative incidence of limited chronic GVHD was 5% (95% CI, 2-15; supplemental Figure 2B).

NRM

Four patients died of transplantation-related causes: 2 because of idiopathic pneumonitis and 1 each of disseminated adenovirus infection after graft failure and cardiac insufficiency. Only 1 of the 4 patients who died because of transplant-related causes had received TBI as part of the preparative regimen, whereas the remaining 3 had been prepared with a chemotherapy-based regimen. These fatal events occurred in the first 100 days after the allograft. The 5-year cumulative incidence of NRM for the whole cohort of patients is 5% (95% CI 2-13; Figure 1A).

Figure 1.

Outcomes of TCRαβ/CD19-depleted haplo-HSCT. (A) NRM of the whole cohort of patients enrolled in the trial. (B) Cumulative incidence of relapse of the whole cohort of patients enrolled in the trial. (C) LFS of the whole cohort of patients enrolled in the trial. (D) LFS according to the disease type (ALL [continuous green line] vs AML [dotted red line]). E, events; N, number.

Figure 1.

Outcomes of TCRαβ/CD19-depleted haplo-HSCT. (A) NRM of the whole cohort of patients enrolled in the trial. (B) Cumulative incidence of relapse of the whole cohort of patients enrolled in the trial. (C) LFS of the whole cohort of patients enrolled in the trial. (D) LFS according to the disease type (ALL [continuous green line] vs AML [dotted red line]). E, events; N, number.

Close modal

Relapse

With a median follow-up of 46 months (range, 26-60), 19 patients (24%) relapsed at a median of 6.3 months (range 2-22), the CI of relapse being 24% (95% CI, 16-36; Figure 1B). Fifteen of these 19 relapses occurred in first year after transplantation. The CI of relapse was 23% (95% CI, 14-37) for the 56 children with ALL and 28% (95% CI, 14-58) for the 24 children with AML (P = not significant). In univariable analysis (Table 2), CD34+ cell number above the median value and a TBI-containing regimen were associated with lower risk of leukemia recurrence; use of TBI remained significant in multivariable analysis (hazard ratio 0.36; 95% CI 0.14-0.91; P = .03).

Survival and LFS

Fifty-eight patients (72% of the whole population) were alive at time of the last follow-up: 57 were in continuous CR after haplo-HSCT, whereas 1 was alive after posttransplantation leukemia relapse.

The 5-year OS probability was 72% (95% CI, 62-82) for the whole study population. Overall, the 5-year LFS was 71% (95% CI, 61-81; Figure 1C); it was 71% (95% CI, 60-83) and 68% (95% CI, 47-88) in patients transplanted for ALL and AML, respectively (P = not significant; Figure 1D). The 5-year probability of EFS was 69.5% (95% CI 58-78.4), whereas the 5-year probability of GRFS was 71% (95% CI, 61-81; Figure 2D).

Figure 2.

Outcomes of TCRαβ/CD19-depleted haplo-HSCT and comparison with transplants from HLA-identical sibling and MUD. (A) LFS probability according to the use of TBI in the conditioning regimen (yes, continuous blue line; no, dotted red line). (B) LFS probability according to NK alloreactivity (evaluated using the KIR-KIR-L model of prediction) (yes, dotted red line; no, continuous blue line). (C) Comparison of LFS probability among patients transplanted from different donors (HLA-haploidentical relative [dashed green line], HLA-identical sibling [continuous blue line], and UD [dotted red line]) during the same period. (D) Comparison of GRFS probability among patients transplanted from different donors (HLA-haploidentical relative [dashed green line], HLA-identical sibling [continuous blue line], and UD [dotted red line]) during the same period. MUD, matched unrelated donor; SIB, HLA-identical sibling.

Figure 2.

Outcomes of TCRαβ/CD19-depleted haplo-HSCT and comparison with transplants from HLA-identical sibling and MUD. (A) LFS probability according to the use of TBI in the conditioning regimen (yes, continuous blue line; no, dotted red line). (B) LFS probability according to NK alloreactivity (evaluated using the KIR-KIR-L model of prediction) (yes, dotted red line; no, continuous blue line). (C) Comparison of LFS probability among patients transplanted from different donors (HLA-haploidentical relative [dashed green line], HLA-identical sibling [continuous blue line], and UD [dotted red line]) during the same period. (D) Comparison of GRFS probability among patients transplanted from different donors (HLA-haploidentical relative [dashed green line], HLA-identical sibling [continuous blue line], and UD [dotted red line]) during the same period. MUD, matched unrelated donor; SIB, HLA-identical sibling.

