• Haploidentical transplant achieves outcomes similar to those of identical-sibling transplant for AML patients in first remission.

  • Haploidentical transplant is a valid postremission treatment of intermediate- or high-risk AML patients lacking an identical donor.

The effects of HLA-identical sibling donor (ISD) hematopoietic stem cell transplantation (HSCT) on adults with intermediate- or high-risk acute myeloid leukemia (AML) in the first complete remission (CR1) are well established. Previous single-center studies have demonstrated similar survival after unmanipulated haploidentical donor (HID) vs ISD HSCT for hematologic malignancies. To test the hypothesis that haploidentical HSCT would be a valid option as postremission therapy for AML patients in CR1 lacking a matched donor, we designed a disease-specific, prospective, multicenter study. Between July 2010 and November 2013, 450 patients were assigned to undergo HID (231 patients) or ISD HSCT (219 patients) according to donor availability. Among HID and ISD recipients, the 3-year disease-free survival rate was 74% and 78% (P = .34), respectively; the overall survival rate was 79% and 82% (P = .36), respectively; cumulative incidences of relapse were 15% and 15% (P = .98); and those of the nonrelapse-mortality were 13% and 8% (P = .13), respectively. In conclusion, unmanipulated haploidentical HSCT achieves outcomes similar to those of ISD HSCT for AML patients in CR1. Such transplantation was demonstrated to be a valid alternative as postremission treatment of intermediate- or high-risk AML patients in CR1 lacking an identical donor. This trial was registered at www.chictr.org as #ChiCTR-OCH-10000940.

Acute myeloid leukemia (AML) is a common hematologic malignant disease. HLA-identical sibling donor (ISD) hematopoietic stem cell transplantation (HSCT) offers significantly improved overall survival (OS) for intermediate- or high-risk AML patients compared with chemotherapy alone1-5 ; thus, it is recommended as a first-line postremission treatment in this clinical situation. Recent progress in haploidentical donor (HID) transplantation provides the benefits of rapid and near-universal donor availability.6-8  However, whether haploidentical HSCT could serve as a first-line postremission therapy in intermediate- or high-risk AML remains to be determined.

In recent years, our group established an unmanipulated haploidentical blood and marrow transplantation protocol in which anti-thymocyte globulin (ATG) in the conditioning and the powerful posttransplantation graft-versus-host disease (GVHD) prophylaxis probably provided a major contribution to controlling alloreactivity.7,8  Additionally, our recent single-institute, patient self-selected study showed that haploidentical HSCT was superior to chemotherapy alone for patients with intermediate- or high-risk AML in the first complete remission (CR1).9  Undoubtedly, randomized trials are needed to determine if haploidentical transplantation is, in fact, superior to chemotherapy for AML in CR1. However, this may be difficult to determine in this setting because of ethical and practical reasons given the inferior outcomes observed after chemotherapy-based postremission strategies compared with those after allo-HSCT.1,3-5  Conversely, prior single-center analyses demonstrated similar survival after unmanipulated HID HSCT using ATG vs ISD HSCT for hematologic malignancies.10,11 

Taken together, we hypothesized that haploidentical HSCT is a valid option as a postremission treatment of AML in CR1. The results from haploidentical HSCT have not formally been compared in prospective studies with those from ISDs to evaluate the value of haploidentical HSCT for the specific, homogenous disease of intermediate- or high-risk AML in CR1. In addition, evaluating a uniform haplo-HSCT protocol in more clinical centers would enable prospective, multicenter trials on the standardized treatment. Therefore, based on the current prospective, multicenter, disease-specific study, we compare the outcomes of consecutive AML patients in CR1 undergoing T-cell–replete haploidentical HSCT uniformly performed at 3 centers with all contemporaneous ISD HSCT.

Study design

We conducted a disease-specific, multicenter, nonrandomized trial. Patients were assigned to undergo haploidentical or ISD HSCT according to donor availability. The primary end point was disease-free survival (DFS), OS, and relapse rates. Secondary end points included an evaluation of immunologic reconstitution. Enrollment began in July 2010 and ended after the accrual goal was met, which occurred in November 2013. The analysis includes data collected as of November 30, 2014. The median follow-up for surviving patients is 952 days (range; 365-1612 days) after HSCT. This article focuses on the primary end points. Analyses of immune reconstitution are ongoing.

Patients

Eligible patients were aged 15 to 60 years and had intermediate- or high-risk AML in CR1 with no contraindications to HSCT. HLA-ISD was the first choice for allotransplantation. If an HLA-ISD was unavailable, subjects without a suitable closely HLA-matched unrelated donor (>8 of 10 matching HLA-A, B, C, DR, and DQ loci; and >5 of 6 matching HLA-A, B, and DR loci) after 2 cycles of consolidation were eligible for HLA-haplotype transplantation. Consecutive patients receiving HID (n = 231) or ISD (n = 219) HSCT during the study period were enrolled. For this comparative analysis to arrive at comparable patient cohorts that received transplants during the same time interval, we excluded patients who received unrelated donor HSCT (n = 58).

The study was performed in accordance with the Declaration of Helsinki and was approved by the local institutional review board. All donors and recipients gave written informed consent before enrollment. All authors vouch for the accuracy and completeness of the reported data and analyses and for the adherence of the study to the protocol. This study was registered as #ChiCTR-OCH-10000940 at www.chictr.org.

