Male recipients of transplants from female (F→M) hematopoietic stem cell donors represent a special group in whom donor T cells that are specific for recipient minor histocompatibility antigens encoded by Y-chromosome genes may contribute to a graft-versus-leukemia (GVL) effect and to graft-versus-host disease (GVHD). We examined the contribution of donor/patient sex to the risk for relapse and GVHD in 3238 patients who underwent HLA-identical sibling hematopoietic stem cell transplantation (HSCT) for hematopoietic malignancies at a single institution. Compared with other sex combinations, male recipients of female transplants had the lowest risk for relapse and the greatest odds for GVHD. Remarkably, after controlling for GVHD as a time-dependent covariate, F→M HSCT still exhibited a lower risk for relapse than other sex combinations, demonstrating a selective GVL effect distinct from that contributed by GVHD. A reduction in relapse after F→M HSCT was observed in patients with chronic myelogenous leukemia (CML), acute myelogenous leukemia (AML), and acute lymphoblastic leukemia (ALL). Taken together, these data suggest that minor H antigens encoded or regulated by genes on the Y chromosome contribute to a selective GVL effect against myeloid and lymphoid leukemias after F→M HSCT.

The efficacy of allogeneic hematopoietic stem cell transplantation (HSCT) for leukemia is in part due to a graft-versus-leukemia (GVL) effect, which is mediated by T cells contained in the stem cell inoculum and is often associated with the development of graft-versus-host disease (GVHD).1-3  The lower risk for leukemic relapse for recipients in whom GVHD develops does not always translate into a survival advantage because these patients are at risk for complications related to GVHD-mediated tissue damage and to immunosuppressive drugs administered for the treatment of GVHD.1,4  Nevertheless, identifying the GVL effect has fostered hope that approaches to allogeneic HSCT based on augmenting immune-mediated antitumor activity can improve outcomes. Examples of efforts to exploit the GVL effect for clinical benefit include the infusion of donor lymphocytes to treat recurrent leukemia after transplantation and the use of low-intensity conditioning regimens that lack significant antitumor activity but sufficiently suppress recipient immunity to allow the engraftment of donor T cells capable of mediating GVL activity.5-10  The results of both interventions have validated the potency of the GVL effect, but, unfortunately, current approaches have not evolved sufficiently to allow the separation of GVL activity from GVHD.7,11 

Studies in murine models of allogeneic HSCT and in humans have implicated recipient minor histocompatibility antigens (minor H antigens) as the targets of donor T cells that mediate antileukemic activity and GVHD.2,12-15  Minor H antigens are peptides derived from cellular proteins encoded by polymorphic genes that differ between the transplant donor and recipient and that are presented to CD8+ and CD4+ T cells by major histocompatibility complex (MHC) class 1 and 2 molecules, respectively. In humans, only a small number of minor H antigens have been molecularly characterized, and the contribution of T cells specific for individual determinants to GVL activity or GVHD remains largely speculative. It has been suggested that minor H antigens, which are limited in their expression to hematopoietic cells including leukemic cells, may be targets for a selective GVL effect, whereas those that are broadly expressed are targets for GVHD.16,17  However, the very large number of minor H antigens that are likely encoded by autosomal genes and are involved in allogeneic responses has complicated analysis of the contribution of individual determinants in the clinical setting.