Close modal

Table 2 details the univariable analysis of factors potentially influencing the LFS probability. Only a TBI-containing regimen remained significant in multivariable analysis (hazard ratio 0.30; 95% CI 0.12-0.72; P = .007; see also Figure 2A). Noteworthy, in this study, we were unable to confirm any protective advantage for LFS in patients transplanted from an NK alloreactive donor (evaluated using the KIR-KIR-L model of prediction; Figure 2B).

Comparison of outcomes of haplo-HSCT recipients with those of children given HLA-identical sibling or UD transplantation

In order to compare differences between haplo-HSCT and transplantation from either an HLA-identical sibling or a 10/10 allelic-matched UD, we also analyzed 41 and 51 consecutive AL children of the latter 2 cohorts given an allograft in CR in the same period (see supplemental Table 2). The 3 groups were comparable for many relevant features considered, with the exception of source of stem cells employed (98% and 78% of HLA-identical siblings or UD HSCT recipients were transplanted with bone marrow cells), GVHD prophylaxis (none of the patients of the 2 control groups was given a T-cell–depleted allograft), and human cytomegalovirus (HCMV) serology (see supplemental Table 2 for details).

Haplo-HSCT had a lower incidence of grade 3-4 acute GVHD, of visceral GVHD, and of chronic GVHD (P < .01, <.01, and .03, respectively; supplemental Figure 2A-B). There was no significant difference in the risk of any cause-specific death (GVHD, infection, regimen-related toxicity, and leukemia relapse) among these 3 groups (data not shown). The 5-year probability of LFS and GRFS did not differ between haplo-HSCT recipients and the other 2 cohorts of patients (Figure 2C-D).

Immune recovery of haplo-HSCT recipients

Recovery of lymphoid subsets and of immunoglobulin serum levels is detailed in Table 3, showing prompt recovery of γδ T lymphocytes and NK cells in the early posttransplant period and progressive emergence over time of αβ T lymphocytes. Noteworthy, notwithstanding a dose of rituximab administered before transplantation and depletion of donor B cells from the graft, all children had B-cell engraftment.

Table 3.

Details on immune reconstitution of the 80 patients given haplo-HSCT, median (range)

1 mo3 mo6 mo12 mo
CD3+ T cells/μL 231 (1-1618) 254 (23-1472) 668 (153-2596) 1379 (407-3449) 
CD4+ T cells/μL 19 (0-442) 89 (4-397) 264 (87-1055) 599 (174-1421) 
CD8+ T cells/μL 24 (0-910) 96 (9-1108) 301 (25-1581) 574 (112-2301) 
αβ T cells/μL 47 (1-672) 186 (12-1340) 573 (135-2146) 1291 (259-2795) 
γδ T cells/μL 181 (1-1335) 49 (4-388) 84 (4-752) 94 (10-660) 
CD3-CD56+ NK cells/μL 236 (47-1813) 196 (29-1448) 283 (48-5441) 269 (79-3116) 
CD19+ B cells/μL 0 (0-20) 2 (0-473) 160 (2-1609) 291 (40-1616) 
IgG, mg/dL 973 (104-3153) 580 (340-1041) 597 (213-1487) 711 (377-1351) 
IgA, mg/dL 32 (0-190) 26 (0-186) 33 (0-284) 41 (0-221) 
IgM, mg/dL 10 (0-66) 10 (0-44) 47 (0-496) 64 (0-469) 
1 mo3 mo6 mo12 mo
CD3+ T cells/μL 231 (1-1618) 254 (23-1472) 668 (153-2596) 1379 (407-3449) 
CD4+ T cells/μL 19 (0-442) 89 (4-397) 264 (87-1055) 599 (174-1421) 
CD8+ T cells/μL 24 (0-910) 96 (9-1108) 301 (25-1581) 574 (112-2301) 
αβ T cells/μL 47 (1-672) 186 (12-1340) 573 (135-2146) 1291 (259-2795) 
γδ T cells/μL 181 (1-1335) 49 (4-388) 84 (4-752) 94 (10-660) 
CD3-CD56+ NK cells/μL 236 (47-1813) 196 (29-1448) 283 (48-5441) 269 (79-3116) 
CD19+ B cells/μL 0 (0-20) 2 (0-473) 160 (2-1609) 291 (40-1616) 
IgG, mg/dL 973 (104-3153) 580 (340-1041) 597 (213-1487) 711 (377-1351) 
IgA, mg/dL 32 (0-190) 26 (0-186) 33 (0-284) 41 (0-221) 
IgM, mg/dL 10 (0-66) 10 (0-44) 47 (0-496) 64 (0-469) 