Procedures

Patients were classified as intermediate- or high-risk based on National Comprehensive Cancer Network cytogenetic abnormalities.12  Information on the monosomal karyotype,13  Center for International Blood and Marrow Transplant classification,14  nucleophosmin (NPM), and fms-related tyrosine kinase 3 (FLT3) is listed in Table 1. The protocol required DNA typing of the patient and donor at intermediate resolution for HLA-A, B and at high resolution for DRB1. HID subjects received a graft from a family member sharing 1 HLA haplotype with the recipient but differed to a variable degree for the HLA-A, B, and DR antigens of the unshared HLA haplotype. Granulocyte colony-stimulating factor (G-CSF; 5 µg/kg of body weight per day for 5 days) was used to mobilize the BM and PB. The target mononuclear cell count was >6 × 108/kg of recipient weight. Unmanipulated BM (harvested on day 4 after G-CSF) and PB stem cells (harvested on day 5 after G-CSF) were infused into the recipient on the day of collection. All HID patients received both BM and PB mobilized with G-CSF.

Table 1

Patient and graft characteristics

CharacteristicsHaploidentical, N = 231Identical sibling, N = 219
Age, y, median (range) 28 (15-57) 40 (17-60) 
Gender, no. (%)   
 Male 144 (62) 122 (56) 
 Female 87 (38) 97 (44) 
French-American-British subtype, no. (%)   
 M0 5 (2) 6 (3) 
 M1 11 (5) 23 (10) 
 M2 105 (46) 85 (39) 
 M4 25 (11) 30 (14) 
 M5 68 (29) 54 (25) 
 M6 11 (5) 12 (5) 
 M7 1 (0.4) 1 (0.5) 
 Undetermined 5 (2) 8 (4) 
WBC count at diagnosis 16.9 (0.3-368) 1.8 (0.5-418) 
 <50 000/mm3, no. (%) 159 (69) 164 (75) 
 >50 000/mm3, no. (%) 72 (31) 55 (25) 
Duration from diagnosis to transplant, mo   
 Median (range) 6.0 (2.0-12) 5.0 (2.0-12) 
Cytogenetic risk group,*no. (%)   
 Intermediate, normal 99 (43) 110 (50) 
 Intermediate, abnormal 84 (36) 77 (35) 
 High 48 (21) 32 (15) 
Autosome monosomal karyotype among abnormals   
 No 119 (90) 105 (96) 
 Yes 13 (10) 4 (4) 
Courses required achieving remission, no. (%)   
 1 155 (67) 156 (71) 
 2 60 (26) 50 (23) 
 3-4 16 (7) 13 (6) 
Donor-patient sex matched, no. (%)   
 Male to male 90 (39) 65 (30) 
 Male to female 55 (24) 61 (28) 
 Female to male 51 (22) 57 (26) 
 Female to female 35 (15) 36 (16) 
Donor-patient relation, no. (%)   
 Father donor 76 (33)  
 Mother donor 39 (17)  
 Sibling donor 92 (40) 219 (100) 
 Children donor 24 (10)  
Graft type, no. (%)   
 BM + PB cell 231 (100) 124 (57) 
 BM  14 (6) 
 PB cell  81 (37) 
Median nucleated cells, ×108 cells per kg (range) 7.6 (4.2-15.8) 7.2 (4.0-12.4) 
Median CD34+ cells, ×106/kg (range) 2.5 (0.4-17.0) 2.2 (0.3-8.3) 
Follow-up time in survivors from complete remission, mo   
 No. of evaluable patients 188 (81%) 184 (84%) 
 Median (range) 36 (16-63) 37 (14-66) 
CharacteristicsHaploidentical, N = 231Identical sibling, N = 219
Age, y, median (range) 28 (15-57) 40 (17-60) 
Gender, no. (%)   
 Male 144 (62) 122 (56) 
 Female 87 (38) 97 (44) 
French-American-British subtype, no. (%)   
 M0 5 (2) 6 (3) 
 M1 11 (5) 23 (10) 
 M2 105 (46) 85 (39) 
 M4 25 (11) 30 (14) 
 M5 68 (29) 54 (25) 
 M6 11 (5) 12 (5) 
 M7 1 (0.4) 1 (0.5) 
 Undetermined 5 (2) 8 (4) 
WBC count at diagnosis 16.9 (0.3-368) 1.8 (0.5-418) 
 <50 000/mm3, no. (%) 159 (69) 164 (75) 
 >50 000/mm3, no. (%) 72 (31) 55 (25) 
Duration from diagnosis to transplant, mo   
 Median (range) 6.0 (2.0-12) 5.0 (2.0-12) 
Cytogenetic risk group,*no. (%)   
 Intermediate, normal 99 (43) 110 (50) 
 Intermediate, abnormal 84 (36) 77 (35) 
 High 48 (21) 32 (15) 
Autosome monosomal karyotype among abnormals   
 No 119 (90) 105 (96) 
 Yes 13 (10) 4 (4) 
Courses required achieving remission, no. (%)   
 1 155 (67) 156 (71) 
 2 60 (26) 50 (23) 
 3-4 16 (7) 13 (6) 
Donor-patient sex matched, no. (%)   
 Male to male 90 (39) 65 (30) 
 Male to female 55 (24) 61 (28) 
 Female to male 51 (22) 57 (26) 
 Female to female 35 (15) 36 (16) 
Donor-patient relation, no. (%)   
 Father donor 76 (33)  
 Mother donor 39 (17)  
 Sibling donor 92 (40) 219 (100) 
 Children donor 24 (10)  
Graft type, no. (%)   
 BM + PB cell 231 (100) 124 (57) 
 BM  14 (6) 
 PB cell  81 (37) 
Median nucleated cells, ×108 cells per kg (range) 7.6 (4.2-15.8) 7.2 (4.0-12.4) 
Median CD34+ cells, ×106/kg (range) 2.5 (0.4-17.0) 2.2 (0.3-8.3) 
Follow-up time in survivors from complete remission, mo   
 No. of evaluable patients 188 (81%) 184 (84%) 
 Median (range) 36 (16-63) 37 (14-66) 

Patient age and duration from diagnosis to transplant differed significantly between the 2 groups (P < .001). There were no other significant between-group differences.

