Chronic graft-versus-host disease (cGVHD) remains the major cause of late morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). However, only a few studies specifically focused on children, and little information is available on the antileukemic effect of cGVHD and its impact on disease-free survival (DFS) in children. We retrospectively analyzed 696 children given allogeneic HSCT for malignant (n = 450) or nonmalignant (n = 246) diseases. The donor was an HLA-identical sibling in 461 cases and an alternative donor in 235. Bone marrow was the stem cell source in 647 cases, peripheral blood in 17, and cord blood (CB) in 32. cGVHD developed in 173 children (25%) at a median of 116 days after HSCT. Three-year cGVHD probability was 27%. In multivariate analysis, variables predicting cGVHD were donor and recipient age, grade II to IV acute GVHD, female donor for male recipient, diagnosis of malignancy, and use of total body irradiation; CB transplants had a very low risk of cGVHD (RR = 0.07,P = .0001). cGVHD occurrence increased transplant-related mortality (P < .05). Nevertheless, in hematologic malignancies, patients with cGVHD had a reduced relapse probability compared with children without cGVHD (16% ± 3% versus 39% ± 3%, P = .0001) and a better DFS (68% ± 4% versus 54% ± 3%, P = .01). The antileukemic effect of cGVHD was observed mainly in patients with acute lymphoblastic leukemia (ALL). This study provides novel data on cGVHD in childhood. Use of CB stem cells and preparative regimens without radiotherapy may prevent its development. In patients affected by ALL, cGVHD was associated with a strong graft-versus-leukemia effect, improving DFS.

Chronic graft-versus-host disease (cGVHD) remains the most frequent late complication after allogeneic hematopoietic stem cell transplantation (HSCT) and it represents the major cause of nonrelapse mortality and morbidity in long-term survivors.1-3 In adults, the reported incidence of cGVHD ranges between 30% and 50% of HLA-identical sibling transplant recipients,1 with acute GVHD being recognized as the most important factor predicting the development of the chronic form of the disease.4-6 The increased use of matched unrelated volunteers as donors7 and of peripheral blood as a stem cell source8 9 has led to a further increase of the incidence and severity of this complication.

Chronic GVHD is associated with a graft-versus-leukemia effect (GVL), thus resulting, in the context of transplantation for the treatment of hematologic malignancies, in a decreased incidence of relapse.10-12 Nevertheless, the reduced relapse rate could be offset by an increase in transplant-related mortality (TRM) and counterbalanced by a severely impaired quality of life for patients experiencing the extensive form of the disease.2 3 

Previously published studies documented a lower risk of cGVHD, as well as of acute GVHD, in younger patients.4-6 However, only a few studies specifically focused on children.13-15 For this reason, risk factors for cGVHD in childhood are still poorly defined, and little information is available on the GVL effect of this complication, as well as on its impact on disease-free survival (DFS) and TRM of young patients.

We carried out a retrospective analysis on a large number of children receiving allogeneic HSCT from either a matched or mismatched family donor or an unrelated volunteer, to identify risk factors predicting the development of cGVHD and to assess its impact on clinical outcome.

Patient characteristics

A total of 696 consecutive patients, undergoing allogeneic HSCT between January 1, 1991, and June 30, 1999, in one of the transplantation centers of the Italian Association for Pediatric Hematology and Oncology (AIEOP) and reported to the AIEOP-BMT Registry were included in this analysis. All new transplantations performed were registered yearly by each center, together with the updated follow-up of previously reported transplantations, through disease- and transplant-specific data collection forms.

For the purpose of this study, only patients younger than 18 years of age at the time of HSCT and surviving more than 90 days after the graft were considered for data analysis. Exclusion criteria for the definition of the study population comprised second or subsequent transplantations, the use of a genotypically identical twin as donor, and a diagnosis of solid tumor.

In all donor-recipient pairs, histocompatibility was determined by serology for HLA-A, B, and DR antigens, whereas high-resolution molecular typing of HLA-class II DRB1 was available for all children undergoing HSCT from an unrelated donor. A total of 450 children (65%) had hematologic malignancies; the other 246 children (35%) had a nonmalignant disorder. Children with acute leukemia receiving transplants during first or second complete remission, those with myelodysplastic syndrome (MDS) characterized by a low blast count (ie, refractory anemia and refractory anemia with ringed sideroblasts), as well as patients with chronic myeloid leukemia (CML) in first chronic phase were considered in early phase, whereas all other patients with leukemia or MDS were considered in advanced phase. Of the 696 patients analyzed, 418 were boys and 278 girls. Median age at transplantation was 7 years (range, 0.3-17 years), whereas median donor age was 12 years (range, 0-55 years). The donor was an HLA-identical sibling in 461 cases (66%), a partially matched family donor in 68 cases (10%), and an unrelated volunteer in the remaining 167 patients (24%). The source of stem cells was bone marrow in 647 cases (93%), peripheral blood in 17 (2%), and cord blood (CB) in 32 (5%). GVHD prophylaxis consisted of cyclosporin A (Cs-A) alone in 343 cases (49%) and Cs-A in combination either with methotrexate (MTX) in 127 cases (18%) or with steroids (prednisone [PDN]) in 62 cases (9%). The combination of Cs-A + MTX + in vivo serotherapy (antilymphocyte globulin [ALG] or the monoclonal antibody [mAb] Campath-1G) was adopted for 102 transplants (15%). Nineteen patients (3%) received a T-cell–depleted HSCT.

Details on the 696 patients evaluated, as well as on the transplantation procedure, are reported in Table 1.

Table 1.

Patient characteristics

Characteristics and treatmentsNumberPercentages
Number of patients 696  
Sex, M/F 418/278 60/40  
Median age at transplantation, y (range) 7 (0.3-17)  
Diagnosis   
 Malignant disease 450 65 
  ALL 268 38  
  AML 99 14  
  CML 28 
  MDS 36 5  
  Lymphoma 19 3  
 Nonmalignant disease 246 35  
  Thalassemia 88 12  
  Severe aplastic anemia 35 5  
  Fanconi anemia 23 
  Immunodeficiencies 48 7  
  Inborn errors 52 
Donor   
 HLA-identical family donor 461 66 
 Partially matched family donor 68 10  
 HLA-identical unrelated donor 141 20  
 Partially matched unrelated donor 26 4  
Female donor/male recipient pairs 196 28 
Median donor age, y (range) 12 (0-55)  
Stem cell source   
 Bone marrow 647 93  
 Peripheral blood 17 2  
 Cord blood 32 5  
Conditioning regimen   
 Chemotherapy based 369 53  
 TBI based 327 47  
GVHD prophylaxis   
 Cs-A 348 50 
 Cs-A + MTX 129 19  
 Cs-A + PDN 63 9  
 Cs-A + MTX + ALG or mAb 104 15  
 Cs-A + ALG or mAb ± PDN 33 
 T-cell depletion 19 2  
Acute GVHD   
 Grade 0-I 406 58  
 Grade II-IV 290 42 
Characteristics and treatmentsNumberPercentages
Number of patients 696  
Sex, M/F 418/278 60/40  
Median age at transplantation, y (range) 7 (0.3-17)  
Diagnosis   
 Malignant disease 450 65 
  ALL 268 38  
  AML 99 14  
  CML 28 
  MDS 36 5  
  Lymphoma 19 3  
 Nonmalignant disease 246 35  
  Thalassemia 88 12  
  Severe aplastic anemia 35 5  
  Fanconi anemia 23 
  Immunodeficiencies 48 7  
  Inborn errors 52 
Donor   
 HLA-identical family donor 461 66 
 Partially matched family donor 68 10  
 HLA-identical unrelated donor 141 20  
 Partially matched unrelated donor 26 4  
Female donor/male recipient pairs 196 28 
Median donor age, y (range) 12 (0-55)  
Stem cell source   
 Bone marrow 647 93  
 Peripheral blood 17 2  
 Cord blood 32 5  
Conditioning regimen   
 Chemotherapy based 369 53  
 TBI based 327 47  
GVHD prophylaxis   
 Cs-A 348 50 
 Cs-A + MTX 129 19  
 Cs-A + PDN 63 9  
 Cs-A + MTX + ALG or mAb 104 15  
 Cs-A + ALG or mAb ± PDN 33 
 T-cell depletion 19 2  
Acute GVHD   
 Grade 0-I 406 58  
 Grade II-IV 290 42 