This study is the first to report long-term outcome of a large population of children with AL given αβ T- and B-cell–depleted haplo-HSCT after a myeloablative regimen and enrolled into a prospective, registered phase 2 trial. Our data indicate that, despite not receiving any posttransplantation pharmacological prophylaxis, these patients benefited from a high engraftment rate (98%) and experienced a low incidence of both acute and chronic GVHD, which contributed to the reduced risk of NRM. Remarkably, none of our patients had either grade 3-4 or gut/liver acute GVHD and all cases of chronic GVHD were of limited severity. Moreover, we did not observe any case of EBV-related PTLD, which may occur in immune-compromised individuals in the absence of virus-specific, adaptive T-cell immunity.4  Administration of rituximab before transplantation may have helped prevent EBV-PTLD, and recipient B-cell depletion may have contributed to the low GVHD incidence and severity.30 

In the past, ex vivo T-cell–depleted haplo-HSCTs have been performed either through positive selection of CD34+ cells31,32  or through removal of CD3+ T cells in combination with CD19+ B cells.33,34  Unfortunately, both approaches result in loss of certain cell subsets that may play a positive role in the recipient. In fact, although T cells displaying the αβ TCR are responsible for GVHD, T cells carrying the γδ receptor chains have no alloreactive capacity, but contribute an important anti-infectious activity,35,36  in addition to a possible antileukemia role.37-40  The Vδ2 population recognizes nonpeptide phosphoantigens expressed by leukemia cells, whereas Vδ1 cells expand in response to HCMV reactivation,39  and their presence was associated with complete responses observed in patients with B-cell ALL after T-cell–depleted allogeneic HSCT.38  It is conceivable that the high number of γδ T cells adoptively transferred with the graft in our patients may have contributed to prevent disease recurrence and severe infections. Also donor-derived, mature NK cells, lost in the procedure of positive selection of CD34+ cells and spared in our αβ T-cell–depleted graft, exhibit a graft-versus-leukemia (GVL) effect6,10,14-16,41,42  and participate in the control of opportunistic infections, including HCMV.43-45  In previous studies, we documented that in haplo-HSCT recipients given positively selected CD34+ cells, ∼8 weeks after transplantation are needed to detect mature KIR+ NK cells, and this gap in reconstitution may favor early leukemia relapse in the case of high residual tumor burden and/or rapidly proliferating blasts.14,16  Through the approach of selective αβ T- and B-cell depletion, the recipient immediately benefits from high numbers of donor mature NK cells that can fully display their activity, because the recipient is not exposed to the effect of pharmacological prophylaxis of GVHD, which can impair differentiation/expansion of this lymphocyte subset.46  Altogether, the infusion of cells belonging to innate immunity, together with that of high numbers of committed hematopoietic progenitors and monocyte/dendritic cells (in particular, in patients whose donor was mobilized with granulocyte-colony stimulating factor and plerixafor)9  may have contributed to the low risk of NRM, which we found to be comparable to that observed after transplantation from an HLA-compatible donor, either a sibling, or a UD. We did not document any favorable influence of NK alloreactivity42,47  and of donor KIR B-haplotype15  reported in other studies mainly based on infusion of CD34+ cells, likely because the NK-mediated GVL effect was partially obscured by other cells present in the graft, including γδ T cells.48 

