Purpose:

The anti-leukemic activity of allogeneic hematopoietic cell transplantation (HCT) depends on both the intensity of conditioning regimen and the strength of the graft-versus-leukemia (GVL) effect. However, it is unclear whether the sensitivity of the GVL effects differs between donor type and graft source.

Experimental Design:

We retrospectively evaluated the effect of acute and chronic graft-versus-host disease (GVHD) on transplant outcomes for adults with acute leukemia (n = 6,548) between 2007 and 2017 using a Japanese database. In all analyses, we separately evaluated three distinct cohorts based on donor type [(8/8 allele-matched sibling donor, 8/8 allele-matched unrelated donor, and unrelated single-cord blood (UCB)].

Results:

The multivariate analysis, in which the development of GVHD was treated as a time-dependent covariate, showed a reductive effect of grade I–II acute GVHD on treatment failure (defined as 1-leukemia-free survival; P < 0.001), overall mortality (OM; P < 0.001), relapse (P < 0.001), and non-relapse mortality (NRM; P < 0.001) in patients receiving from UCB. A reductive effect of limited chronic GVHD on treatment failure (P < 0.001), OM (P < 0.001), and NRM (P < 0.001) was also shown in patients receiving from UCB. However, these effects were not always shown in patients receiving from other donors. The beneficial effects of mild acute and chronic GVHD after UCB transplantation on treatment failure were noted relatively in subgroups of patients with acute myelogenous leukemia and a non-remission status.

Conclusions:

These data suggested that the development of mild GVHD could improve survival after UCB transplantation for acute leukemia.

Translational Relevance

To clarify the different impact of graft-versus-host disease (GVHD) on transplant outcomes across donor types, we retrospectively evaluated the effect of acute and chronic GVHD on survival and relapse for adults with acute leukemia (n = 6,548) between 2007 and 2017 using a nationwide Japanese database. Our study demonstrated that the reductive effect of grade I–II acute GVHD on mortality and relapse was shown in patients receiving from unrelated single-cord blood (UCB), but not matched sibling donors (MSD) or matched unrelated donors (MUD). The reductive effect of limited chronic GVHD on mortality was also shown in patients receiving from UCB, but not MSD or MUD. This could be the explanation for the fact that the development of mild GVHD could improve survival after UCB transplantation for acute leukemia.

Allogeneic hematopoietic cell transplantation (HCT) is a potentially curative therapy for adults with acute leukemia. The anti-leukemic activity of allogeneic HCT depends on both the intensity of conditioning regimen and the strength of the graft-versus-leukemia (GVL) effect. It is difficult to evaluate independently the meaningful GVL effect because this effect may occur without the occurrence of clinical graft-versus-host disease (GVHD). Indeed, the GVL effect is based on the phenomenon that patients with GVHD had a lower risk of leukemia relapse compared with patients without GVHD in previous studies (1, 2). Although the incidence and severity of acute and chronic GVHD are associated with donor type and graft source (3–6), the strength of GVL effects between donor type and graft source is unclear.

Several studies have suggested that the GVL effect might be enhanced by HLA-mismatched HCT, such as cord blood transplantation (CBT; refs. 7–10). CBT has unique characteristics, such as a lower incidence and severity of GVHD but does not compromise the GVL effects in spite of less stringent criteria for HLA matching (3–6). Indeed, recent studies have shown that relapse rate was significantly lower after CBT than HCT from matched sibling donors (MSD) or matched unrelated donors (MUD; refs. 3, 11). Therefore, the sensitivity of the GVL effects may differ between donor type and graft source. To clarify the effect of varying severities of acute and chronic GVHD on transplant outcomes, we performed a retrospective analysis using a nationwide Japanese database for a large cohort of adult patients with acute leukemia treated with allogeneic HCT from 8/8 allele-MSD, 8/8 allele-MUD, or unrelated cord blood (UCB).

Data collection

This retrospective study was performed by the Donor/Source Working Group, the Adult Acute Myelogenous Leukemia (AML) Working Group, and the Adult Acute Lymphoblastic Leukemia (ALL) Working Group of the Japan Society for HCT (JSHCT). The clinical data were obtained by the Transplant Registry Unified Management Program (TRUMP) of the Japanese Data Center for HCT (JDCHCT), covering almost all (>300) HCT centers in Japan. Each HCT center is required to report annual follow-up data of consecutive HCTs (12, 13). The inclusion criteria consisted of patients' ages between 16 and 65 years with AML and ALL who received their first allogeneic HCT from MSD, MUD, or single-unit UCB between 2007 and 2017 in Japan, who achieved neutrophil engraftment, and who survived at least 60 days without relapse. We excluded patients that lacked data of survival and GVHD status and those who were conditioned with antithymocyte globulin (ATG). Finally, 6,548 patients were eligible for this study. This retrospective study was performed after approval by the institutional review board of the Institute of Medical Science, The University of Tokyo, where this study was carried out (30–71-B0128), and was conducted in accordance with the Declaration of Helsinki. Written informed consent was obtained from each patient.

Objectives

The primary objective of this study was to clarify the effect of acute and chronic GVHD on leukemia-free survival (LFS) for each donor type. Secondary objectives were to clarify the effect of acute and chronic GVHD on overall survival (OS), hematological relapse, and non-relapse mortality (NRM) for each donor type.

Definitions

The diagnosis and severity of acute and chronic GVHD were assessed by the treating physicians at each center according to consensus criteria (14, 15). The degree of HLA matching was based on allele levels for HLA-A, HLA-B, HLA-C, and HLA-DRB1 loci for HCT from MSD and MUD or antigenic levels for HLA-A, HLA-B, and HLA-DR loci HCT from single-unit cord blood. Myeloablative conditioning (MAC) was defined as total body irradiation with fractionated doses totaling ≥8 Gy, oral busulfan (BU) doses of ≥9 mg/kg, or intravenous BU doses of ≥7.2 mg/kg (16). Poor-risk cytogenetics was defined according to the National Comprehensive Cancer Network Guidelines for AML (17). Poor-risk cytogenetics for ALL included t(9;22), t(4;11), t(8;14), t(14;18), hypodiploid, and complex karyotypes according to the criteria described previously (18). LFS (inverse of treatment failure) was defined as the time from the date of HCT until death, relapse, or survival. OS (inverse of overall mortality) was defined as the time from the date of HCT until death or survival. Relapse was defined as hematological leukemia recurrence. NRM was defined as death without leukemia recurrence after HCT.

Statistical analysis

Patients, diseases, and transplant characteristics between groups were compared using a χ2 test or Fisher's exact test for categorical variables and the Kruskal–Wallis test for continuous variables. The probabilities of LFS and OS were calculated using the Kaplan–Meier method, with a conditional landmark analysis, which could be used to remove guarantee-time bias (19). The probabilities of relapse and NRM were calculated on the basis of cumulative incidence curves to accommodate competing risks, with a conditional landmark analysis. The competing risk for relapse was NRM, whereas the competing risk for NRM was relapse. Univariate analyses were performed using the log-rank test for OS and LFS and Gray's test for relapse and NRM.