Transplantation of stem cells from a female donor to a male recipient (F→M HSCT) is a special circumstance in which donor T cells specific for minor H antigens, encoded by genes on the recipient Y chromosome that are polymorphic to their X-chromosome homologues, may make a contribution to GVHD and GVL activity. Four genes on the Y chromosome have already been identified to encode minor H antigens, and the role of these antigens in GVHD and GVL responses is being investigated.18-22  Hematopoietic and nonhematopoietic cells express epitopes that are encoded by SMCY and are presented by HLA-A2 and -B7 to CD8+ T cells, and this development of T-cell responses to SMCY has been implicated in GVHD in small studies of male recipients of HSC transplants from female donors.23,24  In contrast, an HLA–B8-restricted epitope encoded by UTY is not presented to CD8+ T cells by nonhematopoietic cells in vitro but is expressed on hematopoietic cells, including leukemic progenitor cells.20,25  These observations suggest it is possible that some H-Y antigens may be differentially associated with GVHD and GVL responses after transplanting stem cells from female donors into male recipients. The Y-chromosome genes that encode minor H antigens exhibit substantial polymorphism with their X-homologues, suggesting additional epitopes may be presented by other HLA alleles that will also be targets for T cells mediating GVHD or GVL activity after F→M HSCT.

Earlier studies of transplantation outcomes have shown that male recipients of female HLA-identical HSC transplants experience increased GVHD,4,26-28  but these studies have not specifically examined whether such patients exhibit a GVL effect that is independent of GVHD. We analyzed the effect of donor/recipient sex on GVHD and on relapse of the underlying malignancy in a cohort of more than 3000 patients with hematologic malignancies who underwent myeloablative HSCT from an HLA-identical sibling at a single institution.

Patient population

Retrospective analysis was performed on data from 3238 patients who underwent a myeloablative preparative regimen for a hematologic malignancy and who received an unmanipulated HSC transplant from an HLA-identical sibling between 1969 and 2001 at the Fred Hutchinson Cancer Research Center (FHCRC). Patients who expressed at least one of these class 1 HLA alleles—HLA-A1, -A2, -A3, -B7, -B8, -B40, or -B60—were included in the analysis because these alleles are the most frequent in the North American population and have been shown to present at least one of the defined H-Y antigens or are predicted by computer algorithm to bind polymorphic peptides encoded by one or more Y chromosome genes. More than 85% of the patients who underwent HLA-identical HSCT at the FHCRC for hematologic malignancy in the study period expressed one or more of these HLA alleles and were included in the analysis. Patient and donor characteristics are shown in Table 1. Our interest in this study was to evaluate the GVL effect, and we elected to categorize patients based on tumor burden at the time of transplantation because tumor burden has been identified as a factor in the efficacy of immune-mediated tumor regression. Thus, less advanced disease was defined as chronic myelogenous leukemia in chronic phase (CML-CP), acute leukemia in remission, or myelodysplastic syndrome classified as refractory anemia or refractory anemia with ringed sideroblasts. All others were considered more advanced disease. Assessing and grading acute and chronic GVHD were performed as previously described.29,30 

Table 1.

Patient and donor characteristics



Donor/patient sex

F/F
M/F
F/M
M/M
No.   646   586   858   1148  
Median patient age, y (range)   33 (1-72)   32 (1-64)   32 (1-68)   33 (1-68)  
Median donor age, y (range)   33 (1-78)   32 (1-68)   32 (1-70)   32 (1-72)  
Disease     
    CML   170   134   239   319  
    AML   232   219   240   332  
    ALL   121   93   186   205  
    Myelodysplastic syndrome   44   44   61   89  
    Hodgkin disease   15   7   21   25  
    Multiple myeloma   20   26   19   47  
    Non-Hodgkin lymphoma   37   54   68   106  
    Other   7   9   24   25  
Disease status     
    Less advanced   335   300   414   546  
    More advanced   311   286   444   602  
GVHD prophylaxis     
    CSP/MTX   284   284   411   486  
    Other   362   302   447   662  
Conditioning regimen     
    TBI containing   489   439   610   859  
    Non-TBI containing   157   147   248   289  
Patient CMV serostatus     
    Positive   269   276   283   379  
    Negative   197   208   319   401  
    Unknown   180   102   256   367  
Donor CMV serostatus     
    Positive   264   198   308   345  
    Negative   222   285   304   446  
    Unknown
 