BM, bone marrow; PB, peripheral blood; WBC, white blood cell.

*

Cytogenetic studies were carried out using standard techniques. Molecular screening for fusion genes was offered to all patients. Patients were classified as low, intermediate, or high risk on the basis of National Comprehensive Cancer Network cytogenetic abnormalities. Cytogenetic abnormalities t(8;21)(q22;q22), t(15;17)(q22;q21), and inv(16) or t(16;16)(p13;q22) were considered low risk. Complex cytogenetic abnormalities (defined as at least 3 unrelated cytogenetic clones) and −5/5q-, −7/7q-, abn(3q), t(6;9)(q23;q34), abn(11q23)-non t(9;11), and t(9;22)(q34;q11) were considered high risk. Patients without favorable (low risk) or adverse (high risk) abnormalities or without karyotype information were classified as intermediate risk.12  Patients were not reclassified according to the recently published Center for International Blood and Marrow Transplant criteria because only 10 patients fit in the adverse group.14  Information on FLT3 internal tandem duplication (ITD) was only available in 187 patients (positive in 40 and negative in 147). Information on NPM1 was available in 222 patients (normal karyotype with NPM1 mutation and FLT3-ITD in 11 patients, abnormal karyotype with NPM1 mutation in 11 patients, normal karyotype with NPM1 wild type in 110 patients, and abnormal karyotype with NPM1 wild type in 90 patients).

The conditioning therapy for the HID group was as follows: cytarabine (4 g/m2 per day) IV on days −10 to −9; busulfan (3.2 mg/kg per day) IV on days −8 to −6; cyclophosphamide (1.8 g/m2 per day), IV on days −5 to −4; methyl chloride hexamethylene urea nitrate (Me-CCNU) (250 mg/m2 per day), orally once on day −3; and ATG (2.5 mg/kg per day; Sang Stat, Lyon, France) IV on days −5 to −2. Patients in the ISD group received hydroxycarbamide (80 mg/kg) orally on day −10 and a lower dose of cytarabine (2 g/m2 per day) on day −9, but otherwise, an identical regimen to the HID patients without ATG was employed. GVHD prophylaxis regimen consisted of cyclosporine A, mycophenolate mofetil, and short-term methotrexate.11  Between May 2012 and November 2013, 25 subjects in Peking University were randomized to receive low-dose corticosteroid prophylaxis (www.clinicaltrials.gov, #NCT01607580).

After completion of the study treatment, BM samples were analyzed at 1, 2, 3, 4.5, 6, 9, and 12 months after transplantation and at 6-month intervals thereafter for the monitoring of minimal residual disease (MRD), defined as previously reported.15,16  In Peking University, modified donor lymphocyte infusion (DLI) would be given before hematologic relapse as the intervention therapy (preemptive DLI) after 3 months post-HSCT following a trial of immunosuppressant withdrawal. The detailed criteria for preemptive DLI administration included the following: (1) patients scored as MRD+ if they had 2 consecutive positive results using flow cytometry or Wilms’ tumor gene 1 or were both flow cytometry–positive and Wilms’ tumor gene 1– positive in a single sample within 1 year after transplantation; (2) no uncontrolled GVHD or life-threatening infection; and (3) with donor availability and willingness. Patients with GVHD first received GVHD therapy. After GVHD was controlled, MRD testing was repeated, and those patients that remained MRD+ received modified DLI. In total, 23 patients (13 HID, 10 ISD) required this preemptive approach, which occurred at a median of 171 days (range, 109-319 days) after transplantation. The modified DLI regimen was as previously described.17  When a hematologic relapse was diagnosed after HSCT, posttransplant immune suppression was immediately discontinued. If patients did not develop GVHD within 2 weeks, and if patients agreed to receive targeted therapeutic modified DLI and their donors also agreed to undergo PB stem cell collection again, the patients would receive chemotherapy followed by modified DLI; otherwise, the patients would receive chemotherapy alone.18  Antileukemia chemotherapy before DLI included aclacinomycin (10 mg/m2 per day for 5 days) and Ara-C 100 (mg/m2 per day for 5 days); fludarabine (30 mg/m2 per day for 5 days), Ara-C (1.0 g every 12 hours for 10 doses), and G-CSF; or harringtonine (2 mg/m2 per day for 5 days), aclacinomycin (10 mg/m2 per day for 5 days), and Ara-C (100 mg/m2 per day for 5 days). In total, 18 patients (11 HID, 7 ISD) were given this targeted therapeutic DLI (see “Results”). The median number of CD3+ cells infused in each patient was 0.58 (interquartile range, 0.13–2.03) × 108/kg. Chimerism analyses were done by DNA fingerprinting of short tandem repeat on blood samples.

Statistical analysis

Comparisons of patient characteristics between the 2 groups were performed using the Mann-Whitney U test for continuous variable and χ2 test for categorical data. Relapse, nonrelapse mortality (NRM), engraftment, and GVHD were estimated as cumulative incidences, taking into account competing risks. NRM was defined as death from any cause in the first 28 days post-HSCT or death without evidence of disease recurrence beyond day 28. Relapse was defined as recurrence of BM blasts >5%, reappearance of blasts in the blood, or development of extramedullary disease infiltrates at any site. Assessments of engraftment and chimerism were previously described in detail.11  Chronic GVHD was classified as limited or extensive and was also classified as mild, moderate, or severe by the National Institutes of Health consensus criteria. GVHD was evaluated and graded by a single practitioner within the program.