The Cox proportional hazards regression model was used to estimate hazard ratios (HR) with a 95% confidence interval (CI) for treatment failure (1-LFS), overall mortality (1-OS), relapse, and NRM in the multivariate analyses, by treating the development of GVHD as a time-dependent covariate. To evaluate the HRs for relapse and NRM, patients who experienced NRM or relapse were censored. When evaluating the effect of GVHD on transplant outcomes, acute GVHD and chronic GVHD were separately analyzed. The effect of acute GVHD was evaluated by comparing no acute GVHD versus grade I–II acute GVHD versus grade III–IV acute GVHD for patients who survived at least 60 days without relapse. The effect of chronic GVHD was evaluated by comparing no chronic GVHD versus limited chronic GVHD versus extensive chronic GVHD for patients who survived at least 100 days without relapse. The following variables other than GVHD were considered in the multivariate analysis: age (16–49 years vs. 50–65 years), sex (male vs. female), performance status (0–1 vs. 2–4), disease (AML vs. ALL), cytogenetics (other than poor vs. poor), disease status at HCT (CR vs. non-CR), conditioning regimen (MAC vs. RIC), GVHD prophylaxis (calcineurin inhibitors plus methotrexate vs. others), and transplant year (2007–2012 vs. 2013–2017). When evaluating the effect of chronic GVHD on transplant outcomes, the prior history of acute GVHD (none vs. grade I–II vs. grade III–IV acute GVHD) was included in the multivariate analysis. To adjust for multiple testing for each outcome in univariate and multivariate analysis, P < 0.0166 (0.05/3) was considered statistically considered with the Bonfferoni correction, and P < 0.0041 (0.0166/4) was considered statistically considered for subgroup analysis. P values between 0.0166 or 0.0041 and 0.05 were considered to have a marginal significance.

In all these analyses, we separately evaluated three distinct cohorts based on donor type (MSD, MUD, and UCB). All P values were two-sided, and the data were analyzed by EZR (Saitama Medical Center, Jichi Medical University, Saitama, Japan; ref. 20), a graphical user interface for the R 4.0.4 software program (R Foundation for Statistical Computing).

Characteristics of patients, diseases, and transplant procedures

The characteristics of patients, diseases, and transplant procedures are summarized in Table 1. Among 6,548 patients, 1,322, 2,429, and 2,797 patients received allogeneic HCT from MSD, MUD, and UCB, respectively. The median age at HCT was 42 years, 47 years, and 49 years in MSD, MUD, and UCB recipients, respectively (P < 0.001). The proportions of recipient cytomegalovirus seropositive status (P = 0.004), HCT-CI≥3 (P < 0.001), and PS≥2 (P < 0.001) were different among donor types. MUD transplants were less frequently performed in recent years (P < 0.001). A higher proportion of UCB recipients received RIC regimen (P < 0.001), and calcineurin inhibitors+mycophenolate mofetil-based GVHD prophylaxis (P < 0.001). UCB recipients were more likely to have AML (P < 0.001) and a non-CR status (P < 0.001).

Table 1.

Characteristics of patient, disease, and transplantation according to donor type.