160
 
103
 
246
 
357
 


Donor/patient sex

F/F
M/F
F/M
M/M
No.   646   586   858   1148  
Median patient age, y (range)   33 (1-72)   32 (1-64)   32 (1-68)   33 (1-68)  
Median donor age, y (range)   33 (1-78)   32 (1-68)   32 (1-70)   32 (1-72)  
Disease     
    CML   170   134   239   319  
    AML   232   219   240   332  
    ALL   121   93   186   205  
    Myelodysplastic syndrome   44   44   61   89  
    Hodgkin disease   15   7   21   25  
    Multiple myeloma   20   26   19   47  
    Non-Hodgkin lymphoma   37   54   68   106  
    Other   7   9   24   25  
Disease status     
    Less advanced   335   300   414   546  
    More advanced   311   286   444   602  
GVHD prophylaxis     
    CSP/MTX   284   284   411   486  
    Other   362   302   447   662  
Conditioning regimen     
    TBI containing   489   439   610   859  
    Non-TBI containing   157   147   248   289  
Patient CMV serostatus     
    Positive   269   276   283   379  
    Negative   197   208   319   401  
    Unknown   180   102   256   367  
Donor CMV serostatus     
    Positive   264   198   308   345  
    Negative   222   285   304   446  
    Unknown
 
160
 
103
 
246
 
357
 

All patients received HSC transplants at the FHCRC from HLA-identical siblings between 1969 and 2001.

CSP indicates cyclosporine; MTX, methotrexate; TBI, total body irradiation; and CMV, cytomegalovirus.

Statistical analysis

Multivariable logistic regression models were fit to examine the association of donor/patient sex with the probability of acute grades II to IV GVHD. Proportional hazards regression models were fit to examine the association of sex with the risk for failure for time-to-event endpoints (overall mortality, relapse, chronic GVHD). The impact of acute and chronic GVHD on mortality and relapse was assessed by treating GVHD as a time-dependent covariate. Unadjusted survival estimates were obtained using the Kaplan-Meier method. All 2-sided P values associated with regression models were derived from the Wald test, and no adjustments were made for multiple comparisons.

Acute and chronic GVHD

After controlling for patient and donor age, GVHD prophylaxis regimen, conditioning regimen, patient/donor CMV serostatus, and disease status at the time of HSCT, male patients who underwent HSCT with female donors (F→M) had the greatest odds of acquiring grades II to IV acute GVHD among the 4 donor/recipient sex categories. This difference was statistically significant when the F→M subset was compared with F→F or M→M HSCT patients (Table 2). After adjusting for the same variables, the odds ratio of clinical extensive chronic GVHD was also significantly higher in male recipients with female donors compared with recipients in each of the other donor/recipient sex categories (Table 3). In other studies, donor parity has been identified as a risk factor for the development of acute GVHD after HSCT from HLA-identical female donors,4,26,27  though it has not been a risk factor for overall survival after adjusting for other factors.26  The parity data for our population of HSCT patients was incomplete, and we were unable to evaluate their potential contribution to GVHD. However, the F→F HSCT recipients had the lowest odds ratio of acquiring acute and chronic GVHD, and the median age ranges of the donors in the F→F and F→M groups were identical, suggesting it was unlikely that differences in donor parity were solely responsible for the observed differences in GVHD.

Table 2.

Effect of donor/recipient sex on grades II–IV acute GVHD


Donor/recipient sex

Adjusted odds ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   0.86   0.68-1.08   .20  
Female/female   0.55   0.43-0.68   < .0001  
Male/male
 
0.70
 
0.58-0.86
 
.0005
 

Donor/recipient sex

Adjusted odds ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   0.86   0.68-1.08   .20  
Female/female   0.55   0.43-0.68   < .0001  
Male/male
 
0.70
 
0.58-0.86
 
.0005
 

CI indicates confidence interval; NA, not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and donor/recipient CMV serostatus.

Table 3.