Survival functions were estimated using the Kaplan-Meier method and compared by the log-rank test. The starting point for comparing outcomes between the cohorts was the point when complete remission was declared. Univariate probabilities of survival and DFS were calculated using a left-truncated version of the Kaplan-Meier estimator with 95% confidence intervals (CIs). Cumulative incidence of relapse (CIR) and NRM were calculated using a left-truncated version of cumulative incidence function to accommodate competing risks. To adjust for the differences in baseline characteristics, left-truncated versions of the Cox proportional hazards regression models were used to evaluate the relative risk of subjects receiving either allotransplant by forcing the main interest variable (HID vs ISD, using ISD as the reference group) into the models. Backward elimination with a criterion of P < .10 for retention was used to select a final model. The following variables were analyzed: patient age (>50 years or not),19  sex (female or male), WBC count at diagnosis (>50 × 109/L or not), cytogenetic risk group (intermediate or high), and courses to achieving CR1 (>2 courses or not). The assumption of proportional hazards for each factor in the Cox model was tested. The test indicated that the proportionality assumptions hold. End-point events for DFS included morphologic relapse and NRM in CR1. Patients with MRD were not considered relapsed for DFS determination. Since conventional measures of leukemia-free survival (LFS) fail to take into account the capacity of DLI to produce durable molecular remissions in patients who relapse, thus underestimating the ability of allografting to produce sustained DFS even in patients who have relapsed, the current LFS probabilities were also estimated.20  The SAS V. 9.3 (SAS Institute Inc., Cary, NC) and SPSS 13.0 software packages (SPSS Inc., Chicago, IL) were used for data analyses.

Patient characteristics

As shown in Table 1, the groups were well matched, except that patients undergoing HID-HSCT were significantly younger and there was a little delay from diagnosis to transplantation. Because of the 1-child policy in China over the past 30 years, parent donors accounted for 50% of the HID group. As a result, the HID cohort was younger. The phenomenon of 1 month longer to transplant in the HID vs the ISD group may be explained by the possible hesitation of patients, which is associated with unawareness arising from a lack of a doctor’s referral during chemotherapy in small institutes or insufficient confidence in HID transplantation. Patients undergoing HSCT using HIDs were mismatched at a median of 3 of 6 HLA-A, B, and DRB1 loci by molecular typing (range, 1-3).

Survival

The 3-year probabilities of DFS were 74% (95% CI, 67-81) and 78% (95% CI, 72-85; P = .34) among HID and ISD patients (Figure 1A). The 3-year probabilities of current LFS were 76% (95% CI, 64-87) and 80% (95% CI, 70-91) among HID and ISD patients. The 3-year probabilities of OS were 79% (95% CI, 73-85) and 82% (95% CI, 76-88; P = .36) among HID and ISD patients (Figure 1B). The 3-year cumulative incidences of NRM were 13% (95% CI, 7-19) and 8% (95% CI, 4-12; P = .13; Figure 1C). Subgroup analysis with available FLT-ITD results at diagnosis showed that the 3-year DFS rates were 79% (95% CI, 71-88) and 60% (95% CI, 42-78) among patients without FLT-ITD and patients with FLT-ITD (P = .02).

Figure 1

Outcomes after transplantation according to donor source. The rate of DFS (A) and the rate of OS (B). (C) The incidence of NRM. (D) The incidence of relapse. The incidence of neutrophil engraftment (>500 neutrophils per mm3) (E) and the incidence of platelet engraftment (>20 000 platelets per cubic mm3, without platelet transfusion during the prior 7 days) (F). The incidence of acute GVHD of grades 2 to 4 (G) and the incidence of chronic GVHD (H).

Figure 1

Outcomes after transplantation according to donor source. The rate of DFS (A) and the rate of OS (B). (C) The incidence of NRM. (D) The incidence of relapse. The incidence of neutrophil engraftment (>500 neutrophils per mm3) (E) and the incidence of platelet engraftment (>20 000 platelets per cubic mm3, without platelet transfusion during the prior 7 days) (F). The incidence of acute GVHD of grades 2 to 4 (G) and the incidence of chronic GVHD (H).

Close modal

Univariate analysis treated the patient age in different ways: (1) as a continuous variable; and (2) in addition, a dichotomous variable of 50 years old was used as reported by Yanada et al19  for matched donor transplant. Univariate analysis showed that patient age did not affect DFS as a continuous variable (P = .39), whereas patient age over 50 years old significantly affected DFS (P = .03). This is likely because of the significantly higher proportion of WBC count at diagnosis as well as higher proportion of cytogenetically high-risk patients within the older group as compared with that in the younger group (45% vs 26%, P = .01; 30% vs 17%, P = .03, respectively). Of note, however, the differences in DFS rates between age groups differed according to donor source. DFS was not significantly different for patients over 50 years or not (62% vs 75%, P = .37) in the HID cohort, whereas DFS was significantly lower among those over 50 years than those under 50 years (61% vs 81%, P = .02) in the ISD group. The previously mentioned different proportion of disease characteristics and the more prominent differences within the ISD group are likely to have contributed to differences in DFS between the 2 age groups. In addition, the effect of patient age on DFS disappeared in multivariate analysis (P = .11).

A multivariate analysis failed to show significant differences in DFS, OS, or NRM rates between the 2 cohorts; cytogenetic risk group and WBC count at diagnosis were independent risk factors associated with DFS and OS, and patient age, sex, or courses required to achieving CR1 did not significantly affect mortality (Table 2).