Overall cohortMSDMUDUCBP value OverallP value MSD vs. MUDP value MSD vs. UCBP value MUD vs. UCB
Number of patients 6,548 1,322 2,429 2,797     
Recipient age, median (IQR) 47 (35–57) 42 (32–53) 47 (36–57) 49 (36–59) <0.001 <0.001 <0.001 0.003 
Recipient sex     0.26 0.97 0.24 0.14 
 Male 3,625 (55.4) 743 (56.2) 1,366 (56.3) 1,516 (54.2)     
 Female 2,921 (44.6) 579 (43.8) 1,062 (43.7) 1,280 (45.8)     
 Missing 2 (<0.1) 1 (<0.1) 1 (<0.1)     
Recipient CMV serostatus     0.004 0.10 <0.001 0.14 
 Positive 5,018 (76.6) 977 (73.9) 1,854 (76.3) 2,187 (78.2)     
 Negative 1,252 (19.1) 295 (22.3) 472 (19.4) 485 (17.3)     
 Missing 278 (4.2) 50 (3.8) 103 (4.2) 125 (4.5)     
HCT-CI     <0.001 0.10 <0.001 <0.001 
 0–2 5,607 (85.1) 1,183 (89.5) 2,116 (87.1) 2,308 (82.5)     
 ≥3 716 (11.3) 107 (8.1) 242 (10.0) 367 (13.1)     
 Missing 225 (3.6) 32 (2.4) 71 (2.9) 122 (4.4)     
PS     <0.001 0.42 <0.001 <0.001 
 0–1 6,124 (91.9) 1,252 (94.7) 2,316 (95.3) 2,556 (91.4)     
 ≥2 418 (7.9) 69 (5.2) 112 (4.6) 237 (8.5)     
 Missing 6 (0.1) 1 (<0.1) 1 (<0.1) 4 (0.1)     
Transplant year     <0.001 <0.001 0.23 <0.001 
 2007–2012 2,948 (45.0) 549 (41.5) 1,182 (48.7) 1,217 (43.5)     
 2013–2017 3,600 (55.0) 773 (58.5) 1247 (51.3) 1,580 (56.5)     
Conditioning regimen     <0.001 0.13 <0.001 <0.001 
 MAC 4,975 (76.0) 1,055 (79.8) 1,886 (77.6) 2,034 (72.7)     
 RIC 1,573 (24.0) 267 (20.2) 543 (22.4) 763 (27.3)     
GVHD prophylaxis     <0.001 0.031 <0.001 <0.001 
 CI+MTX 5,230 (79.9) 1,224 (92.6) 2,291 (94.3) 1,715 (61.3)     
 CI+MMF 908 (13.9) 38 (2.9) 68 (2.8) 802 (28.7)     
 Others 410 (6.3) 60 (4.5) 70 (2.9) 280 (10.0)     
Disease     <0.001 0.056 <0.001 <0.001 
 AML 4,521 (69.0) 846 (64.0) 1,630 (67.1) 2,045 (73.1)     
 ALL 2,027 (31.0) 476 (36.0) 799 (32.9) 752 (26.9)     
Disease status     <0.001 0.86 <0.001 <0.001 
 CR 4,622 (70.6) 1,033 (78.1) 1,904 (78.4) 1,685 (60.2)     
 Non-CR 1,926 (29.4) 289 (21.9) 525 (21.6) 1,112 (39.8)     
Cytogenetic risk     0.62 0.67 0.74 0.33 
 Poor 1,846 (28.2) 373 (28.2) 669 (27.5) 804 (28.7)     
 Other than poor 4,702 (71.8) 949 (71.8) 1,760 (72.5) 1,993 (71.3)     
Acute GVHD by 100 days     <0.001 <0.001 <0.001 <0.001 
 None 2,248 (38.4) 562 (47.1) 863 (39.5) 823 (33.2)     
 Grade I-II 3,037 (51.9) 529 (44.3) 1,146 (52.4) 1,362 (55.0)     
 Grade III-IV 572 (9.8) 102 (8.5) 177 (8.1) 293 (11.8)     
Chronic GVHD by 129 days     0.003 0.40 0.021 0.003 
 None 4,445 (79.8) 903 (79.8) 1,703 (81.4) 1,839 (78.3)     
 Limited 570 (10.2) 103 (9.1) 185 (8.8) 282 (12.0)     
 Extensive 557 (10.0) 126 (11.1) 203 (9.7) 228 (9.7)     
Overall cohortMSDMUDUCBP value OverallP value MSD vs. MUDP value MSD vs. UCBP value MUD vs. UCB
Number of patients 6,548 1,322 2,429 2,797     
Recipient age, median (IQR) 47 (35–57) 42 (32–53) 47 (36–57) 49 (36–59) <0.001 <0.001 <0.001 0.003 
Recipient sex     0.26 0.97 0.24 0.14 
 Male 3,625 (55.4) 743 (56.2) 1,366 (56.3) 1,516 (54.2)     
 Female 2,921 (44.6) 579 (43.8) 1,062 (43.7) 1,280 (45.8)     
 Missing 2 (<0.1) 1 (<0.1) 1 (<0.1)     
Recipient CMV serostatus     0.004 0.10 <0.001 0.14 
 Positive 5,018 (76.6) 977 (73.9) 1,854 (76.3) 2,187 (78.2)     
 Negative 1,252 (19.1) 295 (22.3) 472 (19.4) 485 (17.3)     
 Missing 278 (4.2) 50 (3.8) 103 (4.2) 125 (4.5)     
HCT-CI     <0.001 0.10 <0.001 <0.001 
 0–2 5,607 (85.1) 1,183 (89.5) 2,116 (87.1) 2,308 (82.5)     
 ≥3 716 (11.3) 107 (8.1) 242 (10.0) 367 (13.1)     
 Missing 225 (3.6) 32 (2.4) 71 (2.9) 122 (4.4)     
PS     <0.001 0.42 <0.001 <0.001 
 0–1 6,124 (91.9) 1,252 (94.7) 2,316 (95.3) 2,556 (91.4)     
 ≥2 418 (7.9) 69 (5.2) 112 (4.6) 237 (8.5)     
 Missing 6 (0.1) 1 (<0.1) 1 (<0.1) 4 (0.1)     
Transplant year     <0.001 <0.001 0.23 <0.001 
 2007–2012 2,948 (45.0) 549 (41.5) 1,182 (48.7) 1,217 (43.5)     
 2013–2017 3,600 (55.0) 773 (58.5) 1247 (51.3) 1,580 (56.5)     
Conditioning regimen     <0.001 0.13 <0.001 <0.001 
 MAC 4,975 (76.0) 1,055 (79.8) 1,886 (77.6) 2,034 (72.7)     
 RIC 1,573 (24.0) 267 (20.2) 543 (22.4) 763 (27.3)     
GVHD prophylaxis     <0.001 0.031 <0.001 <0.001 
 CI+MTX 5,230 (79.9) 1,224 (92.6) 2,291 (94.3) 1,715 (61.3)     
 CI+MMF 908 (13.9) 38 (2.9) 68 (2.8) 802 (28.7)     
 Others 410 (6.3) 60 (4.5) 70 (2.9) 280 (10.0)     
Disease     <0.001 0.056 <0.001 <0.001 
 AML 4,521 (69.0) 846 (64.0) 1,630 (67.1) 2,045 (73.1)     
 ALL 2,027 (31.0) 476 (36.0) 799 (32.9) 752 (26.9)     
Disease status     <0.001 0.86 <0.001 <0.001 
 CR 4,622 (70.6) 1,033 (78.1) 1,904 (78.4) 1,685 (60.2)     
 Non-CR 1,926 (29.4) 289 (21.9) 525 (21.6) 1,112 (39.8)     
Cytogenetic risk     0.62 0.67 0.74 0.33 
 Poor 1,846 (28.2) 373 (28.2) 669 (27.5) 804 (28.7)     
 Other than poor 4,702 (71.8) 949 (71.8) 1,760 (72.5) 1,993 (71.3)     
Acute GVHD by 100 days     <0.001 <0.001 <0.001 <0.001 
 None 2,248 (38.4) 562 (47.1) 863 (39.5) 823 (33.2)     
 Grade I-II 3,037 (51.9) 529 (44.3) 1,146 (52.4) 1,362 (55.0)     
 Grade III-IV 572 (9.8) 102 (8.5) 177 (8.1) 293 (11.8)     
Chronic GVHD by 129 days     0.003 0.40 0.021 0.003 
 None 4,445 (79.8) 903 (79.8) 1,703 (81.4) 1,839 (78.3)     
 Limited 570 (10.2) 103 (9.1) 185 (8.8) 282 (12.0)     
 Extensive 557 (10.0) 126 (11.1) 203 (9.7) 228 (9.7)     

Note: The P values in bold are statistically significant among donor type (<0.05).

Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CI, calcineurin inhibitor; CMV, cytomegalovirus; CR, complete remission; GVHD, graft-versus-host disease; HCT-CI, hematopoietic cell transplantation-specific comorbidity index; IQR, interquartile range; MAC, myeloablative conditioning; MMF, mycophenolate mofetil; MSD, matched sibling donor; MTX, methotrexate; MUD, matched unrelated donor; PS, performance status; RIC, reduced-intensity conditioning; UCB, unrelated cord blood.

Transplant outcomes

In the entire cohort, the median follow-up period for survivors after HCT was 41 months (range, 2–138 months). The probability of LFS at 3 years was 55% (95% CI, 52%–58%), 58% (95% CI, 56%–60%), and 53% (95% CI, 51%–55%) after HCT from MSD, MUD, and UCB, respectively (Supplementary Fig. S1A). In the multivariate analysis, the hazard risk of treatment failure was significantly lower in MUD (HR, 0.88; P = 0.022) or UCB (HR, 0.86; P = 0.007) recipients compared with MSD recipients (Supplementary Table S1). The probability of OS at 3 years was 63% (95% CI, 60%–66%), 64% (95% CI, 62%–66%), and 58% (95% CI, 56%–60%) after HCT from MSD, MUD, and UCB, respectively (Supplementary Fig. S1B). In the multivariate analysis, there was no significant difference in overall mortality between MSD recipients and both MUD (HR, 0.94; P = 0.30) and UCB (HR, 0.96; P = 0.53) recipients (Supplementary Table S1).

The cumulative incidence of relapse at 3 years was 34% (95% CI, 31%–37%), 28% (95% CI, 26%–30%), and 30% (95% CI, 29%–32%) after HCT from MSD, MUD, and UCB, respectively (Supplementary Fig. S1C). In the multivariate analysis, the hazard risk of relapse was significantly lower in MUD (HR, 0.79; P < 0.001) or UCB (HR, 0.80; P < 0.001) recipients compared with MSD recipients (Supplementary Table S1). The cumulative incidence of NRM at 3 years was 11% (95% CI, 9%–13%), 15% (95% CI, 13%–16%), and 17% (95% CI, 15%–18%) after HCT from MSD, MUD, and UCB, respectively (Supplementary Fig. S1D). In the multivariate analysis, there was no significant difference in NRM between MSD recipients and both MUD (HR, 1.18; P = 0.07) and UCB (HR, 1.10; P = 0.31) recipients (Supplementary Table S1).