Diagnosis of acute and chronic GVHD and of chronic GVHD resolution

The diagnosis of acute and cGVHD was made by the site investigators at each institution on the basis of clinical symptoms, laboratory tests, and, whenever possible, on histopathologic findings of skin, oral mucosa, and gastrointestinal tract.16-18Acute and cGVHD were classified according to previously described criteria.19,20 Limited cGVHD was defined as either or both localized skin involvement and hepatic dysfunction due to chronic GVHD; extensive cGVHD was defined as generalized skin involvement, or localized skin involvement or hepatic dysfunction due to cGVHD plus liver histology showing chronic aggressive hepatitis, bridging necrosis or cirrhosis, or involvement of the eye or minor salivary glands or oral mucosa, or involvement or any other target organ.20 21 

Resolution of cGVHD was defined as the disappearance of all active lesions attributable to cGVHD (whenever possible confirmed by histopathologic findings) or normalization of liver function tests, associated with the discontinuation of any form of immunosuppressive treatment.

Treatment of chronic GVHD

The choice among the different options for treatment of cGVHD was based on the policy in use in each single transplantation center and varied over time. Thirty-three children received a single drug treatment (Cs-A alone in 20 cases, PDN alone in 11, ursodeoxycholic acid in 1, and mAbs in 1), whereas the standard combination of Cs-A and PDN was used in 62 children, with the addition of azathioprine in 14. Thalidomide was used, always in combination with Cs-A or steroids, in 14 patients. Fifteen children received 3 or more immunosuppressive drugs. Extracorporeal photochemotherapy was used, as single treatment modality or in combination with other systemic immunosuppressive drugs, in 26 children.22 23 Finally, 9 children, all with limited cGVHD, did not need any specific immunosuppressive therapy.

The occurrence of cGVHD, as well as DFS, TRM, and relapse probability according to cGVHD occurrence and extension, were calculated by the Kaplan-Meier method; comparisons between probabilities in different patient groups were performed using the log-rank test.24 As suggested by Pepe et al,25 cGVHD was presented, besides the Kaplan-Meier curve, also as the prevalence of cGVHD over time after transplantation (Figure1B). The prevalence function at time t was calculated using the following function: (number of patients with cGVHD at time t)/(number of patients alive and relapse-free at time t).25 26 

Fig. 1.

Cumulative probability of developing cGVHD and its prevalence.

(A) Overall cumulative probability to develop cGVHD after HSCT in our study population and probability to develop the limited or the extensive form of the disease. (B) Overall prevalence of cGVHD and prevalence of the limited and extensive form of the disease. The prevalence function at time t was calculated using the following function: (number of patients with cGVHD at timet)/(number of patients alive and relapse-free at timet).

Fig. 1.

Cumulative probability of developing cGVHD and its prevalence.

(A) Overall cumulative probability to develop cGVHD after HSCT in our study population and probability to develop the limited or the extensive form of the disease. (B) Overall prevalence of cGVHD and prevalence of the limited and extensive form of the disease. The prevalence function at time t was calculated using the following function: (number of patients with cGVHD at timet)/(number of patients alive and relapse-free at timet).

Close modal

In DFS analysis, both relapse and death in remission due to any cause were considered events, whereas in relapse probability analysis, only disease relapse was considered as failure. In TRM analysis, all deaths not due to disease recurrence were considered events. Results were expressed as probability (%) and 95% CI.

A univariate analysis of DFS, relapse probability, and TRM according to cGVHD occurrence and severity was performed for the whole study population and, subsequently, stratified for malignant and nonmalignant disorders. Furthermore, a separate analysis on the impact of cGVHD on relapse probability and DFS was performed also for the 4 types of hematologic malignancies including the larger number of patients: acute lymphoblastic leukemia (ALL; 268 patients), acute myelogenous leukemia (AML; 99 patients), CML (28 patients), and MDS (36 patients).

In the univariate analysis of risk factors for cGVHD development, the following variables were evaluated: patient and donor age, sex mismatch, parity of female donors, diagnosis, type of donor, stem cell source, use of total body irradiation (TBI) as part of the conditioning regimen, GVHD prophylaxis, infused cell dose, and development of grade II to IV acute GVHD. For this purpose, continuous variables were categorized as follows: each variable was first divided into 4 categories at approximately the 25th, 50th, and 75th percentiles. If the relative event rates (ratio of the observed number of events to the expected number of events in a category, assuming no variation across categories) in 2 or more adjacent categories (and the mean times-to-event) were not substantially different, these categories were grouped together. If no clear pattern was observed for the primary outcome, the median was taken as the cut point.27 

For the multivariate analysis of cGVHD, DFS, and relapse probability, the Cox proportional hazard regression model was used, including in the models all the variables with P < .05 in univariate analysis.28 29 

The χ2 test was used to compare differences in percentages.

All P values were 2-sided and P < .05 was considered statistically significant. P > .1 was reported as not significant (NS), whereas P between .05 and .1 was reported in detail.

The SAS package (SAS Institute, Cary, NC) was used for the analysis of the data.

Data were analyzed as of December 31, 2000. Median follow-up was 57 months (range, 18-119 months) for surviving patients and 9 months (range, 3.4-85 months) for patients not surviving.

Incidence of chronic GVHD and chronic GVHD resolution

Of the 696 patients analyzed, 173 (25%) developed cGVHD at a median of 116 days after transplantation (range, 80-600 days). The limited form of the disease was observed in 109 patients (16%), and extensive cGVHD was diagnosed in 64 cases (9%). As shown in Figure1A, 2 years after the transplantation, the overall cumulative probability of cGVHD was 27% (95% CI, 23%-30%). The cumulative probabilities of developing either limited or cGVHD were 17% (95% CI, 14%-20%) and 11% (95% CI, 8%-14%), respectively.

For patients with cGVHD, median Karnofsky score at time of last follow-up was 90%, with 72 of the 173 patients (42%) having a Karnofsky score of 100% and only 7 (4%) with a Karnofsky score below 50%.

The skin was the most common target of cGVHD, being involved in 144 cases (83%); the liver was involved in 49 patients (28%) and the gastrointestinal tract in 42 (24%). Oral cGVHD was described in 12 patients (7%); both eye involvement and joint contractures were reported in 6 cases (3%). Finally, 19 children (11%) had lung involvement from cGVHD.

Figure 1B shows the prevalence of cGVHD over time after HSCT. Prevalence was greater during the first year after HSCT, when it reached a value of 18%, and progressively decreased to about 5% at 7 years after transplantation. The prevalence of limited cGVHD was greater than that of extensive cGVHD during the first 20 months after HSCT, whereas the prevalence of extensive cGVHD was higher than that of limited cGVHD from 20 months afterward.

Chronic GVHD resolved in 121 patients (70%), whereas it was still present at the time of last follow-up or at time of death in the remaining 52 children (30%). The median duration of cGVHD, in the 121 patients in whom this complication resolved, was 5 months (range, 1-93 months).

Figure 2 shows the cumulative probability of continuing cGVHD, which was equal to 16% (95% CI, 6%-25%) 8 years after transplantation, and the probability of death with cGVHD over time after development of the complication, which was 12% (95% CI, 3%-18%).

Fig. 2.

Cumulative probability of continuing cGVHD and of death with cGVHD over time after the onset of cGVHD.

To calculate the probability of continuing cGVHD, cGVHD resolutions were considered as events, whereas patients whose cGVHD did not resolve were censored at time of last follow-up or of death. To calculate the probability of death with cGVHD, only deaths with active cGVHD were considered events, whereas patients whose cGVHD resolved were censored at time of last follow-up or death.