Table 2

Results of multivariate analysis of outcomes and contributing factors

OutcomeHazard ratio (95% CI)P
DFS   
 Identical sibling vs haploidentical 0.83 (0.52-1.30) .42 
 Other significant factors   
  Cytogenetic risk high vs intermediate 1.93 (1.18-3.15) .008 
  WBC >50 000 vs <50 000 per mm3 1.92 (1.21-3.04) .005 
OS   
 Identical sibling vs haploidentical 0.80 (0.47-1.37) .43 
 Other significant factors   
  Cytogenetic risk high vs intermediate 0.62 (0.36-1.06) .08 
  WBC >50 000 vs <50 000 per mm3 0.51 (0.31-0.83) .007 
Relapse   
 Identical sibling vs haploidentical 1.06 (0.58-1.92) .85 
 Other significant factors   
  Cytogenetic risk high vs intermediate 1.88 (1.05-3.76) .04 
  WBC >50 000 vs <50 000 per mm3 2.15 (1.18-3.91) .01 
NRM   
 Identical sibling vs haploidentical 0.58 (0.28-1.19) .14 
OutcomeHazard ratio (95% CI)P
DFS   
 Identical sibling vs haploidentical 0.83 (0.52-1.30) .42 
 Other significant factors   
  Cytogenetic risk high vs intermediate 1.93 (1.18-3.15) .008 
  WBC >50 000 vs <50 000 per mm3 1.92 (1.21-3.04) .005 
OS   
 Identical sibling vs haploidentical 0.80 (0.47-1.37) .43 
 Other significant factors   
  Cytogenetic risk high vs intermediate 0.62 (0.36-1.06) .08 
  WBC >50 000 vs <50 000 per mm3 0.51 (0.31-0.83) .007 
Relapse   
 Identical sibling vs haploidentical 1.06 (0.58-1.92) .85 
 Other significant factors   
  Cytogenetic risk high vs intermediate 1.88 (1.05-3.76) .04 
  WBC >50 000 vs <50 000 per mm3 2.15 (1.18-3.91) .01 
NRM   
 Identical sibling vs haploidentical 0.58 (0.28-1.19) .14 

Relapse

The 3-year CIRs were 15% (95% CI, 9-21) and 15% (95% CI, 9-21; P = .98) among HID and ISD patients (Figure 1D). Multivariate analysis failed to show significant differences in CIR between the 2 cohorts; cytogenetic risk group and WBC count at diagnosis were independent risk factors affecting CIR (Table 2).

At Peking University, full donor chimerism was detected in 22 of the 23 (96%) patients at the time of preemptive DLI. Among the 23 patients (13 HID, 10 ISD) receiving modified DLI as intervention for positive MRD, 7 (4 of 13 HID and 3 of 10 ISD) eventually relapsed at a median of 194 days (range, 85-780 days) after DLI, and 15 patients were alive without relapse with a median of 940 days (range, 214-1349 days) after DLI; the remaining 1 patient died of infection.

Until the last follow-up, 26 and 27 patients experienced relapse after HID- or ISD-HSCT; of the 26 HID patients, 11 received DLI plus chemotherapy, 10 received chemotherapy alone, and 5 received no further therapy. In the DLI plus chemotherapy group, 6 patients achieved the second complete remission (CR2) and are still alive with a median of 323 days after relapse, whereas 3 died of relapse (76, 209, and 397 days after DLI) and the other 2 died of infection. The patients who received chemotherapy alone or no therapy died at a median of 60 days after relapse. Among ISD patients, 27 experienced relapse; of these, 7 received DLI plus chemotherapy, 8 received chemotherapy alone, and 12 received no further therapy. In the DLI plus chemotherapy group, 4 patients achieved CR2 and are still alive with a median of 844 days after relapse, whereas 3 died of relapse (30, 146, and 185 days after DLI). Among the 8 patients who received chemotherapy alone, 2 patients achieved CR2 and are still alive (94 and 194 days after relapse) and 6 died at a median of 36 days after relapse. Although the 12 patients receiving no further therapy died at a median of 30 days after relapse. In total, 10 of the 18 patients who underwent therapeutic DLI and 2 of the other 35 patients who did not undergo DLI achieved CR2 and are still alive (P < .001). Full donor chimerism was detected in 14 of the 18 (80%) patients at the time of targeted therapeutic DLI. The 1-year CIR after preemptive DLI or targeted therapeutic DLI was 22% and 33% (P = .42), respectively.

Engraftment and GVHD

All patients achieved donor-cell engraftment. The median time to achieve neutrophil engraftment was 2 days shorter (P = .004; Figure 1E) after HID-HSCT, whereas that of platelet engraftment was 3 days shorter after ISD-HSCT (P < .001; Figure 1F).

The cumulative incidences of grades 2 to 4 acute GVHD at 100 days were 36% (95% CI, 30-42) and 13% (95% CI, 9-17) after HID- or ISD-HSCT, respectively (P < .001; Figure 1G); the cumulative incidences for grades 3 to 4 acute GVHD at 100 days were 10% (95% CI, 6-14) and 3% (95% CI, 1-5), respectively (P = .004). The cumulative incidences of chronic GVHD at 1 year were 42% (95% CI, 36-48) vs 15% (95% CI, 10-20) for HID vs ISD patients (P < .001; Figure 1H); the cumulative rates of severe chronic GVHD at 1 year were 12% (95% CI, 8-16) and 2% (95% CI, 0-4), respectively (P < .001).

To our knowledge, our study represents the first formal comparison of HID transplantation with allo-HSCT using ISDs in a disease-specific population of patients with intermediate- or high-risk AML in CR1. Although the study was nonrandomized, its strengths include the prospective nature, uniform treatment modality at the multiple centers, contemporaneous population of patients studied, and the number of patients analyzed. Furthermore, the comparison provides a particular opportunity for exploring the up-to-date undefined role of HID HSCT as postremission therapy for intermediate- or high-risk AML patients in CR1. Our study is the first to demonstrate that HID-HSCT is comparable to ISD-HSCT in the rate of DFS as postremission treatment of this portion of patients and the rates of OS and relapse were also similar in the 2 groups.