Effect of donor/recipient sex on clinical extensive chronic GVHD


Donor/recipient sex

Adjusted odds ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   0.68   0.56-0.82   < .0001  
Female/female   0.63   0.51-0.76   < .0001  
Male/male
 
0.64
 
0.54-0.76
 
< .0001
 

Donor/recipient sex

Adjusted odds ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   0.68   0.56-0.82   < .0001  
Female/female   0.63   0.51-0.76   < .0001  
Male/male
 
0.64
 
0.54-0.76
 
< .0001
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and patient/donor CMV serostatus.

Leukemia relapse

We anticipated that the increased risk for acute and chronic GVHD observed in male recipients of HSC transplants from female donors may be associated with a reduction in the risk for leukemic relapse based on the findings of prior studies that have linked acute and chronic GVHD with the GVL effect.1-3,31  As shown in Table 4, this was in fact the case because male HSCT patients with female donors had a lower risk for relapse compared with all other donor/recipient sex categories. The difference was statistically significant when compared with the M→M and F→F cohorts and reflected a similar trend when compared with female recipients and male donors. The statistical model was then expanded to include acute and chronic GVHD as time-dependent covariates. If acute or chronic GVHD is largely responsible for the reduction in the risk for relapse, then adjusting for GVHD should decrease the hazard ratios summarized in Table 4. Remarkably, the hazard ratios changed very little after controlling for GVHD (Table 5), suggesting that the GVHD effect is not the major driver in the reduction of the risk for relapse among male recipients of transplants from female HSC donors.

Table 4.

Effect of donor/recipient sex on relapse


Donor/recipient sex

Adjusted hazard ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   1.21   0.99-1.48   .06  
Female/female   1.26   1.04-1.52   .02  
Male/male
 
1.38
 
1.17-1.64
 
.0002
 

Donor/recipient sex

Adjusted hazard ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   1.21   0.99-1.48   .06  
Female/female   1.26   1.04-1.52   .02  
Male/male
 
1.38
 
1.17-1.64
 
.0002
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and patient/donor CMV serostatus.

Table 5.

Association of donor/recipient sex and relapse is not solely explained by GVHD


Donor/recipient sex

Adjusted hazard ratio*

95% CI

Pr
Female/male   1   NA   NA  
Male/female   1.20   0.96-1.50   .10  
Female/female   1.27   1.02-1.58   .03  
Male/male
 
1.43
 
1.18-1.73
 
.0003
 

Donor/recipient sex

Adjusted hazard ratio*

95% CI

Pr
Female/male   1   NA   NA  
Male/female   1.20   0.96-1.50   .10  
Female/female   1.27   1.02-1.58   .03  
Male/male
 
1.43
 
1.18-1.73
 
.0003
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, patient/donor CMV serostatus, and acute and chronic GVHD.

Earlier studies have demonstrated that the GVL effect of allogeneic HSCT is greater for patients with chronic myelogenous leukemia (CML) than for patients with acute myelogenous leukemia (AML) or acute lymphoblastic leukemia (ALL). Comparing HSCT male recipients/female donors with the other donor/recipient sex categories showed a lower risk for relapse for patients with CML, AML, and ALL (Table 6). Consistent with findings from earlier studies, our analysis showed the hazard ratio for relapse was lower among patients who underwent transplantation for CML than it was among patients who underwent transplantation forAML orALL, though the confidence interval for each of these groups overlapped, indicating that the differences between the disease categories were consistent with what one might expect to have been caused by chance alone. After adjusting for acute and chronic GVHD, the reduction in the risk for relapse among male patients after HSCT with female donors was diminished among the CML patients but was equivalent in patients with AML or ALL (Table 7).

Table 6.