Fig. 2.

Cumulative probability of continuing cGVHD and of death with cGVHD over time after the onset of cGVHD.

To calculate the probability of continuing cGVHD, cGVHD resolutions were considered as events, whereas patients whose cGVHD did not resolve were censored at time of last follow-up or of death. To calculate the probability of death with cGVHD, only deaths with active cGVHD were considered events, whereas patients whose cGVHD resolved were censored at time of last follow-up or death.

Close modal

The analysis concerning the impact of cGVHD therapy on the probability of cGVHD resolution and on patient outcome did not show any statistically significant difference among the different treatment modalities (data not shown).

Risk factors for chronic GVHD

A number of demographic and transplant-related characteristics were evaluated as potential risk factors for cGVHD. Tables2 and 3present the results of univariate and multivariate analysis of risk factors for development of cGVHD.

Table 2.

Univariate analysis of risk factors for cGVHD development

Univariate analysis
No. of
cases
EventsProbability,
%
(95% CI)P
Patient age      
 Younger than 2 y 108 (3-14) .0001  
 2-5 y 122 27 14 (16-31)  
 5-10 y 239 67 30 (24-36)  
 10-15 y 170 46 29 (22-37)  
 15 y or older 57 24 44 (31-58)  
Donor age      
 Younger than 2 y 49 (0-10) .0002  
 2-5 y 84 10 (3-16)  
 5-10 y 139 32 25 (17-32)  
 10-15 y 100 26 27 (18-36)  
 15-20 y 75 21 30 (19-40)  
 20-30 y 96 30 36 (25-46)  
 30-40 y 96 31 39 (29-49)  
 40 y or older 49 19 42 (27-57)  
 Unknown    
Sex      
 Female donor and male recipient 196 62 34 (27-41) .01 
 Other 497 109 23 (20-27)  
 Unknown    
Parity*      
 Female donor, no pregnancy 34 18 58 (40-76) NS 
 Female donor, 1 or more
pregnancies 
37 15 44 (27-61)  
Diagnosis      
 Nonmalignant disease 246 31 13 (9-17) .001  
 Malignant disease 450 142 35 (30-39)  
Donor      
 HLA-identical family 461 105 24 (20-28) .019  
 Partially matched family 68 15 27 (15-39)  
 HLA-identical unrelated 141 48 37 (29-46)  
 Partially matched unrelated 26 20 (4-35)  
Stem cell source      
 Bone marrow 647 168 28 (24-31) .027  
 Peripheral blood 17 30 (4-55)  
 Cord blood 32 (0-9)  
Stem cell source      
 Bone marrow + peripheral blood 664 172 28 (24-31) .0073  
 Cord blood 32 (0-9)  
Conditioning regimen      
 Chemotherapy based 369 60 17 (13-21) .0001  
 TBI based 327 113 38 (33-44)  
GVHD prophylaxis      
 Cs-A 348 82 25 (20-30) .0052 
 Cs-A + MTX 129 32 27 (19-35)  
 Cs-A + PDN 63 13 23 (12-34)  
 Cs-A + MTX + ALG or mAb 104 39 42 (31-42)  
 Cs-A + ALG or mAb ± PDN 33 15 (1-29)  
 T-cell depletion 19 16 (0-32)  
GVHD prophylaxis      
 Cs-A + MTX + ALG or mAb 104 39 42 (31-52) .0001  
 Other prophylaxis 592 134 24 (21-28)  
Cell dose infused      
 Less than 2 × 108/kg 117 37 35 (26-44) .09  
 2 × 108/kg or more 556 132 25 (21-29)  
 Unknown 23    
Acute GVHD grade      
 Grade 0-I 406 54 14 (11-18) .0001  
 Grade II-IV 290 119 45 (39-51)  
Univariate analysis
No. of
cases
EventsProbability,
%
(95% CI)P
Patient age      
 Younger than 2 y 108 (3-14) .0001  
 2-5 y 122 27 14 (16-31)  
 5-10 y 239 67 30 (24-36)  
 10-15 y 170 46 29 (22-37)  
 15 y or older 57 24 44 (31-58)  
Donor age      
 Younger than 2 y 49 (0-10) .0002  
 2-5 y 84 10 (3-16)  
 5-10 y 139 32 25 (17-32)  
 10-15 y 100 26 27 (18-36)  
 15-20 y 75 21 30 (19-40)  
 20-30 y 96 30 36 (25-46)  
 30-40 y 96 31 39 (29-49)  
 40 y or older 49 19 42 (27-57)  
 Unknown    
Sex      
 Female donor and male recipient 196 62 34 (27-41) .01 
 Other 497 109 23 (20-27)  
 Unknown    
Parity*      
 Female donor, no pregnancy 34 18 58 (40-76) NS 
 Female donor, 1 or more
pregnancies 
37 15 44 (27-61)  
Diagnosis      
 Nonmalignant disease 246 31 13 (9-17) .001  
 Malignant disease 450 142 35 (30-39)  
Donor      
 HLA-identical family 461 105 24 (20-28) .019  
 Partially matched family 68 15 27 (15-39)  
 HLA-identical unrelated 141 48 37 (29-46)  
 Partially matched unrelated 26 20 (4-35)  
Stem cell source      
 Bone marrow 647 168 28 (24-31) .027  
 Peripheral blood 17 30 (4-55)  
 Cord blood 32 (0-9)  
Stem cell source      
 Bone marrow + peripheral blood 664 172 28 (24-31) .0073  
 Cord blood 32 (0-9)  
Conditioning regimen      
 Chemotherapy based 369 60 17 (13-21) .0001  
 TBI based 327 113 38 (33-44)  
GVHD prophylaxis      
 Cs-A 348 82 25 (20-30) .0052 
 Cs-A + MTX 129 32 27 (19-35)  
 Cs-A + PDN 63 13 23 (12-34)  
 Cs-A + MTX + ALG or mAb 104 39 42 (31-42)  
 Cs-A + ALG or mAb ± PDN 33 15 (1-29)  
 T-cell depletion 19 16 (0-32)  
GVHD prophylaxis      
 Cs-A + MTX + ALG or mAb 104 39 42 (31-52) .0001  
 Other prophylaxis 592 134 24 (21-28)  
Cell dose infused      
 Less than 2 × 108/kg 117 37 35 (26-44) .09  
 2 × 108/kg or more 556 132 25 (21-29)  
 Unknown 23    
Acute GVHD grade      
 Grade 0-I 406 54 14 (11-18) .0001  
 Grade II-IV 290 119 45 (39-51)  
*

The impact of parity on cGVHD occurrence was evaluated in the 117 donor-recipient pairs with a female donor older than 20 years; data on parity were available for 71 of these 117 donors (61%).

Table 3.

Multivariate analysis of risk factors for cGVHD development

Multivariate analysis
RR(95% CI)P
Patient age    
 Younger than 5 y 1.00   
 5-15 y 1.39 (0.93 -2.08) .1  
 15 y or older 1.82 (1.06 -3.12) .03  
Donor age    
 Younger than 5 y 1.00   
 5-20 y 3.26 (1.57 -6.77) .001  
 20 y or older 5.73 (2.56 -12.8) .0001  
Sex    
 Female donor and male recipient vs others 1.52 (1.11 -2.07) .01 
Diagnosis    
 Malignant vs nonmalignant 1.80 (1.13 -2.88) .01 
Donor    
 HLA-identical family vs other donors .69 (0.41 -1.17) NS  
Stem cell source    
 CB vs bone marrow + peripheral blood .07 (0.03 -0.16) .0001  
Conditioning regimen    
 TBI vs chemotherapy 1.56 (1.08 -2.26) .02  
GVHD prophylaxis    
 Cs-A + MTX + ALG or mAb vs other
prophylaxis 
1.55 (0.92 -2.51) .08  
Acute GVHD grade    
 Grade II-IV vs grade 0-I 2.14 (1.78 -2.59) .0001 
Multivariate analysis
RR(95% CI)P
Patient age    
 Younger than 5 y 1.00   
 5-15 y 1.39 (0.93 -2.08) .1  
 15 y or older 1.82 (1.06 -3.12) .03  
Donor age    
 Younger than 5 y 1.00   
 5-20 y 3.26 (1.57 -6.77) .001  
 20 y or older 5.73 (2.56 -12.8) .0001  
Sex    
 Female donor and male recipient vs others 1.52 (1.11 -2.07) .01 
Diagnosis    
 Malignant vs nonmalignant 1.80 (1.13 -2.88) .01 
Donor    
 HLA-identical family vs other donors .69 (0.41 -1.17) NS  
Stem cell source    
 CB vs bone marrow + peripheral blood .07 (0.03 -0.16) .0001  
Conditioning regimen    
 TBI vs chemotherapy 1.56 (1.08 -2.26) .02  
GVHD prophylaxis    
 Cs-A + MTX + ALG or mAb vs other
prophylaxis 
1.55 (0.92 -2.51) .08  
Acute GVHD grade    
 Grade II-IV vs grade 0-I 2.14 (1.78 -2.59) .0001 