Previously published, nonprospective studies enrolling smaller cohorts of patients with shorter follow-up have analyzed the outcome of children given αβ T- and B-cell–depleted haplo-HSCT. Maschan et al analyzed the outcome of children with high-risk AML, who received transplantation from UD (n = 20) and haploidentical donors (n = 13) after this graft manipulation. Twenty-eight patients were given posttransplantation pharmacological immune suppression, including tacrolimus until day +30 and methotrexate in 21 patients, tacrolimus in 5, methotrexate in 2, whereas 5 patients did not receive prophylaxis.13  Notably, recipients of haploidentical grafts more commonly developed isolated skin GVHD, whereas gastrointestinal involvement was more common in UD HSCT. Cumulative incidence of relapse at 2 years in the 13 haplo-HSCT recipients was 40% (95% CI: 20-80), whereas the LFS probability was 59% (95% CI: 31-87). Lang et al recently published the retrospective analysis of immune recovery in a cohort of 41 pediatric patients, with AL, myelodysplastic syndrome, and nonmalignant diseases (n = 5), who received αβ T- and B-cell–depleted allografts from a haploidentical relative after reduced-toxicity regimens.49  Grade 3-4 acute GVHD occurred in 15% of patients; with a median follow-up of 1.6 years, 21 of the 41 patients were alive and relapse was the major cause of death (n = 17). Also in our cohort of patients, disease recurrence was the main cause of treatment failure, the CI of relapse being 24%. The lower incidence of relapse in our patients can be attributed, at least partly, to the use of fully myeloablative conditioning regimens and to the lack of posttransplantation GVHD prophylaxis, potentially able to impair the innate immunity-mediated GVL effect. Support to the former interpretation is given by the observation that a better outcome was observed when we used conditioning regimens including TBI, which, albeit more toxic in the long term for children,50-52  displays potent antileukemia activity53  potentially compensating for the lack of αβ T-cell–mediated GVL effect. In addition, we hypothesize that the accurate identification and determination of alloreactive NK cells, as well as a refined analysis of the main activating NK receptors, allowed selecting donors with high antileukemia activity, thus contributing to reduce the risk of relapse. In view of all these considerations, we cannot dissect the relative contribution of the different components of transplant package to the good outcome of our patients.

Our results document that this type of haplo-HSCT offers comparable risks of NRM and relapse with respect to transplantation from HLA-identical siblings or allelic-matched UDs. This finding is corroborated by the observation that the 71% probability of LFS at 5 years observed in our 54 children with ALL is superimposable to that recently reported by Peters et al54  in 306 children transplanted from an UD using a standardized protocol for transplantation/GVHD prophylaxis (71% at 4 years). Moreover, our results compare favorably with the 5-year LFS of 30% and 34% in the larger cohort of 22 CR1 and 48 CR2 ALL patients given haplo-HSCT with CD34+ cells reported so far.31 

In the last few years, alternative platforms, such as that based on posttransplantation infusion of cyclophosphamide, have been developed.55  Although largely used in adults, few studies have been published on the use of this approach for modulating alloreactivity in AL children.56,57  Although certainly cheaper than the αβ T- and B-cell depletion, the use of posttransplantation cyclophosphamide seems to be associated with a risk of leukemia recurrence higher than that observed in our cohort.57  Future studies will further clarify the relative advantages and limitations of these 2 different haplo-HSCT platforms.

In summary, our data indicate that, through more refined approaches of graft manipulation, haplo-HSCT offers the opportunity to transplant virtually every child in need of an allograft, with an expected outcome comparable to that obtained when the donor is an HLA-matched sibling or an allelic-matched volunteer. Because we have been able to successfully transplant adolescents and patients with a body weight >40 kg, it is reasonable to hypothesize that this approach is feasible to be translated to adults, maybe through repeated apheretic collections of the donor, employed in 7 cases in our cohort. We are now running a new prospective trial, based on post–haplo-HSCT infusion of titrated numbers of donor-derived αβ T cells transduced with a suicide gene for controlling possible alloreactive reactions,58  with the aim of accelerating recovery of adaptive immunity and, possibly, reducing the risk of leukemia recurrence.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

This work was supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC) special grant “5xmille”-9962 (F.L., A.M., and L.M.), “My first AIRC” grant 15925 (A.B.), AIRC IG-16764 (D. Pende), Ministero della Salute (RF-2010-2316606) (F.L.), Ricerca Corrente (F.L.), Regione Lazio grant FILAS (F.L.), and Ministero dell’Istruzione, Università e Ricerca grant Progetto di Rilevante Interesse Nazionale, PRIN 2010 (F.L.).