Effect of acute GVHD on transplant outcomes

In the univariate analysis with a conditional landmark analysis at 100 days, the probability of LFS was significantly lower in patients who developed grade III–IV acute GVHD compared with those who did not develop acute GVHD after HCT from MUD (P < 0.001; Fig. 1AC). The probability of OS was significantly lower in patients who developed grade III–IV acute GVHD compared with those who did not develop acute GVHD after HCT from MSD (P = 0.011), MUD (P < 0.001), and UCB (P < 0.001; Fig. 1D–F). Interestingly, the probabilities of LFS and OS were significantly higher in patients who developed grade I–II acute GVHD compared with those who did not develop acute GVHD only after HCT from UCB (P < 0.001 for LFS, P < 0.001 for OS; Fig. 1C and F).

Figure 1.

The effect of acute GVHD on leukemia-free survival (A–C) and overall survival (D–F) according to donor type. Kaplan–Meier survival curves are plotted with a conditional landmark analysis at 100 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of acute GVHD on treatment failure (G) and overall mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, and transplant year. The P values in bold are statistically significant (P < 0.0166).

Figure 1.

The effect of acute GVHD on leukemia-free survival (A–C) and overall survival (D–F) according to donor type. Kaplan–Meier survival curves are plotted with a conditional landmark analysis at 100 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of acute GVHD on treatment failure (G) and overall mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, and transplant year. The P values in bold are statistically significant (P < 0.0166).

Close modal

In the multivariate analysis, when no acute GVHD was used as the reference group among each donor type, development of grade I–II acute GVHD was significantly associated with lower treatment failure (P < 0.001) and overall mortality (P < 0.001) in patients after HCT from UCB but not in other donor types. The development of grade III–IV acute GVHD was significantly associated with higher treatment failure in patients after HCT from MUD (P < 0.001), and higher overall mortality in patients after HCT from MUD (P < 0.001), and UCB (P < 0.001; Fig. 1G and H).

In the univariate analysis with a conditional landmark analysis at 100 days, the cumulative incidence of relapse was significantly lower in patients who developed grade III–IV acute GVHD compared with those who did not develop acute GVHD after HCT from UCB (P < 0.001), but not MSD and MUD (Fig. 2A–C). The cumulative incidence of NRM was significantly higher in patients who developed grade III–IV acute GVHD compared with those who did not develop acute GVHD for each donor type (P < 0.001; Fig. 2D–F). Interestingly, the cumulative incidence of relapse (P < 0.001) was significantly lower in patients who developed grade I–II acute GVHD compared with those who did not develop acute GVHD only in patients after HCT from UCB (Fig. 2C).

Figure 2.

The effect of acute GVHD on relapse (A–C) and non-relapse mortality (D–F) according to donor type. Cumulative incidence curves are plotted with a conditional landmark analysis at 100 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of acute GVHD on relapse (G) and non-relapse mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, and transplant year. The P values in bold are statistically significant (P < 0.0166).

Figure 2.

The effect of acute GVHD on relapse (A–C) and non-relapse mortality (D–F) according to donor type. Cumulative incidence curves are plotted with a conditional landmark analysis at 100 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of acute GVHD on relapse (G) and non-relapse mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, and transplant year. The P values in bold are statistically significant (P < 0.0166).

Close modal

In the multivariate analysis, when no acute GVHD was used as the reference group among each donor type, development of grade III–IV acute GVHD was associated with lower relapse in patients after HCT from MSD (P = 0.007), and UCB (P < 0.001). The development of grade III–IV acute GVHD was significantly associated with higher NRM in patients after HCT from MSD (P < 0.001), MUD (P < 0.001), and UCB (P < 0.001). Interestingly, development of grade I–II acute GVHD was also associated with significant lower relapse (P < 0.001) and NRM (P < 0.001) only in patients after HCT from UCB (Fig. 2G and H).

We also separately evaluated the effect of grade I and II acute GVHD on transplant outcomes in multivariate analysis using acute GVHD as time-dependent covariates. When no acute GVHD was used as the reference group among each donor type, development of grade I and II acute GVHD was also significantly associated with lower treatment failure, overall mortality, relapse, and NRM in patients after HCT from UCB but not in other donor types (Supplementary Fig. S2).

The causes of death according to the severity of acute GVHD for each donor type are summarized in Supplementary Table S2A. The most common cause of death was relapse in patients who did not develop acute GVHD and did develop grade I–II acute GVHD for each donor type. Only among UCB recipients, relapse (P = 0.002) and pulmonary complications (P = 0.011) were a less common cause of death in patients who developed grade I–II acute GVHD compared with those who did not develop acute GVHD. GVHD was a more common cause of death in patients who developed grade III–IV acute GVHD compared with those who did not develop acute GVHD, regardless of the donor type.

Effects of chronic GVHD on transplant outcomes

In the univariate analysis with a conditional landmark analysis at 129 days, which was the median time of development of chronic GVHD, the probabilities of LFS and OS were significantly lower in patients who developed extensive chronic GVHD compared with those who did not develop chronic GVHD after HCT from MSD (P = 0.011 for LFS, P = 0.001 for OS) and MUD (P = 0.002 for LFS, P < 0.001 for OS; Fig. 3A, B, D, and E). In contrast, the probabilities of LFS and OS were significantly higher in patients who developed limited chronic GVHD compared with those who did not develop chronic GVHD after HCT from UCB (P = 0.010 for LFS, P = 0.001 for OS; Fig. 3C and F).

Figure 3.

The effect of chronic GVHD on leukemia-free survival (A–C) and overall survival (D–F) according to donor type. Kaplan–Meier survival curves are plotted with a conditional landmark analysis at 129 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of chronic GVHD on treatment failure (G) and overall mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, transplant year, and prior history of acute GVHD. The P values in bold are statistically significant (P < 0.0166).

Figure 3.

The effect of chronic GVHD on leukemia-free survival (A–C) and overall survival (D–F) according to donor type. Kaplan–Meier survival curves are plotted with a conditional landmark analysis at 129 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of chronic GVHD on treatment failure (G) and overall mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, transplant year, and prior history of acute GVHD. The P values in bold are statistically significant (P < 0.0166).

Close modal

In the multivariate analysis, when no chronic GVHD was used as the reference group among each donor type, development of limited chronic GVHD was significantly associated with lower treatment failure (P < 0.001) and overall mortality (P < 0.001) only in patients after HCT from UCB (Fig. 3G and H).

In the univariate analysis with a conditional landmark analysis at 129 days, there were no significant differences of relapse incidence in patients who developed limited or extensive chronic GVHD compared with those who did not develop chronic GVHD for each donor type (Fig. 4A–C). Meanwhile, the cumulative incidence of NRM was significantly higher in patients who developed extensive chronic GVHD compared with those who did not develop chronic GVHD for each donor type (P < 0.001 for MSD, MUD, UCB; Fig. 4D–F). Interestingly, the cumulative incidence of NRM was significantly lower in patients who developed limited chronic GVHD compared with those who did not develop chronic GVHD only in patients after HCT from UCB (P < 0.001; Fig. 4F).