Several variables were strongly associated with an increased risk of cGVHD in univariate analysis. In particular, older patient age was significantly associated with cGVHD, with children younger than 5 years having a probability of less than 14% of developing this complication, those aged 5 to 15 years a probability of about 30%, and those older than 15 years a probability of 44% (P = .0001). Also donor age was a significant risk factor for cGVHD occurrence in univariate analysis, with a continuous increase in the incidence of this complication from less than 10% for donors younger than 5 years, to more than 40% for donors older than 40 years of age (P = .0002). Female donor and male recipient combination was another factor predicting cGVHD development (34% versus 23%, P = .01). Furthermore, patients affected by hematologic malignancies had a greater incidence of cGVHD (35% versus 13%, P = .01), as well as those treated with TBI (38% versus 17%, P = .0001) and those with previous grade II to IV acute GVHD (45% versus 14%, P = .0001). CB transplants had a significantly lower probability of cGVHD (3%) compared with bone marrow or peripheral blood transplants (28%,P = .0073). As reported in Table 2, in our cohort of patients, parity of female donors did not show any effect on the risk of cGVHD development.

In multivariate analysis, the following 6 characteristics were associated with an increased probability of developing cGVHD: patient age 15 years or older (RR = 1.82, P = .03), donor age 5 years or older (RR = 3.36, P < .001), female donor and male recipient (RR = 1.52, P = .01), use of TBI as part of the conditioning regimen (RR = 1.56, P = .02), diagnosis of hematologic malignancy (RR = 1.80,P = .01), and, with the highest relative risk, previous grade II to IV acute GVHD (RR = 2.14, P = .0001). On the contrary, transplants of CB stem cells were characterized by a very low incidence of cGVHD, with only 1 child of 32 presenting this complication (RR = 0.07, P = .001).

Causes of death and TRM

Overall, 191 (27%) of the 696 patients analyzed died; 146 (28%) belonged to the group of 523 subjects without cGVHD, whereas 45 (26%) belonged to the 173 children with cGVHD.

Details on the causes of death for patients with and without cGVHD are reported in Table 4. Disease progression was the main cause of death in 102 patients, whereas 89 children died of transplant-related causes. In patients with cGVHD, we observed a significantly higher percentage of transplant-related deaths (69%) as compared to subjects without cGVHD (40%), and a lower percentage of deaths due to disease progression (31% versus 60%,P = .0006). In the group of children who had cGVHD, this complication was indicated as the primary cause of death in 11 of the 31 patients who died due to transplant-related causes (35%), so that 6% of the 173 patients with cGVHD died because of this complication.

Table 4.

Causes of death for patients with and without cGVHD

No chronic
GVHD
(523 patients)
Chronic
GVHD
(173 patients)
Total
(696 patients)
Disease progression (%) 88 (60) 14 (31) 102  (53) 
Transplant-related causes (%) 58 (40) 31 (69) 89  (47)  
 Bacterial pneumonia  
 HCMV interstitial pneumonia  
 Bacterial infection 10  
 Fungal infection  
 Viral infection  
 EBV-PTLD  
 Encephalitis  
 ARDS  
 Idiopathic interstitial pneumonia  
 Autoimmune hemolytic anemia  
 Thrombotic thrombocytopenic
purpura 
 
 Liver failure  
 Renal failure  
 Cardiac insufficiency  
 Multiorgan failure  
 Hemorrhage 10  
 cGVHD 11  
Total 146 45 191 
No chronic
GVHD
(523 patients)
Chronic
GVHD
(173 patients)
Total
(696 patients)
Disease progression (%) 88 (60) 14 (31) 102  (53) 
Transplant-related causes (%) 58 (40) 31 (69) 89  (47)  
 Bacterial pneumonia  
 HCMV interstitial pneumonia  
 Bacterial infection 10  
 Fungal infection  
 Viral infection  
 EBV-PTLD  
 Encephalitis  
 ARDS  
 Idiopathic interstitial pneumonia  
 Autoimmune hemolytic anemia  
 Thrombotic thrombocytopenic
purpura 
 
 Liver failure  
 Renal failure  
 Cardiac insufficiency  
 Multiorgan failure  
 Hemorrhage 10  
 cGVHD 11  
Total 146 45 191 

χ2 = 11.76, P = .0006 for the comparison of causes of death (disease progression versus transplant-related cause) in subjects with and without cGVHD.

For the 11 patients with cGVHD reported as the main cause of death, the ultimate cause of death was bronchiolitis obliterans in 4 cases, HCMV interstitial pneumonia in 1 case, bacterial pneumonia in 1 case, other bacterial infections in 3 cases, hemorrhagic complications in 1 case, and multiorgan failure in 1 case. HCMV indicates human cytomegalovirus; EBV-PTLD, Epstein-Barr virus–related posttransplantation lymphoproliferative disease; ARDS, acute respiratory distress syndrome.

In the 11 cases where cGVHD was reported as the main cause of death, the ultimate cause of death was bronchiolitis obliterans in 4 cases, cytomegalovirus interstitial pneumonia in 1 case, bacterial pneumonia in 1 case, other bacterial infections in 3 cases, hemorrhagic complications in 1 case, and multiorgan failure in 1 case.

For patients treated for nonmalignant disorders, the cumulative probability of TRM was 13% (95% CI, 7%-19%) for children without cGVHD, 5% (95% CI, 0%-15%) for those with the limited form of the disease, and 64% (95% CI, 11%-100%) for those with extensive cGVHD (P = .016). For subjects treated for malignant diseases, the cumulative probability of TRM was 9% (95% CI, 5%-13%) for children without cGVHD, 15% (95% CI, 5%-25%) for those with limited disease, and 24% (95% CI, 12%-36%) for those with extensive cGVHD (P = .036).

Relapse probability

Of the 450 children who underwent HSCT for treatment of hematologic malignancy, 127 (28%) experienced disease relapse. Analyzing the risk of relapse according to cGVHD occurrence, we observed that the 6-year cumulative relapse probability was significantly lower (P = .0001) in children with cGVHD, being 16% (95% CI, 10%-22%) compared with a value of 39% (95% CI, 33%-45%) in patients without cGVHD (Figure 3).

Fig. 3.

Six-year relapse probability after HSCT for children affected by hematologic malignancies, according to cGVHD development.

Patients experiencing cGVHD had a significantly lower risk of relapse (P = .0001).

Fig. 3.

Six-year relapse probability after HSCT for children affected by hematologic malignancies, according to cGVHD development.

Patients experiencing cGVHD had a significantly lower risk of relapse (P = .0001).

Close modal

Tables 5 and6 show the results of univariate and multivariate analysis of the probability of relapse in the 450 children affected by hematologic malignancies. cGVHD confirmed its protective effect against relapse also in multivariate analysis (RR = 0.32,P = .0001, for patients with cGVHD versus those without cGVHD).

Table 5.