Contribution: F.L., A.B., P.M., R.H., A.M., and L.M. designed the study and the concept; M.F., D. Pende, and R. Meazza provided the donor NK cell characterization and donor selection; F.L., P.M., L.P.B., D. Pagliara, B.L., G.M.M., M.A., R.M.P., L.S., and A.B. supervised the transplantation of patients and collection of data; G.L.P. oversaw the graft manipulation; L.G. conducted the radiotherapy; V.B. and G.L.P. studied the immune reconstitution; R.R., R. Masetti, P.M., and F.L. analyzed and interpreted the data (eg, statistical analysis, computational analysis); F.L., A.B., and L.M. were in charge of the study supervision; and all authors contributed to the writing, review, and/or revision of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Franco Locatelli, Dipartimento di Oncoematologia Pediatrica, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Pediatrico Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Roma, Italy; e-mail: franco.locatelli@opbg.net.

1.
Mancusi
A
,
Ruggeri
L
,
Velardi
A
.
Haploidentical hematopoietic transplantation for the cure of leukemia: from its biology to clinical translation
.
Blood
.
2016
;
128
(
23
):
2616
-
2623
.
2.
Martelli
MF
,
Aversa
F
.
Haploidentical transplants using ex vivo T-cell depletion
.
Semin Hematol
.
2016
;
53
(
4
):
252
-
256
.
3.
Reisner
Y
,
Hagin
D
,
Martelli
MF
.
Haploidentical hematopoietic transplantation: current status and future perspectives
.
Blood
.
2011
;
118
(
23
):
6006
-
6017
.
4.
Bollard
CM
,
Rooney
CM
,
Heslop
HE
.
T-cell therapy in the treatment of post-transplant lymphoproliferative disease
.
Nat Rev Clin Oncol
.
2012
;
9
(
9
):
510
-
519
.
5.
Aversa
F
,
Terenzi
A
,
Tabilio
A
, et al
.
Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse
.
J Clin Oncol
.
2005
;
23
(
15
):
3447
-
3454
.
6.
Locatelli
F
,
Pende
D
,
Maccario
R
,
Mingari
MC
,
Moretta
A
,
Moretta
L
.
Haploidentical hemopoietic stem cell transplantation for the treatment of high-risk leukemias: how NK cells make the difference
.
Clin Immunol
.
2009
;
133
(
2
):
171
-
178
.
7.
Chaleff
S
,
Otto
M
,
Barfield
RC
, et al
.
A large-scale method for the selective depletion of alphabeta T lymphocytes from PBSC for allogeneic transplantation
.
Cytotherapy
.
2007
;
9
(
8
):
746
-
754
.
8.
Li Pira
G
,
Malaspina
D
,
Girolami
E
, et al
.
Selective depletion of αβ T cells and B cells for human leukocyte antigen-haploidentical hematopoietic stem cell transplantation. A three-year follow-up of procedure efficiency
.
Biol Blood Marrow Transplant
.
2016
;
22
(
11
):
2056
-
2064
.
9.
Rutella
S
,
Filippini
P
,
Bertaina
V
, et al
.
Mobilization of healthy donors with plerixafor affects the cellular composition of T-cell receptor (TCR)-αβ/CD19-depleted haploidentical stem cell grafts
.
J Transl Med
.
2014
;
12
:
240
.
10.
Locatelli
F
,
Merli
P
,
Rutella
S
.
At the bedside: innate immunity as an immunotherapy tool for hematological malignancies
.
J Leukoc Biol
.
2013
;
94
(
6
):
1141
-
1157
.
11.
Bertaina
A
,
Merli
P
,
Rutella
S
, et al
.
HLA-haploidentical stem cell transplantation after removal of αβ+ T and B cells in children with nonmalignant disorders
.
Blood
.
2014
;
124
(
5
):
822
-
826
.
12.
Laberko
A
,
Bogoyavlenskaya
A
,
Shelikhova
L
, et al
.
Risk factors for and the clinical impact of cytomegalovirus and Epstein-Barr virus infections in pediatric recipients of TCR-α/β- and CD19-depleted grafts