In our analysis, a 1.5-year NRM rate of 10% was achieved in the HID patients, which is comparable with the low 1-year NRM of 7% observed in studies of T-cell–replete HID HSCT using posttransplantation cyclophosphamide in nonmyeloablative settings.21  Our analysis demonstrates that low rates of NRM can be achieved among AML patients in CR1 undergoing both HID and ISD transplantation performed in multi-institutions. Nonetheless, both groups are relatively young, and this may have contributed to reduce the severity of NRM. It should also be noted that NRM was substantially higher with haploidentical transplants compared with those with matched related donors, although it did not reach statistical significance. Regarding GVHD, similar to our recently updated results,22  the rates of GVHD observed after HID transplantation in our population were also comparable to those described in 53 HID transplantations using posttransplantation cyclophosphamide reported by Bashey et al.23  However, the rates of GVHD observed after ISD transplantation in our population were much lower than those after nonmyeloablative ISD transplant described in Bashey’s report.23  Recently, a multicenter phase 2 study from the Chinese Bone Marrow Transplant Cooperative Group showed that a combination of cyclosporine A, methotrexate, and mycophenolate mofetil for GVHD prophylaxis significantly decreased the risk of acute GVHD compared with historical controls in ISD transplantation.24,25  This finding is confirmed in our ISD cohort. Thus, the higher incidence of GVHD in the HID group is partly because of the relatively low incidence in the ISD cohort. Furthermore, the GVHD rates in HID group were quite acceptable as discussed previously and the rates of GVHD-related death were similar between the 2 groups (data not shown) because of effective therapy. The CIR of 15% at 3 years in the HID group was similar to that of 12% reported in our previous study9  and to patients who underwent ISD-HSCT in the present study but lower than those reported in other studies.26-28  Posttransplantation monitoring of MRD and modified DLI intervention guided by MRD level might have further decreased the relapse rate.16,17 

Survival should be the primary end point for assessing the efficacy of a treatment modality for leukemia. Patient survival in this study was similar to that of our own previous study,9  but higher than that observed in other previous reports26-28  because of a more favorable relapse rate and a much lower NRM. Several factors likely contributed to our superior transplantation outcomes. First, the patients in other studies have often included AML patients in CR1 undergoing HSCT with very high-risk features such as poor cytogenetic, high WBC count at diagnosis, delayed CR,26-28  while patients in our present study with these high-risk features accounted for 56% of all study population. Second, as mentioned previously, younger age in our present study than in other studies (median age of 28 vs 33 to 37 in other haploidentical report26-28  or 50 years old in ISD transplant), intensive GVHD prophylaxis, and effective infection control may have attributed to the much lower NRM rate (13% vs 32% to 36% at 3 years26-28 ). Third, quantification of MRD after allo-HSCT is reportedly predictive of relapse, and individualized intervention can decrease relapse and improve survival in high-risk AML patients after transplantation. All the more, although the effects of HLA-matched allo-HSCT on patients with AML in CR1 are well established,1-5  the application of haploidentical HSCT in AML needs to be broadened.29,30  Our observations support the rationale for considering haploidentical HSCT as a front-line postremission treatment option for adults with AML without favorable cytogenetics who lack a matched donor. However, one should be particularly cautious in interpreting the results, given the age difference between the 2 groups. Although OS is similar, a matched sibling transplant should still be considered the standard of care, compared with haploidentical transplants since it has lower morbidity (GVHD) and NRM.

As a limitation to our current study, the patients were not randomized to receive HID- or ISD-HSCT for ethical and practical reasons. Although biologically selected postremission treatment with HID-HSCT or ISD-HSCT based on the availability of a matched donor might have minimized this limitation to some extent, there were still imbalanced features between the 2 groups (the HID cohort was younger and had longer time to transplant). In addition, insufficient confidence in HID transplant resulted in less HID transplant (maybe more patients with very high-risk features were tended to be “selected” into HID group as a result) than the number could be, although consecutive patients entering either transplant group were analyzed. We made adjustments in the multivariate analysis that produced no effects of age on DFS. This finding is consistent with those from our own previous studies and previous studies from other groups,31-33  but it contradicts some reports.19,34,35  We did left-truncation analysis to adjust for the time-to-transplant bias. Despite our effort on these adjustments, the possible influence of the imbalanced factors on comparable outcomes between the 2 study cohorts cannot be eliminated completely. Another limitation is that insufficient data on FLT3, comorbidities and NK cell alloreactivity limit our ability to incorporate these important variables into analysis, although subgroup results on FLT3 were inconsistent with those reported by Brunet et al.36  Controversial effects of natural killer cell alloreactivity between the Europe and America study and our study as reviewed in detail by our group.37,38 

In summary, this comparison suggests that outcomes after transplantation using HIDs with ATG are comparable to those after ISDs transplantation for AML patients in remission. HID transplantation as postremission therapy should be recommended as a valid alternative option for patients with intermediate- or high-risk AML in CR1 for whom no matched donor is available.

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.

The authors thank Prof Bob Löwenberg, Prof Nancy Berliner, and Prof Jacob Rowe for kindly reviewing this paper; Dr Zhuang Tao (Beijing Medical Development Clinical Research Institute) for the careful statistical review; and the principal investigators and the skilled teams at each of the participating sites.

This work was partly supported by the Collaborative Innovation Center of Hematology China, the Key Program of National Natural Science Foundation of China (grant 81230013), the National Natural Science Foundation of China (grant 81400143), and the Scientific Research Foundation for Capital Medicine Development (grant 2011-4022-08).