Figure 4.

The effect of chronic GVHD on relapse (A–C) and non-relapse mortality (D–F) according to donor type. Cumulative incidence curves are plotted with a conditional landmark analysis at 129 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of chronic GVHD on relapse (G) and non-relapse mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, transplant year, and prior history of acute GVHD. The P values in bold are statistically significant (P < 0.0166).

Figure 4.

The effect of chronic GVHD on relapse (A–C) and non-relapse mortality (D–F) according to donor type. Cumulative incidence curves are plotted with a conditional landmark analysis at 129 days after HCT. Forest plots for the adjusted hazard ratios and 95% confidence intervals of severity of chronic GVHD on relapse (G) and non-relapse mortality (H) in the multivariate analysis. All covariates other than GVHD included in multivariate models were age, sex, performance status, disease, cytogenetics, disease status at HCT, conditioning regimen, GVHD prophylaxis, transplant year, and prior history of acute GVHD. The P values in bold are statistically significant (P < 0.0166).

Close modal

In the multivariate analysis, when no chronic GVHD was used as the reference group for each donor type, development of extensive chronic GVHD was associated with a significant lower relapse in patients after HCT from MUD (P < 0.001) and a lower relapse but with marginal significance in patients after HCT from MSD (P = 0.016). The development of extensive chronic GVHD was also significantly associated with higher NRM in patients after HCT from MSD (P = 0.011), MUD (P < 0.001), and UCB (P = 0.002). Interestingly, development of limited chronic GVHD was associated with a significant lower NRM (P < 0.001) and a lower relapse but with marginal significance (P = 0.021) only in patients after HCT from UCB (Fig. 4G and H).

The causes of death according to the severity of chronic GVHD for each donor type are summarized in Supplementary Table S2B. The most common cause of death was relapse in patients who did not develop chronic GVHD and did develop limited or extensive chronic GVHD for each donor type. Relapse was a less common cause of death in patients who developed limited chronic GVHD compared with those who did not develop chronic GVHD in MUD (P = 0.011) and UCB (P = 0.017) recipients. GVHD was a more common cause of death in patients who developed extensive chronic GVHD compared with those who did not develop chronic GVHD, regardless of the donor type.

Effect of GVHD on transplant outcomes according to disease type and status

We also evaluated the effect of GVHD on transplant outcomes stratified according to disease type and status, which could affect the effects of GVHD on transplant outcomes, for each donor type.

The reductive effects of grade I–II acute GVHD on treatment failure and overall mortality after UCB transplantation were commonly noted in almost all subgroups of patients with AML and ALL, and those in CR and with non-CR status at HCT. However, the reductive effects of grade I–II acute GVHD on NRM after UCB transplantation were noted only in subgroups of patients with AML. The reductive effect of grade I–II acute GVHD on relapse after UCB transplantation was noted in subgroups of patients with AML and ALL, and those in CR status (Supplementary Fig. S3).

The reductive effects of limited chronic GVHD on treatment failure and NRM after UCB transplantation were noted in some subgroups of patients with AML and those with non-CR status at HCT, but not in those with ALL and with CR status. The reductive effects of limited chronic GVHD on overall mortality after UCB transplantation were noted in almost all subgroups of patients with AML and those in CR and with non-CR status at HCT, but not in those with ALL. The reductive effects of extensive chronic GVHD on relapse were noted in patients with CR status after MUD transplantation (Supplementary Fig. S4).

Effect of HLA disparity and GVHD prophylaxis regimen in CBT

Finally, we evaluated whether HLA disparity and GVHD prophylaxis regimen affected the beneficial effects of mild GVHD on transplant outcomes after CBT. The beneficial effects of grade I–II acute GVHD on LFS, OS, and relapse were noted only in the HLA 4/6–matched group, but not in the HLA 5–6/6–matched group (Supplementary Fig. S5A–S5D). The beneficial effects of limited chronic GVHD on LFS, OS, and NRM were also noted only in the HLA 4/6–matched group, but not in the HLA 5–6/6–matched group (Supplementary Fig. S5E–S5H). GVHD prophylaxis regimens did not affect the beneficial effects of mild GVHD on outcomes after CBT (Supplementary Fig. S6).

We evaluated the differential GVL effect associated with GVHD in acute leukemia based on donor types in a large cohort from a nationwide Japanese database. Our data showed that the reductive effects of grade I–II acute GVHD and limited chronic GVHD on treatment failure and mortality were commonly noted in patients receiving from UCB. Interestingly, the reductions of treatment failure and overall mortality were due not only to the reduction of relapse rate, but also the reduction of NRM in patients receiving from UCB. These data suggested that the development of mild GVHD could improve survival after CBT for acute leukemia.

Recent retrospective studies have demonstrated that relapse rate is similar between different donor types, such as MSD, 8/8MUD, and UCB in patients with acute leukemia (3–6). Although the incidence and severity of GVHD could be independent of donor type, the strength of GVT effects between donor type is limited (21–23). Ringdén and colleagues (21) demonstrated that the GVL effect from HLA-identical siblings is similar to those from MUDs for acute leukemia patients undergoing allogeneic HCT. Our study clearly demonstrated that a survival benefit of the GVL effect was commonly present only in CBT patients with mild GVHD, which is consistent with a previous report published by the JSHCT GVHD working group (24). Their report showed that the development of mild acute and chronic GVHD reduced the relapse rate after single CBT in previous cohort of adults with acute leukemia and myelodysplastic syndrome from 2000 to 2012 (24). All these data suggest that the GVL effects after CBT might be stronger than those after HCT from MSD or MUD. Although the exact mechanism of stronger GVL effects after CBT is unclear, this might be partly due to the enhancement of GVL effect by HLA-mismatched donor transplantation (25), because our data also demonstrated that the reductive effect of grade I–II acute GVHD on relapse was noted only in the HLA 4/6–matched group. Indeed, lower relapse rates after CBT were associated with an increased number of HLA mismatches (7, 8) and a specific HLA locus mismatch (9, 10), suggesting that the GVL effect after CBT is an attractive concept for acute leukemia.

The strength of the GVT effects associated with GVHD is also dependent on disease type (18, 26–33). Although some studies showed that ALL was more sensitive to GVT effects associated with GVHD than AML (26, 30, 33), several studies showed that AML was more sensitive to GVHD than ALL (28, 29). Meanwhile, the strength of the GVT effects according to disease burden at HCT is controversial (21, 28, 33–35). An early study showed that the reductive effect of GVHD on relapse was stronger in patients with advanced leukemia compared with those in CR (33), whereas recent reports have shown that the GVL effect was not present in advanced leukemia (21, 34). Indeed, our study showed that the reductive effect of mild acute GVHD on relapse was noted only in CBT patients with AML and ALL, and those in CR status. The marginal reduction of relapse by the development of mild chronic GVHD was noted only in CBT patients with AML. Importantly, the survival benefit of GVL effects was present only in CBT patients with mild GVHD. Recently, Milano and colleagues (3), reported a significant reduction of relapse after CBT but only in minimal residual disease (MRD)–positive groups compared with HCT from matched or mismatched unrelated donors, suggesting that the GVL effects after CBT might be induced by MRD-positive status. However, we included various methods to detect MRD at HCT from our registry-data but could not evaluate the association between MRD status and GVL effects. Therefore, the association between MRD status and stronger GVL effect after CBT is a future matter of investigation.