.
Biol Blood Marrow Transplant
.
2017
;
23
(
3
):
483
-
490
.
13.
Maschan
M
,
Shelikhova
L
,
Ilushina
M
, et al
.
TCR-alpha/beta and CD19 depletion and treosulfan-based conditioning regimen in unrelated and haploidentical transplantation in children with acute myeloid leukemia
.
Bone Marrow Transplant
.
2016
;
51
(
5
):
668
-
674
.
14.
Moretta
L
,
Locatelli
F
,
Pende
D
,
Marcenaro
E
,
Mingari
MC
,
Moretta
A
.
Killer Ig-like receptor-mediated control of natural killer cell alloreactivity in haploidentical hematopoietic stem cell transplantation
.
Blood
.
2011
;
117
(
3
):
764
-
771
.
15.
Oevermann
L
,
Michaelis
SU
,
Mezger
M
, et al
.
KIR B haplotype donors confer a reduced risk for relapse after haploidentical transplantation in children with ALL
.
Blood
.
2014
;
124
(
17
):
2744
-
2747
.
16.
Pende
D
,
Marcenaro
S
,
Falco
M
, et al
.
Anti-leukemia activity of alloreactive NK cells in KIR ligand-mismatched haploidentical HSCT for pediatric patients: evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity
.
Blood
.
2009
;
113
(
13
):
3119
-
3129
.
17.
Cooley
S
,
Weisdorf
DJ
,
Guethlein
LA
, et al
.
Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia
.
Blood
.
2010
;
116
(
14
):
2411
-
2419
.
18.
Venstrom
JM
,
Pittari
G
,
Gooley
TA
, et al
.
HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1
.
N Engl J Med
.
2012
;
367
(
9
):
805
-
816
.
19.
Cooley
S
,
Weisdorf
DJ
,
Guethlein
LA
, et al
.
Donor killer cell Ig-like receptor B haplotypes, recipient HLA-C1, and HLA-C mismatch enhance the clinical benefit of unrelated transplantation for acute myelogenous leukemia
.
J Immunol
.
2014
;
192
(
10
):
4592
-
4600
.
20.
Ciurea
SO
,
Thall
PF
,
Milton
DR
, et al
.
Complement-binding donor-specific anti-HLA antibodies and risk of primary graft failure in hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2015
;
21
(
8
):
1392
-
1398
.
21.
Glucksberg
H
,
Storb
R
,
Fefer
A
, et al
.
Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors
.
Transplantation
.
1974
;
18
(
4
):
295
-
304
.
22.
Lee
SJ
,
Vogelsang
G
,
Flowers
ME
.
Chronic graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2003
;
9
(
4
):
215
-
233
.
23.
Holtan
SG
,
DeFor
TE
,
Lazaryan
A
, et al
.
Composite end point of graft-versus-host disease-free, relapse-free survival after allogeneic hematopoietic cell transplantation
.
Blood
.
2015
;
125
(
8
):
1333
-
1338
.
24.
Kaplan
EL
,
Meier
P
.
Non parametric estimation from incomplete observations
.
J Am Stat Assoc
.
1958
;
53
(
282
):
457
-
481
.
25.
Pepe
MS
,
Longton
G
,
Pettinger
M
,
Mori
M
,
Fisher
LD
,
Storb
R
.
Summarizing data on survival, relapse, and chronic graft-versus-host disease after bone marrow transplantation: motivation for and description of new methods
.
Br J Haematol
.
1993
;
83
(
4
):
602
-
607
.
26.
Klein
JP
,
Rizzo
JD
,
Zhang
MJ
,
Keiding
N
.
Statistical methods for the analysis and presentation of the results of bone marrow transplants. Part 2: regression modeling
.
Bone Marrow Transplant
.
2001
;
28
(
11
):
1001
-
1011
.
27.
Klein
JP
,
Rizzo
JD
,
Zhang
MJ
,
Keiding
N
.
Statistical methods for the analysis and presentation of the results of bone marrow transplants. Part I: unadjusted analysis
.
Bone Marrow Transplant
.
2001
;
28
(
10
):
909
-
915
.
28.
Gray
RJ
.