Effect of donor/recipient sex on relapse among patients who underwent transplantation for CML, AML, and ALL




Adjusted hazard ratio*

95% CI

P
CML    
    Other sex combinations   1   NA   NA  
    Female/male   0.68   0.48-0.98   .04  
AML    
    Other sex combinations   1   NA   NA  
    Female/male   0.87   0.65-1.15   .33  
ALL    
    Other sex combinations   1   NA   NA  
    Female/male
 
0.77
 
0.59-1.02
 
.07
 



Adjusted hazard ratio*

95% CI

P
CML    
    Other sex combinations   1   NA   NA  
    Female/male   0.68   0.48-0.98   .04  
AML    
    Other sex combinations   1   NA   NA  
    Female/male   0.87   0.65-1.15   .33  
ALL    
    Other sex combinations   1   NA   NA  
    Female/male
 
0.77
 
0.59-1.02
 
.07
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and patient/donor CMV serostatus.

Table 7.

Effect of donor/recipient sex on relapse among patients who underwent transplantation for CML, AML, and ALL, controlling for GVHD




Adjusted hazard ratio*

95% CI

P
CML    
    Other sex combinations   1   NA   NA  
    Female/male   0.78   0.53-1.15   .21  
AML    
    Other sex combinations   1   NA   NA  
    Female/male   0.83   0.60-1.16   .27  
ALL    
    Other sex combinations   1   NA   NA  
    Female/male
 
0.75
 
0.54-1.04
 
.09
 



Adjusted hazard ratio*

95% CI

P
CML    
    Other sex combinations   1   NA   NA  
    Female/male   0.78   0.53-1.15   .21  
AML    
    Other sex combinations   1   NA   NA  
    Female/male   0.83   0.60-1.16   .27  
ALL    
    Other sex combinations   1   NA   NA  
    Female/male
 
0.75
 
0.54-1.04
 
.09
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and patient/donor CMV serostatus.

Survival

Increased GVHD and GVL activity would be expected to have opposing effects on survival, with GVHD having a detrimental effect and the reduction in relapse having a beneficial effect. When overall survival was analyzed, female recipients had higher unadjusted survival rates compared with male recipients (Figure 1). After adjusting for the same variables as before, the reduced survival in male recipients was largely confined to the F→M group (Table 8). Male recipients with female donors had a risk for death that was statistically significantly greater than that for recipients in other donor/recipient sex categories, and the odds ratio for M→M HSCT overlapped with that of F→F and M→M (Table 8). The increased risk for death with F→M HSCT was also observed when patients were stratified according to type of leukemia, but the difference is considered to be only a trend among ALL patients (Table 9). Confidence intervals for these disease-specific comparisons overlap, however, indicating that the observed differences among the 3 disease groups are consistent with what would be expected to have been caused by chance alone.

Figure 1.

Probability of survival in transplant recipients over time. Unadjusted overall survival was compared among different donor/patient sex groups as a function of time.

Figure 1.

Probability of survival in transplant recipients over time. Unadjusted overall survival was compared among different donor/patient sex groups as a function of time.

Close modal
Table 8.

Effect of donor/recipient sex on death


Donor/recipient sex

Adjusted hazard ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   0.82   0.72-0.94   .004  
Female/female   0.81   0.72-0.92   .002  
Male/male
 
0.87
 
0.78-0.97
 
.01
 

Donor/recipient sex

Adjusted hazard ratio*

95% CI

P
Female/male   1   NA   NA  
Male/female   0.82   0.72-0.94   .004  
Female/female   0.81   0.72-0.92   .002  
Male/male
 
0.87
 
0.78-0.97
 
.01
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and patient/donor CMV serostatus.

Table 9.