Our study had several limitations. One limitation was the study was based on a retrospective registry-based data in Japan. Therefore, there could have been a selection bias and unavailable data, such as a mutation profile for leukemia (36, 37), HLA-DPB1 mismatch (10, 25), killer immunoglobulin-like receptors (KIR) ligand mismatch (38), and post-transplant maintenance therapy (39, 40), which could have affected the GVL effects or relapse rate after HCT. Second, the development of acute and chronic GVHD frequently overlapped, and the treatment intervention and response for acute and chronic GVHD could not be considered to have evaluated the GVL effects. Therefore, it was difficult to clarify the relevant effects of acute and chronic GVHD on relapse in our study. In addition, a beneficial effect of mild acute GVHD has been controversial. Previous studies showed a positive impact of grade I acute GVHD (28) and grade II acute GVHD (41, 42), but a negative impact of grade II acute GVHD (29). In our study, a beneficial effect of mild acute GVHD was noted with patients who developed each grade I and II acute GVHD only after HCT from UCB, as well as combined grade I–II acute GVHD, which was consistent with previous reports (18, 24). Third, the data for single-unit CBT alone were included in our study, because double-unit CBT was not approved for use outside of clinical trials during the study period in Japan. Several retrospective studies have shown that double-unit CBT mediated stronger GVL effects than single-unit CBT (43, 44), but this effect was not confirmed in prospective studies (45, 46). Moreover, the beneficial effects of survival on mild acute and chronic GVHD after CBT are independent of not only the lower relapse but also lower NRM, which is consistent with a previous report in Japan (24, 47). Although the exact mechanism of the reductive effect of mild GVHD on NRM after CBT is unclear, we speculate that development of mild GVHD may affect delayed or impaired immune reconstitution after CBT without excess of adverse effects, which could contribute to lower incidence of NRM. Fourth, recent studies have shown that the GVL effect from haploidentical donors is similar or superior to those from HLA-identical siblings for patients with acute leukemia undergoing allogeneic HCT (22, 23). However, haploidentical HCT data were not included in our study, due to the relatively small number of patients during the study period. Further studies are warranted to compare the GVL effects after CBT with those after haploidentical HCT. Fifth, racial differences could affect rates of acute and chronic GVHD not only in MRD recipients (48), but also UCB recipients (49). Because previous study showed that the risk of acute GVHD was significantly lower in the Japanese patients compared with the Caucasian patients (48). Therefore, it remains to be established whether our results can be applied to other racial groups.

In conclusion, our registry-based data demonstrated that the development of mild acute and chronic GVHD was associated with lower relapse and NRM after CBT, which contributed to the improvement of leukemia-free and OS for acute leukemia. These data indicated that the development of mild GVHD could improve survival after UCB transplantation for acute leukemia.

J. Kanda reports personal fees from Takeda Phamaceutical, Celgene, Novartis Pharma, Astellas Pharma, Chugai Pharmaceutical, Kyowa Kirin, Otsuka Pharmaceutical, Bristol Myers Squibb, JCR Pharmaceuticals, MSD, Daiichi Sankyo, Sanofi, Janssen Pharmaceutical, and Ono Pharmaceutical outside the submitted work. S. Hirabayashi reports personal fees and other support from Revorf Co., Ltd. outside the submitted work. N. Uchida reports personal fees from Chugai Pharmaceutical Co., Astellas Pharma Inc., Otsuka Pharmaceutical Co., Sumitomo Dainippon Pharma Co., and Novartis Pharma Inc. during the conduct of the study. M. Sawa reports personal fees from Chugai, Pfizer, Astellas, Nippon-Shinyaku, Ono, MSD, BMS, Kyowa Kirin, Asahi-Kasei, Novartis, Eisai, Otsuka, Sumitomo Dainippon, Sanofi, Takeda, Celgene, Mochida, Shire, and Mundiphama outside the submitted work. Y. Atsuta reports other support from AbbVie GK, Astellas Pharma Inc., Mochida Pharmaceutical Co., Ltd., Meiji Seika Pharma Co, Ltd., Chugai Pharmaceutical Co., Ltd., and Kyowa Kirin Co., Ltd. outside the submitted work. F. Kimura reports grants from Astellas, Takeda, Taiho, Ono, MSD, Eisai, Kyowa Kirin, Chugai, and Daiichi Sankyo, and grants and non-financial support from Bristol Myers Squibb outside the submitted work. No disclosures were reported by the other authors.

T. Konuma: Conceptualization, formal analysis, supervision, investigation, methodology, writing–original draft, project administration. J. Kanda: Resources, writing–review and editing. Y. Kuwatsuka: Resources, writing–review and editing. M. Yanada: Resources, writing–review and editing. T. Kondo: Resources. S. Hirabayashi: Resources, writing–review and editing. S. Kako: Resources. Y. Akahoshi: Resources. N. Uchida: Resources. N. Doki: Resources. Y. Ozawa: Resources. M. Tanaka: Resources. T. Eto: Resources. M. Sawa: Resources. S. Yoshioka: Resources. T. Kimura: Resources. Y. Kanda: Resources. T. Fukuda: Resources. Y. Atsuta: Resources, funding acquisition. F. Kimura: Resources, supervision.