A class of K-sample tests for comparing the cumulative incidence of a competing risk
.
Ann Stat
.
1988
;
16
(
3
):
1141
-
1154
.
29.
Scrucca
L
,
Santucci
A
,
Aversa
F
.
Regression modeling of competing risk using R: an in depth guide for clinicians
.
Bone Marrow Transplant
.
2010
;
45
(
9
):
1388
-
1395
.
30.
Christopeit
M
,
Schütte
V
,
Theurich
S
,
Weber
T
,
Grothe
W
,
Behre
G
.
Rituximab reduces the incidence of acute graft-versus-host disease
.
Blood
.
2009
;
113
(
13
):
3130
-
3131
.
31.
Klingebiel
T
,
Cornish
J
,
Labopin
M
, et al
;
Pediatric Diseases and Acute Leukemia Working Parties of the European Group for Blood and Marrow Transplantation (EBMT)
.
Results and factors influencing outcome after fully haploidentical hematopoietic stem cell transplantation in children with very high-risk acute lymphoblastic leukemia: impact of center size: an analysis on behalf of the Acute Leukemia and Pediatric Disease Working Parties of the European Blood and Marrow Transplant group
.
Blood
.
2010
;
115
(
17
):
3437
-
3446
.
32.
Marks
DI
,
Khattry
N
,
Cummins
M
, et al
.
Haploidentical stem cell transplantation for children with acute leukaemia
.
Br J Haematol
.
2006
;
134
(
2
):
196
-
201
.
33.
Bethge
WA
,
Faul
C
,
Bornhäuser
M
, et al
.
Haploidentical allogeneic hematopoietic cell transplantation in adults using CD3/CD19 depletion and reduced intensity conditioning: an update
.
Blood Cells Mol Dis
.
2008
;
40
(
1
):
13
-
19
.
34.
Lang
P
,
Teltschik
HM
,
Feuchtinger
T
, et al
.
Transplantation of CD3/CD19 depleted allografts from haploidentical family donors in paediatric leukaemia
.
Br J Haematol
.
2014
;
165
(
5
):
688
-
698
.
35.
Bonneville
M
,
O’Brien
RL
,
Born
WK
.
Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity
.
Nat Rev Immunol
.
2010
;
10
(
7
):
467
-
478
.
36.
Chiplunkar
S
,
Dhar
S
,
Wesch
D
,
Kabelitz
D
.
gammadelta T cells in cancer immunotherapy: current status and future prospects
.
Immunotherapy
.
2009
;
1
(
4
):
663
-
678
.
37.
Dolstra
H
,
Fredrix
H
,
van der Meer
A
,
de Witte
T
,
Figdor
C
,
van de Wiel-van Kemenade
E
.
TCR gamma delta cytotoxic T lymphocytes expressing the killer cell-inhibitory receptor p58.2 (CD158b) selectively lyse acute myeloid leukemia cells
.
Bone Marrow Transplant
.
2001
;
27
(
10
):
1087
-
1093
.
38.
Godder
KT
,
Henslee-Downey
PJ
,
Mehta
J
, et al
.
Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation
.
Bone Marrow Transplant
.
2007
;
39
(
12
):
751
-
757
.
39.
Airoldi
I
,
Bertaina
A
,
Prigione
I
, et al
.
γδ T-cell reconstitution after HLA-haploidentical hematopoietic transplantation depleted of TCR-αβ+/CD19+ lymphocytes
.
Blood
.
2015
;
125
(
15
):
2349
-
2358
.
40.
Scheper
W
,
van Dorp
S
,
Kersting
S
, et al
.
γδT cells elicited by CMV reactivation after allo-SCT cross-recognize CMV and leukemia
.
Leukemia
.
2013
;
27
(
6
):
1328
-
1338
.
41.
Leung
W
,
Iyengar
R
,
Turner
V
, et al
.
Determinants of antileukemia effects of allogeneic NK cells
.
J Immunol
.
2004
;
172
(
1
):
644
-
650
.
42.
Ruggeri
L
,
Capanni
M
,
Urbani
E
, et al
.
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants
.
Science
.
2002
;
295
(
5562
):
2097
-
2100
.
43.
Cook
M
,
Briggs
D
,
Craddock
C
, et al
.
Donor KIR genotype has a major influence on the rate of cytomegalovirus reactivation following T-cell replete stem cell transplantation
.
Blood
.
2006
;
107
(
3
):
1230
-
1232
.
44.
Mancusi