Univariate analysis of relapse probability in the 450 patients with malignant disease

Univariate analysis
Number of
cases
EventsProbability,
%
(95% CI)P
Patient age      
 Younger than 5 y 110 30 28 (20 -37) NS 
 5-10 y 174 51 34 (26 -41)  
 10-15 y 125 32 29 (20 -38)  
 15 y or older 41 14 41 (22 -60)  
Donor age      
 Younger than 5 y 81 23 31 (20 -42) NS  
 5-10 y 95 24 27 (17 -36)  
 10-15 y 64 18 33 (19 -46)  
 15-20 y 55 14 28 (15 -40)  
 20-30 y 69 22 34 (23 -46)  
 30-40 y 56 17 37 (21 -52)  
 40 y or older 30 44 (15 -72)  
Sex      
 Female donor and male
recipient 
103 20 21 (13 -29) .0706 
 Other 320 90 32 (26 -38)  
Diagnosis      
 ALL 268 87 35 (29 -41) NS  
 AML 99 24 31 (19 -42)  
 MDS 36 25 (10 -40)  
 CML 28 18 (0 -39)  
 Lymphoma 19 28 (7 -49)  
Disease phase      
 Early 285 66 26 (21 -32) .0003  
 Advanced 165 61 42 (34 -51)  
Donor      
 HLA-identical family 306 85 30 (25 -36) NS  
 Other donor 144 42 36 (26 -46)  
Stem cell source      
 Bone marrow + peripheral
blood 
434 121 31 (26 -36) NS  
 Cord blood 16 52 (19 -85)  
GVHD prophylaxis      
 Cs-A + MTX + ALG or mAb 81 25 34 (23 -45) NS  
 Other prophylaxis 369 102 31 (26 -36)  
Conditioning regimen      
 Chemotherapy based 148 41 33 (24 -43) NS  
 TBI based 302 86 31 (26 -37)  
Acute GVHD      
 0-I 223 77 38 (31 -45) .0005  
 II-IV 227 49 25 (18 -31)  
Chronic GVHD      
 Absent 308 107 39 (33 -45) .0001  
 Present 142 20 16 (9 -22)  
Chronic GVHD      
 Absent 308 107 39 (33 -45) .0001  
 Limited 89 16 20 (11 -28)  
 Extensive 53 10 (0 -19)  
Univariate analysis
Number of
cases
EventsProbability,
%
(95% CI)P
Patient age      
 Younger than 5 y 110 30 28 (20 -37) NS 
 5-10 y 174 51 34 (26 -41)  
 10-15 y 125 32 29 (20 -38)  
 15 y or older 41 14 41 (22 -60)  
Donor age      
 Younger than 5 y 81 23 31 (20 -42) NS  
 5-10 y 95 24 27 (17 -36)  
 10-15 y 64 18 33 (19 -46)  
 15-20 y 55 14 28 (15 -40)  
 20-30 y 69 22 34 (23 -46)  
 30-40 y 56 17 37 (21 -52)  
 40 y or older 30 44 (15 -72)  
Sex      
 Female donor and male
recipient 
103 20 21 (13 -29) .0706 
 Other 320 90 32 (26 -38)  
Diagnosis      
 ALL 268 87 35 (29 -41) NS  
 AML 99 24 31 (19 -42)  
 MDS 36 25 (10 -40)  
 CML 28 18 (0 -39)  
 Lymphoma 19 28 (7 -49)  
Disease phase      
 Early 285 66 26 (21 -32) .0003  
 Advanced 165 61 42 (34 -51)  
Donor      
 HLA-identical family 306 85 30 (25 -36) NS  
 Other donor 144 42 36 (26 -46)  
Stem cell source      
 Bone marrow + peripheral
blood 
434 121 31 (26 -36) NS  
 Cord blood 16 52 (19 -85)  
GVHD prophylaxis      
 Cs-A + MTX + ALG or mAb 81 25 34 (23 -45) NS  
 Other prophylaxis 369 102 31 (26 -36)  
Conditioning regimen      
 Chemotherapy based 148 41 33 (24 -43) NS  
 TBI based 302 86 31 (26 -37)  
Acute GVHD      
 0-I 223 77 38 (31 -45) .0005  
 II-IV 227 49 25 (18 -31)  
Chronic GVHD      
 Absent 308 107 39 (33 -45) .0001  
 Present 142 20 16 (9 -22)  
Chronic GVHD      
 Absent 308 107 39 (33 -45) .0001  
 Limited 89 16 20 (11 -28)  
 Extensive 53 10 (0 -19)  
Table 6.

Multivariate analysis of relapse probability in the 450 patients with malignant disease

Multivariate analysis
RR(95% CI)P
Sex    
 Female donor and male recipient vs others 0.82 (0.56-1.21) NS 
Disease phase    
 Advanced vs early 2.11 (1.48 -3.00) .0001  
Acute GVHD    
 II-IV vs 0-I 0.91 (0.65 -1.27) NS 
cGVHD    
 Present vs absent 0.32 (0.20 -0.52) .0001 
Multivariate analysis
RR(95% CI)P
Sex    
 Female donor and male recipient vs others 0.82 (0.56-1.21) NS 
Disease phase    
 Advanced vs early 2.11 (1.48 -3.00) .0001  
Acute GVHD    
 II-IV vs 0-I 0.91 (0.65 -1.27) NS 
cGVHD    
 Present vs absent 0.32 (0.20 -0.52) .0001 

Variables with P < .1 in univariate analysis were included in the multivariate analysis.

Stratifying the analysis by type of malignancy, we observed that the GVL effect associated with cGVHD was stronger in the 268 patients with ALL. In fact, the relapse probability for children with ALL and cGVHD was 14% (95% CI, 6%-21%) compared with the 47% (95% CI, 39%-54%) observed in children with ALL but without cGVHD (P = .0001). On the contrary, for the 99 patients with AML, the relapse probability was identical (29%) in both groups of patients. Among the 20 children with MDS, development of cGVHD was associated with a lower relapse probability, even though the advantage was not statistically significant (0% versus 36%,P = .053). Because only 3 of the 28 children with CML experienced a relapse, it was impossible to analyze the role of cGVHD on leukemia recurrence in this group of patients.

Survival and DFS

Overall, 482 of the 696 patients analyzed (69%) are alive and disease free, with a cumulative probability of DFS of 67% (95% CI, 63%-71%).

Figure 4 shows the 6-year cumulative probability of DFS for patients with either nonmalignant disorders (Figure 4A) or hematologic malignancies (Figure 4B), respectively. For children with nonmalignant diseases, no statistically significant difference was observed for DFS between patients without or with cGVHD (85% versus 77%, P = NS), this finding being due to the fact that most of patients with cGVHD had the limited form of the disease. On the contrary, considering children receiving transplants for malignant diseases, patients who developed cGVHD had a significantly better DFS, as compared to children without cGVHD (68% versus 54%, P = .0013).

Fig. 4.

Probability of DFS.

Six-year probability of DFS according to the development of cGVHD is shown for patients affected by nonmalignant disorders (A) and for those affected by hematologic malignancies (B). The difference was not statistically significant for nonmalignant disorders, whereas in the group of patients affected by malignant diseases, the advantage for subjects with cGVHD was statistically significant (P = .0013).

Fig. 4.

Probability of DFS.

Six-year probability of DFS according to the development of cGVHD is shown for patients affected by nonmalignant disorders (A) and for those affected by hematologic malignancies (B). The difference was not statistically significant for nonmalignant disorders, whereas in the group of patients affected by malignant diseases, the advantage for subjects with cGVHD was statistically significant (P = .0013).

Close modal

Tables 7 and8 summarize the results of univariate and multivariate analysis of DFS probability for the 450 children affected by malignant diseases. As for relapse, the positive impact of cGVHD on DFS was confirmed also in multivariate analysis, where this variable was associated with a relative risk of DFS of 2.07 (P = .0001).

Table 7.