The authors thank all of the physicians and staff at the hospital, the Japan Marrow Donor Program, and the cord blood banks who provided the clinical data to the Transplant Registry Unified Management Program of the Japanese Data Center for Hematopoietic Cell Transplantation. This work was supported in part by the Practical Research Project for Allergic Diseases and Immunology (Research Technology of Medical Transplantation) from Japan Agency for Medical Research and Development, AMED under grant 18ek0510023h0002.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ringdén
O
,
Karlsson
H
,
Olsson
R
,
Omazic
B
,
Uhlin
M
. 
The allogeneic graft-versus-cancer effect
.
Br J Haematol
2009
;
147
:
614
33
.
2.
Fleischhauer
K
,
Hsu
KC
,
Shaw
BE
. 
Prevention of relapse after allogeneic hematopoietic cell transplantation by donor and cell source selection
.
Bone Marrow Transplant
2018
;
53
:
1498
507
.
3.
Milano
F
,
Gooley
T
,
Wood
B
,
Woolfrey
A
,
Flowers
ME
,
Doney
K
, et al
Cord-blood transplantation in patients with minimal residual disease
.
N Engl J Med
2016
;
375
:
944
53
.
4.
Eapen
M
,
Rocha
V
,
Sanz
G
,
Scaradavou
A
,
Zhang
MJ
,
Arcese
W
, et al
Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis
.
Lancet Oncol
2010
;
11
:
653
60
.
5.
Terakura
S
,
Atsuta
Y
,
Tsukada
N
,
Kobayashi
T
,
Tanaka
M
,
Kanda
J
, et al
Comparison of Outcomes of 8/8 and 7/8 Allele-matched unrelated bone marrow transplantation and single-unit cord blood transplantation in adults with acute leukemia
.
Biol Blood Marrow Transplant
2016
;
22
:
330
38
.
6.
Takahashi
S
,
Ooi
J
,
Tomonari
A
,
Konuma
T
,
Tsukada
N
,
Oiwa-Monna
M
, et al
Comparative single-institute analysis of cord blood transplantation from unrelated donors with bone marrow or peripheral blood stem-cell transplants from related donors in adult patients with hematologic malignancies after myeloablative conditioning regimen
.
Blood
2007
;
109
:
1322
30
.
7.
Sanz
J
,
Jaramillo
FJ
,
Planelles
D
,
Montesinos
P
,
Lorenzo
I
,
Moscardó
F
, et al
Impact on outcomes of human leukocyte antigen matching by allele-level typing in adults with acute myeloid leukemia undergoing umbilical cord blood transplantation
.
Biol Blood Marrow Transplant
2014
;
20
:
106
10
.
8.
Cunha
R
,
Loiseau
P
,
Ruggeri
A
,
Sanz
G
,
Michel
G
,
Iori
AP
, et al
Impact of HLA mismatch direction on outcomes after umbilical cord blood transplantation for hematological malignant disorders: a retrospective Eurocord-EBMT analysis
.
Bone Marrow Transplant
2014
;
49
:
24
9
.
9.
Konuma
T
,
Kato
S
,
Ishii
H
,
Takeda
R
,
Oiwa-Monna
M
,
Tojo
A
, et al
HLA-DRB1 mismatch is associated with a decreased relapse in adult acute myeloid leukemia after single-unit myeloablative cord blood transplantation
.
Ann Hematol
2015
;
94
:
1233
5
.
10.
Yabe
T
,
Azuma
F
,
Kashiwase
K
,
Matsumoto
K
,
Orihara
T
,
Yabe
H
, et al
HLA-DPB1 mismatch induces a graft-versus-leukemia effect without severe acute GVHD after single-unit umbilical cord blood transplantation
.
Leukemia
2018
;
32
:
168
75
.
11.
Zheng
CC
,
Zhu
XY
,
Tang
BL
,
Zhang
XH
,
Zhang
L
,
Geng
LQ
, et al
Clinical separation of cGvHD and GvL and better GvHD-free/relapse-free survival (GRFS) after unrelated cord blood transplantation for AML
.
Bone Marrow Transplant
2017
;
52
:
88
94
.
12.
Atsuta
Y
. 
Introduction of Transplant Registry Unified Management Program 2 (TRUMP2): scripts for TRUMP data analyses, part I (variables other than HLA-related data)
.
Int J Hematol
2016
;
103
:
3
10
.
13.
Kanda
J
. 
Scripts for TRUMP data analyses. Part II (HLA-related data): statistical analyses specific for hematopoietic stem cell transplantation
.
Int J Hematol
2016
;
103
:
11
9
.
14.
Przepiorka
D
,
Weisdorf
D
,
Martin
P
,
Klingemann
HG
,
Beatty
P
,
Hows
J
, et al
1994 consensus conference on acute GVHD grading
.
Bone Marrow Transplant
1995
;
15
:
825
8
.
15.
Sullivan
KM
,
Agura
E
,
Anasetti
C
,
Appelbaum
F
,
Badger
C
,
Bearman
S
, et al
Chronic graft-versus-host disease and other late complications of bone marrow transplantation
.
Semin Hematol
1991
;
28
:
250
9
.
16.
Giralt
S
,
Ballen
K
,
Rizzo
D
,
Bacigalupo
A
,
Horowitz
M
,
Pasquini
M
, et al
Reduced-intensity conditioning regimen workshop: defining the dose spectrum. Report of a workshop convened by the center for international blood and marrow transplant research
.
Biol Blood Marrow Transplant
2009
;
15
:
367
9
.
17.
National Comprehensive Cancer Network
. 
NCCN clinical practice guidelines in oncology, acute myeloid leukemia (Version 2.2016)
.
Available
from: https://www.nccn.org/professionals/physician_gls/pdf/aml.pdf.
18.
Yeshurun
M
,
Weisdorf
D
,
Rowe
JM
,
Tallman
MS
,
Zhang
MJ
,
Wang
HL
, et al
The impact of the graft-versus-leukemia effect on survival in acute lymphoblastic leukemia
.
Blood Adv
2019
;
3
:
670
80
.
19.
Giobbie-Hurder
A
,
Gelber
RD
,
Regan
MM
. 
Challenges of guarantee-time bias
.
J Clin Oncol
2013
;
31
:
2963
9
.
20.
Kanda
Y
. 
Investigation of the freely available easy-to-use software “EZR” for medical statistics
.
Bone Marrow Transplant
2013
;
48
:
452
8
.
21.
Ringdén
O
,
Pavletic
SZ
,
Anasetti
C
,
Barrett
AJ
,
Wang
T
,
Wang
D
, et al
The graft-versus-leukemia effect using matched unrelated donors is not superior to HLA-identical siblings for hematopoietic stem cell transplantation
.
Blood
2009
;
113
:
3110
8
.
22.
Ringdén
O
,
Labopin
M
,
Ciceri
F
,
Velardi
A
,
Bacigalupo
A
,
Arcese
W
, et al
Is there a stronger graft-versus-leukemia effect using HLA-haploidentical donors compared with HLA-identical siblings?
Leukemia
2016
;
30
:
447
55
.
23.
Yu
S
,
Huang
F
,
Wang
Y
,
Xu
Y
,
Yang
T
,
Fan
Z
, et al
Haploidentical transplantation might have superior graft-versus-leukemia effect than HLA-matched sibling transplantation for high-risk acute myeloid leukemia in first complete remission: a prospective multicentre cohort study
.
Leukemia
2020
;
34
:
1433
43
.
24.
Kanda
J
,
Morishima
Y
,
Terakura
S
,
Wake
A
,
Uchida
N
,
Takahashi
S
, et al
Impact of graft-versus-host disease on outcomes after unrelated cord blood transplantation
.
Leukemia
2017
;
31
:
663
8
.
25.
Fleischhauer
K
,
Beelen
DW
. 