A
,
Ruggeri
L
,
Urbani
E
, et al
.
Haploidentical hematopoietic transplantation from KIR ligand-mismatched donors with activating KIRs reduces nonrelapse mortality
.
Blood
.
2015
;
125
(
20
):
3173
-
3182
.
45.
Muccio
L
,
Bertaina
A
,
Falco
M
, et al
.
Analysis of memory-like natural killer cells in human cytomegalovirus-infected children undergoing αβ+T and B cell-depleted hematopoietic stem cell transplantation for hematological malignancies
.
Haematologica
.
2016
;
101
(
3
):
371
-
381
.
46.
Wang
H
,
Grzywacz
B
,
Sukovich
D
, et al
.
The unexpected effect of cyclosporin A on CD56+CD16− and CD56+CD16+ natural killer cell subpopulations
.
Blood
.
2007
;
110
(
5
):
1530
-
1539
.
47.
Ruggeri
L
,
Mancusi
A
,
Capanni
M
, et al
.
Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value
.
Blood
.
2007
;
110
(
1
):
433
-
440
.
48.
Lamb
LS
Jr
,
Henslee-Downey
PJ
,
Parrish
RS
, et al
.
Increased frequency of TCR gamma delta + T cells in disease-free survivors following T cell-depleted, partially mismatched, related donor bone marrow transplantation for leukemia
.
J Hematother
.
1996
;
5
(
5
):
503
-
509
.
49.
Lang
P
,
Feuchtinger
T
,
Teltschik
HM
, et al
.
Improved immune recovery after transplantation of TCRαβ/CD19-depleted allografts from haploidentical donors in pediatric patients
.
Bone Marrow Transplant
.
2015
;
50
(
suppl 2
):
S6
-
S10
.
50.
Giorgiani
G
,
Bozzola
M
,
Locatelli
F
, et al
.
Role of busulfan and total body irradiation on growth of prepubertal children receiving bone marrow transplantation and results of treatment with recombinant human growth hormone
.
Blood
.
1995
;
86
(
2
):
825
-
831
.
51.
Socié
G
,
Curtis
RE
,
Deeg
HJ
, et al
.
New malignant diseases after allogeneic marrow transplantation for childhood acute leukemia
.
J Clin Oncol
.
2000
;
18
(
2
):
348
-
357
.
52.
Willard
VW
,
Leung
W
,
Huang
Q
,
Zhang
H
,
Phipps
S
.
Cognitive outcome after pediatric stem-cell transplantation: impact of age and total-body irradiation
.
J Clin Oncol
.
2014
;
32
(
35
):
3982
-
3988
.
53.
Shi-Xia
X
,
Xian-Hua
T
,
Hai-Qin
X
,
Bo
F
,
Xiang-Feng
T
.
Total body irradiation plus cyclophosphamide versus busulphan with cyclophosphamide as conditioning regimen for patients with leukemia undergoing allogeneic stem cell transplantation: a meta-analysis
.
Leuk Lymphoma
.
2010
;
51
(
1
):
50
-
60
.
54.
Peters
C
,
Schrappe
M
,
von Stackelberg
A
, et al
.
Stem-cell transplantation in children with acute lymphoblastic leukemia: a prospective international multicenter trial comparing sibling donors with matched unrelated donors-The ALL-SCT-BFM-2003 trial
.
J Clin Oncol
.
2015
;
33
(
11
):
1265
-
1274
.
55.
Fuchs
EJ
.
HLA-haploidentical blood or marrow transplantation with high-dose, post-transplantation cyclophosphamide
.
Bone Marrow Transplant
.
2015
;
50
(
suppl 2
):
S31
-
S36
.
56.
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
.
57.
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
.
58.
Zhou
X
,
Dotti
G
,
Krance
RA
, et al
.
Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation
.
Blood
.
2015
;
125
(
26
):
4103
-
4113
.
59.
Locatelli
F
,
Schrappe
M
,
Bernardo
ME
,
Rutella
S
.
How I treat relapsed childhood acute lymphoblastic leukemia
.
Blood
.
2012
;
120
(
14
):
2807
-
2816
.

Author notes

*

A.B. and L.M. contributed equally to this study.

Sign in via your Institution