Univariate analysis of DFS probability in the 450 patients with malignant disease

Univariate analysis
No. of
cases
EventsProbability,
%
(95% CI)P
Patient age      
 Younger than 5 y 110 35 66 (56 -76) NS 
 5-10 y 174 72 57 (49 -64)  
 10-15 y 125 52 57 (48 -66)  
 15 y or older 41 17 54 (35 -72)  
Donor age      
 Younger than 5 y 81 31 61 (50 -72) .09  
 5-0 y 95 31 65 (55 -76)  
 10-15 y 64 22 63 (49 -76)  
 15-20 y 55 18 65 (52 -78)  
 20-30 y 69 32 52 (40 -64)  
 30-40 y 56 24 48 (34 -62)  
 40 y or older 30 14 45 (21 -70)  
Sex      
 Female donor and male
recipient 
103 53 59 (50 -67) NS 
 Other 320 123 59 (53 -65)  
Diagnosis      
 ALL 268 116 56 (50 -62) NS  
 AML 99 30 64 (53 -76)  
 MDS 36 15 54 (35 -73)  
 CML 28 66 (45 -87)  
 Lymphoma 19 63 (41 -85)  
Disease phase      
 Early 285 93 65 (59 -71) .0001  
 Advanced 165 83 48 (39 -56)  
Donor      
 HLA-identical family 306 103 65 (59 -70) .0003  
 Other donors 144 73 45 (34 -55)  
Stem cell source      
 Bone marrow + peripheral
blood 
434 167 59 (54 -64) .1032  
 Cord blood 16 38 (9 -66)  
GVHD prophylaxis      
 Cs-A + MTX + ALG or mAb 81 39 51 (39 -62) .0533  
 Other prophylaxis 369 167 61 (55 -66)  
Conditioning regimen      
 Chemotherapy based 148 52 59 (50 -69) NS  
 TBI based 302 124 58 (52 -64)  
Acute GVHD      
 0-I 223 89 58 (51 -65) NS  
 II-IV 227 87 60 (53 -67)  
cGVHD      
 Absent 308 134 55 (49 -60) .0013  
 Present 142 42 68 (60 -76)  
cGVHD      
 Absent 308 134 55 (49 -60) .0055  
 Limited 89 26 68 (58 -79)  
 Extensive 53 16 68 (54 -81)  
Univariate analysis
No. of
cases
EventsProbability,
%
(95% CI)P
Patient age      
 Younger than 5 y 110 35 66 (56 -76) NS 
 5-10 y 174 72 57 (49 -64)  
 10-15 y 125 52 57 (48 -66)  
 15 y or older 41 17 54 (35 -72)  
Donor age      
 Younger than 5 y 81 31 61 (50 -72) .09  
 5-0 y 95 31 65 (55 -76)  
 10-15 y 64 22 63 (49 -76)  
 15-20 y 55 18 65 (52 -78)  
 20-30 y 69 32 52 (40 -64)  
 30-40 y 56 24 48 (34 -62)  
 40 y or older 30 14 45 (21 -70)  
Sex      
 Female donor and male
recipient 
103 53 59 (50 -67) NS 
 Other 320 123 59 (53 -65)  
Diagnosis      
 ALL 268 116 56 (50 -62) NS  
 AML 99 30 64 (53 -76)  
 MDS 36 15 54 (35 -73)  
 CML 28 66 (45 -87)  
 Lymphoma 19 63 (41 -85)  
Disease phase      
 Early 285 93 65 (59 -71) .0001  
 Advanced 165 83 48 (39 -56)  
Donor      
 HLA-identical family 306 103 65 (59 -70) .0003  
 Other donors 144 73 45 (34 -55)  
Stem cell source      
 Bone marrow + peripheral
blood 
434 167 59 (54 -64) .1032  
 Cord blood 16 38 (9 -66)  
GVHD prophylaxis      
 Cs-A + MTX + ALG or mAb 81 39 51 (39 -62) .0533  
 Other prophylaxis 369 167 61 (55 -66)  
Conditioning regimen      
 Chemotherapy based 148 52 59 (50 -69) NS  
 TBI based 302 124 58 (52 -64)  
Acute GVHD      
 0-I 223 89 58 (51 -65) NS  
 II-IV 227 87 60 (53 -67)  
cGVHD      
 Absent 308 134 55 (49 -60) .0013  
 Present 142 42 68 (60 -76)  
cGVHD      
 Absent 308 134 55 (49 -60) .0055  
 Limited 89 26 68 (58 -79)  
 Extensive 53 16 68 (54 -81)  
Table 8.

Multivariate analysis of DFS probability in the 450 patients with malignant disease

Multivariate analysis
RR(95% CI)P
Donor age    
 Younger than 20 y vs 20 y or older 1.23 (0.79 -1.90) NS  
Disease phase    
 Advanced vs early 0.57 (0.42 -0.78) .0004 
Donor    
 HLA-identical family vs other donors 1.52 (0.95 -2.43) .08  
GVHD prophylaxis    
 Cs-A + MTX + ALG or mAb vs other prophylaxis .97 (0.58 -1.64) NS 
cGVHD    
 Present vs absent 2.04 (1.43 -2.91) .0001 
Multivariate analysis
RR(95% CI)P
Donor age    
 Younger than 20 y vs 20 y or older 1.23 (0.79 -1.90) NS  
Disease phase    
 Advanced vs early 0.57 (0.42 -0.78) .0004 
Donor    
 HLA-identical family vs other donors 1.52 (0.95 -2.43) .08  
GVHD prophylaxis    
 Cs-A + MTX + ALG or mAb vs other prophylaxis .97 (0.58 -1.64) NS 
cGVHD    
 Present vs absent 2.04 (1.43 -2.91) .0001 

Variables with P < .1 in univariate analysis were included in the multivariate analysis.

As already observed for relapse probability, also for DFS the favorable influence of cGVHD was observed only for children with ALL, where the cumulative probability of DFS was 74% (95% CI, 64%-83%) for patients developing cGVHD and 47% (95% CI, 38%-53%) for children without cGVHD (P = .0001), whereas the effect on other hematologic malignancies was nonsignificant. In fact, in the 99 patients with AML, the DFS probability was 59% (95% CI, 36%-82%) for children with cGVHD and 68% for those without cGVHD (P = NS), and in the 28 children with CML it was 64% (95% CI, 35%-92%) and 71% (95% CI, 44%-97%) for subjects with and without cGVHD, respectively (P = NS). Finally, also considering the 20 children with MDS, the outcome of patients with and without cGVHD was similar, DFS being 67% (95% CI, 36%-97%) and 64% (95% CI, 35%-92%), respectively (P = NS).

Chronic GVHD remains a substantial problem of allogeneic HSCT. Several studies, published in recent years, introduced the notion that recipient age is a major risk factor for the development of cGVHD and that children have a lower probability of presenting this complication.4-6 However, this information, as well as our clinical knowledge of cGVHD and of its risk factors, are based primarily on results of analyses performed in adults.

In our study, conducted on the largest pediatric population reported so far, we observed a cumulative probability of cGVHD of 27%, which is similar to probabilities ranging from 22% to 29% reported by 3 other analyses conducted on smaller cohorts of pediatric patients in recent years.13-15 This probability is nearly half of the estimated probability of 40% to 50% described in adults.4-6 In our group of patients younger than 18 years, older patient and donor age were predictive of a higher risk of cGVHD both in univariate and in multivariate analysis. On the other hand, the type of donor and of GVHD prophylaxis was not predictive of the development of this complication. In multivariate analysis, other factors, such as the female donor–male recipient combination, a diagnosis of malignant disease, the use of TBI, and, as expected, previous grade II to IV acute GVHD, were characterized by a significantly higher risk of cGVHD. Furthermore, in agreement with the observation of Rocha et al,30 use of CB progenitor cells was confirmed to be characterized by a very low probability of cGVHD. In contrast, we could not confirm the observation of an increased risk of cGVHD in subjects receiving peripheral blood stem cells.8 9 This was possibly due to the limited number of patients receiving peripheral blood stem cells.