HLA mismatching as a strategy to reduce relapse after alternative donor transplantation
.
Semin Hematol
2016
;
53
:
57
64
.
26.
Weiden
PL
,
Flournoy
N
,
Thomas
ED
,
Prentice
R
,
Fefer
A
,
Buckner
CD
, et al
Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts
.
N Engl J Med
1979
;
300
:
1068
73
.
27.
Horowitz
MM
,
Gale
RP
,
Sondel
PM
,
Goldman
JM
,
Kersey
J
,
Kolb
HJ
, et al
Graft-versus-leukemia reactions after bone marrow transplantation
.
Blood
1990
;
75
:
555
62
.
28.
Kanda
Y
,
Izutsu
K
,
Hirai
H
,
Sakamaki
H
,
Iseki
T
,
Kodera
Y
, et al
Effect of graft-versus-host disease on the outcome of bone marrow transplantation from an HLA-identical sibling donor using GVHD prophylaxis with cyclosporin A and methotrexate
.
Leukemia
2004
;
18
:
1013
9
.
29.
Storb
R
,
Gyurkocza
B
,
Storer
BE
,
Sorror
ML
,
Blume
K
,
Niederwieser
D
, et al
Graft-versus-host disease and graft-versus-tumor effects after allogeneic hematopoietic cell transplantation
.
J Clin Oncol
2013
;
31
:
1530
8
.
30.
Stern
M
,
de Wreede
LC
,
Brand
R
,
van Biezen
A
,
Dreger
P
,
Mohty
M
, et al
Sensitivity of hematological malignancies to graft-versus-host effects: an EBMT megafile analysis
.
Leukemia
2014
;
28
:
2235
40
.
31.
Boyiadzis
M
,
Arora
M
,
Klein
JP
,
Hassebroek
A
,
Hemmer
M
,
Urbano-Ispizua
A
, et al
Impact of chronic graft-versus-host disease on late relapse and survival on 7,489 patients after myeloablative allogeneic hematopoietic cell transplantation for leukemia
.
Clin Cancer Res
2015
;
21
:
2020
8
.
32.
Baron
F
,
Labopin
M
,
Savani
BN
,
Beohou
E
,
Niederwieser
D
,
Eder
M
, et al
Graft-versus-host disease and graft-versus-leukaemia effects in secondary acute myeloid leukaemia: a retrospective, multicentre registry analysis from the acute leukaemia working party of the EBMT
.
Br J Haematol
2020
;
188
:
428
37
.
33.
Sullivan
KM
,
Weiden
PL
,
Storb
R
,
Witherspoon
RP
,
Fefer
A
,
Fisher
L
, et al
Influence of acute and chronic graft-versus-host disease on relapse and survival after bone marrow transplantation from HLA-identical siblings as treatment of acute and chronic leukemia
.
Blood
1989
;
73
:
1720
8
.
34.
Ringdén
O
,
Labopin
M
,
Gluckman
E
,
Reiffers
J
,
Vernant
JP
,
Jouet
JP
, et al
Graft-versus-leukemia effect in allogeneic marrow transplant recipients with acute leukemia is maintained using cyclosporin a combined with methotrexate as prophylaxis. acute leukemia working party of the european group for blood and marrow transplantation
.
Bone Marrow Transplant
1996
;
18
:
921
9
.
35.
Akahoshi
Y
,
Igarashi
A
,
Fukuda
T
,
Uchida
N
,
Tanaka
M
,
Ozawa
Y
, et al
Impact of graft-versus-host disease and graft-versus-leukemia effect based on minimal residual disease in Philadelphia chromosome-positive acute lymphoblastic leukemia
.
Br J Haematol
2020
;
190
:
84
92
.
36.
Papaemmanuil
E
,
Gerstung
M
,
Bullinger
L
,
Gaidzik
VI
,
Paschka
P
,
Roberts
ND
, et al
Genomic classification and prognosis in acute myeloid leukemia
.
N Engl J Med
2016
;
374
:
2209
21
.
37.
Roberts
KG
,
Mullighan
CG
. 
Genomics in acute lymphoblastic leukaemia: insights and treatment implications
.
Nat Rev Clin Oncol
2015
;
12
:
344
57
.
38.
Mehta
RS
,
Rezvani
K
. 
Can we make a better match or mismatch with KIR genotyping?
Hematology Am Soc Hematol Educ Program
2016
;
2016
:
106
18
.
39.
Molica
M
,
Breccia
M
,
Foa
R
,
Jabbour
E
,
Kadia
TM
. 
Maintenance therapy in AML: the past, the present and the future
.
Am J Hematol
2019
;
94
:
1254
65
.
40.
Warraich
Z
,
Tenneti
P
,
Thai
T
,
Hubben
A
,
Amin
H
,
McBride
A
, et al
Relapse prevention with tyrosine kinase inhibitors after allogeneic transplantation for Philadelphia chromosome-positive acute lymphoblast leukemia: a systematic review
.
Biol Blood Marrow Transplant
2020
;
26
:
e55
-e64.
41.
McCurdy
SR
,
Kanakry
CG
,
Tsai
HL
,
Kasamon
YL
,
Showel
MM
,
Bolaños-Meade
J
, et al
Grade II acute graft-versus-host disease and higher nucleated cell graft dose improve progression-free survival after HLA-haploidentical transplant with post-transplant cyclophosphamide
.
Biol Blood Marrow Transplant
2018
;
24
:
343
52
.
42.
McCurdy
SR
,
Kanakry
CG
,
Tsai
HL
,
Gojo
I
,
Smith
BD
,
Gladstone
DE
, et al
Development of grade II acute graft-versus-host disease is associated with improved survival after myeloablative HLA-matched bone marrow transplantation using single-agent post-transplant cyclophosphamide
.
Biol Blood Marrow Transplant
2019
;
25
:
1128
35
.
43.
Verneris
MR
,
Brunstein
CG
,
Barker
J
,
MacMillan
ML
,
DeFor
T
,
McKenna
DH
, et al
Relapse risk after umbilical cord blood transplantation: enhanced graft-versus-leukemia effect in recipients of 2 units
.
Blood
2009
;
114
:
4293
9
.
44.
Balligand
L
,
Galambrun
C
,
Sirvent
A
,
Roux
C
,
Pochon
C
,
Bruno
B
, et al
Single-unit versus double-unit umbilical cord blood transplantation in children and young adults with residual leukemic disease
.
Biol Blood Marrow Transplant
2019
;
25
:
734
42
.
45.
Wagner
JE
 Jr
,
Eapen
M
,
Carter
S
,
Wang
Y
,
Schultz
KR
,
Wall
DA
, et al
One-unit versus two-unit cord-blood transplantation for hematologic cancers
.
N Engl J Med
2014
;
371
:
1685
94
.
46.
Michel
G
,
Galambrun
C
,
Sirvent
A
,
Pochon
C
,
Bruno
B
,
Jubert
C
, et al
Single- vs. double-unit cord blood transplantation for children and young adults with acute leukemia or myelodysplastic syndrome
.
Blood
2016
;
127
:
3450
7
.
47.
Konuma
T
,
Ishiyama
K
,
Igarashi
A
,
Uchida
N
,
Ozawa
Y
,
Fukuda
T
, et al
Effects of acute and chronic graft-versus-myelodysplastic syndrome on long-term outcomes following allogeneic hematopoietic cell transplantation
.
Clin Cancer Res
2020
;
26
:
6483
93
.
48.
Oh
H
,
Loberiza
FR
 Jr
,
Zhang
MJ
,
Ringdén
O
,
Akiyama
H
,
Asai
T
, et al
Comparison of graft-versus-host-disease and survival after HLA-identical sibling bone marrow transplantation in ethnic populations
.
Blood
2005
;
105
:
1408
16
.
49.
Kuwatsuka
Y
,
Atsuta
Y
,
Horowitz
MM
,
Inagaki
J
,
Kanda
J
,
Kato
K
, et al
Graft-versus-host disease and survival after cord blood transplantation for acute leukemia: a comparison of Japanese versus White populations
.
Biol Blood Marrow Transplant
2014
;
20
:
662
7
.

Supplementary data