Contribution: Y.W. and Q.-F.L. wrote the manuscript; X.-J.H. and D.-P.W. designed the protocol; all authors contributed patients, provided clinical and laboratory data, and revised and corrected the manuscript; Y.W. and X.-J.H. performed the analysis; and X.-J.H., Q.-F.L., and D.-P.W. approved and recommended the protocol within each institute.

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

Correspondence: Xiao-Jun Huang, Peking University People’s Hospital, Peking University Institute of Hematology, Beijing 100044, China; e-mail: xjhrm@medmail.com.cn; and De-Pei Wu, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; e-mail: wudepei@medmail.com.cn.

1
Döhner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
European LeukemiaNet
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
2
Basara
 
N
Schulze
 
A
Wedding
 
U
, et al. 
East German Study Group Hematology and Oncology (OSHO)
Early related or unrelated haematopoietic cell transplantation results in higher overall survival and leukaemia-free survival compared with conventional chemotherapy in high-risk acute myeloid leukaemia patients in first complete remission.
Leukemia
2009
, vol. 
23
 
4
(pg. 
635
-
640
)
3
Cornelissen
 
JJ
van Putten
 
WL
Verdonck
 
LF
, et al. 
Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom?
Blood
2007
, vol. 
109
 
9
(pg. 
3658
-
3666
)
4
Schlenk
 
RF
Döhner
 
K
Mack
 
S
, et al. 
Prospective evaluation of allogeneic hematopoietic stem-cell transplantation from matched related and matched unrelated donors in younger adults with high-risk acute myeloid leukemia: German-Austrian trial AMLHD98A.
J Clin Oncol
2010
, vol. 
28
 
30
(pg. 
4642
-
4648
)
5
Koreth
 
J
Schlenk
 
R
Kopecky
 
KJ
, et al. 
Allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission: systematic review and meta-analysis of prospective clinical trials.
JAMA
2009
, vol. 
301
 
22
(pg. 
2349
-
2361
)
6
Reisner
 
Y
Hagin
 
D
Martelli
 
MF
Haploidentical hematopoietic transplantation: current status and future perspectives.
Blood
2011
, vol. 
118
 
23
(pg. 
6006
-
6017
)
7
Chang
 
YJ
Huang
 
XJ
Haploidentical SCT: the mechanisms underlying the crossing of HLA barriers.
Bone Marrow Transplant
2014
, vol. 
49
 
7
(pg. 
873
-
879
)
8
Wang
 
Y
Liu
 
DH
Liu
 
KY
, et al. 
Long-term follow-up of haploidentical hematopoietic stem cell transplantation without in vitro T cell depletion for the treatment of leukemia: nine years of experience at a single center.
Cancer
2013
, vol. 
119
 
5
(pg. 
978
-
985
)
9
Huang
 
XJ
Zhu
 
HH
Chang
 
YJ
, et al. 
The superiority of haploidentical related stem cell transplantation over chemotherapy alone as postremission treatment for patients with intermediate- or high-risk acute myeloid leukemia in first complete remission.
Blood
2012
, vol. 
119
 
23
(pg. 
5584
-
5590
)
10
Luo
 
Y
Xiao
 
H
Lai
 
X
, et al. 
T-cell-replete haploidentical HSCT with low-dose anti-T-lymphocyte globulin compared with matched sibling HSCT and unrelated HSCT.
Blood
2014
, vol. 
124
 
17
(pg. 
2735
-
2743
)
11
Lu
 
DP
Dong
 
L
Wu
 
T
, et al. 
Conditioning including antithymocyte globulin followed by unmanipulated HLA-mismatched/haploidentical blood and marrow transplantation can achieve comparable outcomes with HLA-identical sibling transplantation.
Blood
2006
, vol. 
107
 
8
(pg. 
3065
-
3073
)
12
Members NAMLP. Acute myeloid leukemia. NCCN practice guidelines in oncology version 2.2011. http://www.docin.com/p-220085937.html. Accessed May 3, 2012
13
Breems
 
DA
Van Putten
 
WL
De Greef
 
GE
, et al. 
Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype.
J Clin Oncol
2008
, vol. 
26
 
29
(pg. 
4791
-
4797
)
14
Armand
 
P
Kim
 
HT
Zhang
 
MJ
, et al. 
Classifying cytogenetics in patients with acute myelogenous leukemia in complete remission undergoing allogeneic transplantation: a Center for International Blood and Marrow Transplant Research study.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
2
(pg. 
280
-
288
)
15
Zhao
 
XS
Jin
 
S
Zhu
 
HH
, et al. 
Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT.
Bone Marrow Transplant
2012
, vol. 
47
 
4
(pg. 
499
-
507
)
16
Zhao
 
XS
Yan
 
CH
Liu
 
DH
, et al. 
Combined use of WT1 and flow cytometry monitoring can promote sensitivity of predicting relapse after allogeneic HSCT without affecting specificity.
Ann Hematol
2013
, vol. 
92
 
8
(pg. 
1111
-
1119
)
17
Yan
 
CH
Liu
 
DH
Liu
 
KY
, et al. 
Risk stratification-directed donor lymphocyte infusion could reduce relapse of standard-risk acute leukemia patients after allogeneic hematopoietic stem cell transplantation.
Blood
2012
, vol. 
119
 
14
(pg. 
3256
-
3262
)
18
Yan
 
CH
Wang
 
JZ
Liu
 
DH
, et al. 
Chemotherapy followed by modified donor lymphocyte infusion as a treatment for relapsed acute leukemia after haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion: superior outcomes compared with chemotherapy alone and an analysis of prognostic factors.
Eur J Haematol
2013
, vol. 
91
 