Our study confirms previously published reports documenting that male patients given a transplant from a female donor experience more GVHD,6 an observation that has been biologically explained hypothesizing that male antigens (ie, H-Y) not shared by females are recognized and attacked by the donor's immune system.31 32 However, our data are particularly intriguing because most female donors were young and, thus, not sensitized toward male antigens by previous pregnancies. Furthermore, even if evaluated on a limited number of subjects, parity of female donors was not a risk factor for cGVHD development; this suggests that even a primary immune response toward these molecules may produce cGVHD.

Children with nonmalignant disorders had a reduced risk of developing cGVHD. Several factors may be considered to explain this observation. Because children with these diseases do not benefit at all from GVHD, it is reasonable to hypothesize that the most effective pharmacologic strategies for both GVHD prevention and therapy of acute GVHD were used in these patients. Moreover, it has been already reported that these patients can have a mixed chimerism,33-35 and this condition is associated with reduced susceptibility to GVHD, probably through mechanisms of central tolerance with negative selection of both host-reactive and donor-reactive T cells.36 Finally, some of the children with nonmalignant disorders (ie, those with aplastic anemia or with congenital immunodeficiencies) were given less intensive preparative regimens and it has been hypothesized that the cytokine storm, which is dependent on the intensity of the conditioning regimen, triggers development of GVHD.37 In this regard, the finding of an increased risk of cGVHD in children given TBI as part of the preparative regimen is also of particular interest. Moreover, because it has been suggested as a relevant role of recipient's thymus in promoting tolerance of donor cells, we speculate that the higher risk of cGVHD in children given TBI can be partly explained by a radiation-induced damage to thymic epithelial cells.38-40 

We also found an increased risk of cGVHD in patients receiving a transplant from an older donor. The detrimental effect of increased donor age on cGVHD has been recently reported in a cohort of patients given HSCT from unrelated volunteers.41 The biologic mechanisms accounting for this effect of donor age remain unclear and deserve further investigation.

Considerable experimental and clinical data suggest a role of the immune system in controlling cancer and, particularly, leukemia, through the GVL effect.12 42-45 In our study, we document a strong protective effect of cGVHD against relapse in children with ALL, whereas the advantage was lower and not statistically significant for patients with MDS and absent for those with AML. Moreover, in children with ALL, the increased TRM associated with cGVHD was largely offset by the impressive reduction in relapse probability, thus resulting in a significantly better survival and DFS.

Despite a beneficial GVL effect against relapse, cGVHD remains a major determinant of poor long-term outcome and of impaired quality of life after allogeneic HSCT.2 For this reason, several efforts have been made to identify factors having an unfavorable prognostic significance, to select subjects with higher risk of either death or invalidating sequelae (as severe joint contractures requiring physical therapy, impairment of lung function, keratoconjunctivitis sicca with corneal lesions, malnutrition due to gastrointestinal involvement), who need to be treated promptly and more intensively.46-49Over the last few years, new drugs and different treatment modalities have been developed for patients with cGVHD,50 even though the optimal treatment is still controversial and no single therapy has been demonstrated to be superior and most effective.51 

In our study population, subjects developing cGVHD had a significantly higher TRM, as compared to those without the disease, even though only 6% of children with cGVHD died directly or indirectly because of this complication. The negative effect of cGVHD was particularly evident in patients with the extensive form of the disease and for those affected by nonmalignant disorders. These observations emphasize the need for a more effective prophylaxis for patients undergoing transplantation with the aim of curing nonmalignant disorders and a more effective treatment for those with extensive GVHD or with poor prognostic clinical features.

Our data document that children have a lower incidence of cGVHD compared with adults. Use of CB stem cells and of conditioning regimens without TBI, as well as better strategies for preventing acute GVHD occurrence, could further decrease the risk of this complication. We provide evidence for a strong antileukemia effect of cGVHD for ALL, but not for other hematologic malignancies. Despite an increased TRM, the probability of DFS in ALL was not impaired by the occurrence of this complication.

We would like to thank Nicoletta Sacchi, at the Italian Bone Marrow Donor Registry, for the help in collecting part of the data on parity of female donors.

Prepublished online as Blood First Edition Paper, May 17, 2002; DOI 10.1182/blood-2001-11-0059.

Partly supported by grants from Associazione Italiana Ricerca sul Cancro (AIRC), Consiglio Nazionale delle Ricerche (CNR), Ministero dell'Università e della Ricerca Scientifica e Tecnologica (MURST), and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico S. Matteo to F.L. and by a grant from IRCCS Policlinico San Matteo to M.Z.

M.Z. and A.P. contributed equally to this work.

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 U.S.C. section 1734.

1
Atkinson
K
Chronic graft-versus-host disease.
Bone Marrow Transplant.
5
1990
69
82
2
Deeg
HJ
Leisenring
W
Storb
R
et al
Long-term outcome after marrow transplantation for severe aplastic anemia.
Blood.
91
1998
3637
3645
3
Kulkarni
S
Powles
R
Treleaven
J
et al
Chronic graft versus host disease is associated with long-term risk for pneumococcal infections in recipients of bone marrow transplants.
Blood.
95
2000
3683
3686
4
Atkinson
K
Horowitz
MM
Gale
RP
et al
Risk factors for chronic graft-versus-host disease after HLA-identical sibling bone marrow transplantation.
Blood.
75
1990
2459
2464
5
Ochs
LA
Miller
WJ
Filipovich
AH
et al
Predictive factors for chronic graft-versus-host disease after histocompatible sibling donor bone marrow transplantation.
Bone Marrow Transplant.
13
1994
455
460
6
Carlens
S
Ringdén
O
Remberger
M
et al
Risk factors for chronic graft-versus-host disease after bone marrow transplantation: a retrospective single centre analysis.
Bone Marrow Transplant.
22
1998
755
761
7
Marks
DI
Cullis
JO
Ward
KN
Lacey
S
Syzdlo
R
Hughes
TP
Allogeneic bone marrow transplantation for chronic myeloid leukemia using sibling and volunteer unrelated donors: a comparison of complications in the first 2 years.
Ann Intern Med.
119
1993
207
214
8
Cutler
C
Giri
S
Jeyapalan
S
Paniagua
D
Viswanathan
A
Antin
JH
Acute and chronic graft-versus-host disease after allogeneic peripheral-blood stem-cell and bone marrow transplantation: a meta-analysis.
J Clin Oncol.
19
2001
3685
3691
9
Przepiorka
D
Anderlini
P
Saliba
R
et al
Chronic graft-versus-host disease after allogeneic blood stem cell transplantation.
Blood.
98
2001
1695
1700
10
Ringdén
O
Labopin
M
Gluckman
E
et al
Strong antileukemic effect of chronic graft-versus-host disease in allogeneic marrow transplant recipients having acute leukemia treated with methotrexate and cyclosporine.
Transplant Proc.
29
1997
733
734
11
Zikos
P
Van-Lint
MT
Lamparelli
T
et al
Allogeneic hemopoietic stem cell transplantation for patients with high risk acute lymphoblastic leukemia: favorable impact of chronic graft-versus-host disease on survival and relapse.
Haematologica.
83
1998
896
903
12
Brunet
S
Urbano-Ispizua
A
Ojeda
E
et al
Favourable effect of the combination of acute and chronic graft-versus-host disease on the outcome of allogeneic peripheral blood stem cell transplantation for advanced haematological malignancies.
Br J Haematol.
114
2001
544
550
13
Locatelli
F
Uderzo
C
Dini
G
et al
Graft-versus-host disease in children: the AIEOP-BMT Group experience with cyclosporin A.
Bone Marrow Transplant.
12
1993
627
633
14
Eisner
MD
August
CS
Impact of donor and recipient characteristics on the development of acute and chronic graft-versus-host disease following pediatric bone marrow transplantation.
Bone Marrow Transplant.
15
1995
663
668
15
Kondo
M
Kojima
S
Horibe
K
Kato
K
Matsuyama
T
Risk factors for chronic graft-versus-host disease after allogeneic stem cell transplantation in children.
Bone Marrow Transplant.
27
2001
727
730
16
Gilliam
AC
Murphy
GF
Cellular pathology of cutaneous graft-versus-host disease.
Graft-vs.-Host Disease.
Ferrara
JL
Deeg
HJ
Burakoff
SJ
1997
291
313
Marcel Dekker
New York, NY
17
Crawford
JM
Graft-versus-host disease of the liver.
Graft-vs.-Host Disease.
Ferrara
JL
Deeg
HJ
Burakoff
SJ
1997
315
336
Marcel Dekker
New York, NY
18
Mowat
A
Intestinal graft-versus-host disease.
Graft-vs.-Host Disease.
Ferrara
JL
Deeg
HJ
Burakoff
SJ
1997
337
384
Marcel Dekker
New York, NY
19
Glucksberg
H
Storb
R
Fefer
A
et al
Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors.
Transplantation.
18
1974
295
304
20
Shulman
HM
Sullivan
KM
Weiden
PL
et al
Chronic graft-versus-host syndrome in man: a long-term clinicopathological study of 20 Seattle patients.
Am J Med.
69
1980
204
217
21
Ringdén
O
Deeg
HJ
Clinical spectrum of graft-versus-host disease.
Graft-vs.-Host Disease.
Ferrara
JL
Deeg
HJ
Burakoff
SJ
1997
525
559
Marcel Dekker
New York, NY
22
Rossetti
F
Zulian
F
Dall'Amico
R
Messina
C
Montini
G
Zacchello
F
Extracorporeal photochemotherapy as single therapy for extensive, cutaneous, chronic graft-versus-host disease.
Transplantation.
59
1995
149
151
23
Salvaneschi
L
Perotti
C
Zecca
M
et al
Extracorporeal photochemotherapy for treatment of acute and chronic GVHD in childhood.
Transfusion.
41
2001
1299
1305
24
Kaplan
EL
Meier
P
Nonparametral estimation from incomplete observations.
J Am Stat Assoc.
53
1958
457
481
25
Pepe
MS
Longton
G
Pettinger
M
Mori
M
Fisher
LD
Storb
R
Summarizing data on survival, relapse, and chronic graft-versus-host disease after bone marrow transplantation: motivation for and description of new methods.
Br J Haematol.
83
1993
602
607
26
Pepe
MS
Longton
G
Thornquist
M
A qualifier Q for the survival function to describe the prevalence of a transient condition.
Sat Med.
10
1991
413
421
27
Byar
DP
Identification of prognostic factors.
Cancer Clinical Trials: Methods and Practice. Oxford
Buyse
ME
Staquet
MJ
Sylvester
RJ
1988
423
443
Oxford Medical Publications
United Kingdom
28
Cox
DR
Regression models and life tables (with discussion).
J Royal Stat Soc B.
34
1972
187
202
29
Klein JP, Rizzo JD, Zhang M-J, Keiding N. Statistical methods for the analysis and presentation of the results of bone marrow transplants. Vol 28. Part 2: regression modeling. Bone Marrow Transplant. 2001;1001-1011.
30
Rocha
V
Wagner
JE
Sobocinski
KA
et al
Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling.
N Engl J Med.
342
2000
1846
1854
31
Goulmy
E
Termijtelen
A
Bradley
BA
van Rood
JJ
Y-antigen killing by T cells of women is restricted by HLA.
Nature.
266
1977
544
545
32
Goulmy
E
Human minor histocompatibility antigens: new concepts for marrow transplantation and adoptive immunotherapy.
Immunol Rev.
157
1997
125
140
33
Hill
RS
Petersen
FB
Storb
R
et al
Mixed hematologic chimerism after allogeneic marrow transplantation for severe aplastic anemia is associated with a higher risk of graft rejection and a lessened incidence of acute graft-versus-host disease.
Blood.
67
1986
811
816
34
Andreani
A
Manna
M
Lucarelli
G
et al
Persistence of mixed chimerism in patients transplanted for the treatment of thalassemia.
Blood.
87
1996
3494
3499
35
Locatelli
F
Bruno
B
Zecca
M
et al
Cyclosporin A and short-term methotrexate versus cyclosporin A as graft-versus-host disease prophylaxis in patients with severe aplastic anemia given allogeneic bone marrow transplantation from an HLA-identical sibling: results of a GITMO/EBMT randomized trial.
Blood.
96
2000
1690
1697
36
Sykes
M
Eisenthal
A
Sachs
DH
Mechanism of protection from graft-vs-host disease in murine mixed allogeneic chimeras, I: development of a null cell population suppressive of cell-mediated lympholysis responses and derived from the syngeneic bone marrow component.
J Immunol.
140
1988
2903
2911
37
Ferrara
JL
Deeg
HJ
Graft-versus-host disease.
N Engl J Med.
324
1991
667
674
38
Beschorner
WE
Di Gennaro
KA
Hess
AD
Santos
GW
Cyclosporine and the thymus: influence of irradiation and age on thymic immunopathology and recovery.
Cell Immunol.
110
1987
350
364
39
Shinozawa
T
Beschorner
WE
Hess
AD
The thymus and prolonged administration of cyclosporine: irreversible immunopathologic changes associated with autologous pseudo-graft-versus-host disease.
Transplantation.
50
1990
106
111
40
Hess
AD
Thoburn
CJ
Immunobiology and immunotherapeutic implications of syngeneic/autologous graft-versus-host disease.
Immunol Rev.
157
1997
111
123
41
Kollman
C
Howe
CWS
Anasetti
C
et al
Donor characteristics as risk factors in recipients after transplantation of bone marrow from unrelated donors: the effect of age.
Blood.
98
2001
2043
2051
42
Weiden
PL
Sullivan
KM
Flournoy
N
Storb
R
Thomas
ED
Antileukemic effect of chronic graft-versus-host disease.
N Engl J Med.
304
1981
1529
1533
43
Barrett
AJ
van Rhee
F
Graft-versus-leukaemia.
Baillieres Clin Haematol.
10
1997
337
355
44
Locatelli
F
Zecca
M
Rondelli
R
et al
Graft versus host disease prophylaxis with low-dose cyclosporine-A reduces the risk of relapse in children with acute leukemia given HLA-identical sibling bone marrow transplantation: results of a randomized trial.
Blood.
95
2000
1572
1579
45
Ringdén
O
Labopin
M
Gorin
NC
et al
Is there a graft-versus-leukaemia effect in the absence of graft-versus-host disease in patients undergoing bone marrow transplantation for acute leukaemia?
Br J Haematol.
111
2000
1130
1137
46
Sullivan
KM
Witherspoon
RP
Storb
R
et al
Prednisone and azathioprine compared with prednisone and placebo for treatment of chronic graft-v-host disease: prognostic influence of prolonged thrombocytopenia after allogeneic marrow transplantation.
Blood.
72
1988
546
554
47
Wingard
JR
Piantadosi
S
Vogelsang
GB
et al
Predictors of death from chronic graft-versus-host disease after bone marrow transplantation.
Blood.
74
1989
1428
1435
48
Sullivan
KM
Shulman
HM
Storb
R
et al
Chronic graft-versus-host disease in 52 patients: adverse natural course and successful treatment with combination immunosuppression.
Blood.
57
1981
267
276
49
Akpek
G
Zahurak
L
Piantadosi
S
et al
Development of a prognostic model for grading chronic graft-versus-host disease.
Blood.
97
2001
1219
1226
50
Zecca
M
Locatelli
F
Management of graft-versus-host disease in paediatric bone marrow transplant recipients.
Paediatr Drugs.
2
2000
29
55
51
Vogelsang
GB
How I treat chronic graft-versus-host disease.
Blood.
97
2001
1196
1201

Author notes

Marco Zecca, Oncoematologia Pediatrica, IRCCS Policlinico San Matteo, P.le Golgi, 2, I-27100 Pavia, Italy; e-mail:m.zecca@smatteo.pv.it.

Sign in via your Institution