4
(pg. 
304
-
314
)
19
Yanada
 
M
Emi
 
N
Naoe
 
T
, et al. 
Allogeneic myeloablative transplantation for patients aged 50 years and over.
Bone Marrow Transplant
2004
, vol. 
34
 
1
(pg. 
29
-
35
)
20
Craddock
 
C
Szydlo
 
RM
Klein
 
JP
, et al. 
Estimating leukemia-free survival after allografting for chronic myeloid leukemia: a new method that takes into account patients who relapse and are restored to complete remission.
Blood
2000
, vol. 
96
 
1
(pg. 
86
-
90
)
21
Brunstein
 
CG
Fuchs
 
EJ
Carter
 
SL
, et al. 
Blood and Marrow Transplant Clinical Trials Network
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
, vol. 
118
 
2
(pg. 
282
-
288
)
22
Wang
 
Y
Chang
 
YJ
Xu
 
LP
, et al. 
Who is the best donor for a related HLA haplotype-mismatched transplant?
Blood
2014
, vol. 
124
 
6
(pg. 
843
-
850
)
23
Bashey
 
A
Zhang
 
X
Sizemore
 
CA
, et al. 
T-cell-replete HLA-haploidentical hematopoietic transplantation for hematologic malignancies using post-transplantation cyclophosphamide results in outcomes equivalent to those of contemporaneous HLA-matched related and unrelated donor transplantation.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1310
-
1316
)
24
Lai
 
Y
Ma
 
J
Schwarzenberger
 
P
, et al. 
Combination of CsA, MTX and low-dose, short-course mycophenolate mofetil for GVHD prophylaxis.
Bone Marrow Transplant
2009
, vol. 
43
 
1
(pg. 
61
-
67
)
25
Lai
 
YR
Chen
 
YH
Hu
 
DM
, et al. 
Multicenter phase ii study of a combination of cyclosporine a, methotrexate and mycophenolate mofetil for GVHD prophylaxis: results of the Chinese Bone Marrow Transplant Cooperative Group (CBMTCG).
J Hematol Oncol
2014
, vol. 
7
 pg. 
59
 
26
Aversa
 
F
Reisner
 
Y
Martelli
 
MF
The haploidentical option for high-risk haematological malignancies.
Blood Cells Mol Dis
2008
, vol. 
40
 
1
(pg. 
8
-
12
)
27
Ciceri
 
F
Labopin
 
M
Aversa
 
F
, et al. 
Acute Leukemia Working Party (ALWP) of European Blood and Marrow Transplant (EBMT) Group
A survey of fully haploidentical hematopoietic stem cell transplantation in adults with high-risk acute leukemia: a risk factor analysis of outcomes for patients in remission at transplantation.
Blood
2008
, vol. 
112
 
9
(pg. 
3574
-
3581
)
28
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
, vol. 
23
 
15
(pg. 
3447
-
3454
)
29
Appelbaum
 
FR
Pursuing the goal of a donor for everyone in need.
N Engl J Med
2012
, vol. 
367
 
16
(pg. 
1555
-
1556
)
30
Chang
 
YJ
Huang
 
XJ
Haploidentical hematopoietic stem cell transplantation with unmanipulated granulocyte colony stimulating factor mobilized marrow and blood grafts.
Curr Opin Hematol
2012
, vol. 
19
 
6
(pg. 
454
-
461
)
31
Ringdén
 
O
Horowitz
 
MM
Gale
 
RP
, et al. 
Outcome after allogeneic bone marrow transplant for leukemia in older adults.
JAMA
1993
, vol. 
270
 
1
(pg. 
57
-
60
)
32
Huang
 
XJ
Liu
 
DH
Liu
 
KY
, et al. 
Haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion for the treatment of hematological malignancies [published correction appears in Bone Marrow Transplant. 2008;42(4):295].
Bone Marrow Transplant
2006
, vol. 
38
 
4
(pg. 
291
-
297
)
33
Farag
 
SS
Elder
 
PJ
Marcucci
 
G
, et al. 
Radiation-free regimens result in similar outcomes of allogeneic hematopoietic progenitor cell transplantation in patients aged ≥50 years compared to younger adults with low-risk disease.
Bone Marrow Transplant
2003
, vol. 
31
 
2
(pg. 
87
-
93
)
34
Lee
 
SJ
Klein
 
J
Haagenson
 
M
, et al. 
High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation.
Blood
2007
, vol. 
110
 
13
(pg. 
4576
-
4583
)
35
Saber
 
W
Opie
 
S
Rizzo
 
JD
Zhang
 
MJ
Horowitz
 
MM
Schriber
 
J
Outcomes after matched unrelated donor versus identical sibling hematopoietic cell transplantation in adults with acute myelogenous leukemia.
Blood
2012
, vol. 
119
 
17
(pg. 
3908
-
3916
)
36
Brunet
 
S
Labopin
 
M
Esteve
 
J
, et al. 
Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis.
J Clin Oncol
2012
, vol. 
30
 
7
(pg. 
735
-
741
)
37
Huang
 
XJ
Zhao
 
XY
Liu
 
DH
Liu
 
KY
Xu
 
LP
Deleterious effects of KIR ligand incompatibility on clinical outcomes in haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion.
Leukemia
2007
, vol. 
21
 
4
(pg. 
848
-
851
)
38
Zhao
 
XY
Chang
 
YJ
Huang
 
XJ
Conflicting impact of alloreactive NK cells on transplantation outcomes after haploidentical transplantation: do the reconstitution kinetics of natural killer cells create these differences?
Biol Blood Marrow Transplant
2011
, vol. 
17
 
10
(pg. 
1436
-
1442
)

Author notes

Y.W. and Q.-F.L. contributed equally to this study.

Sign in via your Institution