Effect of donor/recipient sex on death among patients who underwent transplantation for CML, AML, and ALL




Adjusted hazard ratio*

95% CI

P
CML    
    Other sex combinations   1   NA   NA  
    Female/male   1.61   1.30-1.99   < .0001  
AML    
    Other sex combinations   1   NA   NA  
    Female/male   1.22   1.03-1.45   .02  
ALL    
    Other sex combinations   1   NA   NA  
    Female/male
 
1.18
 
0.97-1.44
 
.09
 



Adjusted hazard ratio*

95% CI

P
CML    
    Other sex combinations   1   NA   NA  
    Female/male   1.61   1.30-1.99   < .0001  
AML    
    Other sex combinations   1   NA   NA  
    Female/male   1.22   1.03-1.45   .02  
ALL    
    Other sex combinations   1   NA   NA  
    Female/male
 
1.18
 
0.97-1.44
 
.09
 

NA indicates not applicable.

*

Adjusted for patient and donor age, GVHD prophylaxis, disease status, conditioning regimen, and patient/donor CMV serostatus.

Donor T cells play a crucial role in the antileukemic activity of allogeneic HSCT, but they are also responsible for the development of GVHD.1-3,16,32,33  There is substantial interest in identifying the molecular nature of minor H antigens that are targets of GVL responses and GVHD because this may assist in developing strategies for improving the efficacy of transplantation. Genes on the Y chromosome have been identified to encode minor H antigens that may contribute to GVHD and GVL responses in male recipients of stem cell grafts obtained from female donors. Prior studies that have analyzed transplantation outcomes in relation to the sex of the recipients and donors found that male recipients of transplants from female HSC donors had an increased risk for acute and chronic GVHD.1,3  The data reported here in a large number of HLA-matched HSCT patients at a single institution also found that male recipients of grafts from female donors had a significantly higher probability for acute and chronic GVHD than male recipients with male donors or female recipients with female donors. Of interest, female recipients with male donors also had a higher likelihood of GVHD, albeit lower than male recipients with female donors. This finding has been observed previously after HLA-matched, related HSCT,4,26  but it remains unexplained. It has been suggested that proteins encoded by the X chromosome may be selectively expressed in female but not in male cells and that they provide minor H antigens that could be recognized after transplantation in females with cells from male donors.4  An alternative is that autosomal genes regulated by sex hormones could be differentially expressed in males compared with females and could encode minor H antigens. The differential expression of an autosomal gene by donor and recipient cells as a consequence of a gene deletion in the donor has recently been shown to result in the generation of a minor H antigen in humans, providing a precedent for such a mechanism.34 

In addition to the increase in GVHD, we found that male recipients with female donors had a decreased risk for relapse compared with all other sex categories. Gratwohl et al28  have shown that male recipients with CML in first chronic phase who receive grafts from female donors have lower relapse rates in the interval between 2 and 5 years after transplantation than female recipients with female donors. When the patients in our study were stratified based on type of leukemia, a statistically significant reduction in relapse was also observed for male patients with CML who received transplants from female donors compared with other recipient/donor sex categories. In addition, male recipients who underwent transplantation for AML and ALL with female donors exhibited a reduced risk for relapse, and this approached statistical significance for ALL. In contrast to the findings of Gratwohl et al,28,35  we found, when comparing relapse rates of CML-CP among male recipients of transplants from female HSC donors with other recipient/donor sex combinations, there was no departure from the assumption of proportional hazards (data not shown), suggesting that in our cohort the beneficial GVL effect observed was not limited to a particular window of time.

Given the link between recipient/donor sex and GVHD, it was reasonable to hypothesize that the association of recipient/donor sex with risk for relapse was mediated through GVHD. Previous studies have shown that recipients of allogeneic HLA-matched HSCT who do not acquire GVHD are less likely to experience relapses than recipients undergoing autologous HSCT or HSCT from a syngeneic donor,1,36  suggesting there is a GVL effect associated with allografting that is independent of clinical GVHD. We determined the risk for leukemic relapse for each of the recipient/donor sex categories, controlling for the presence of acute and chronic GVHD as time-dependent covariates. Male recipients of HSC transplants from female donors still exhibited a lower risk for relapse compared with other sex combinations, demonstrating that the reduction in relapse in this group could not solely be explained by the presence of GVHD. When the patients were stratified according to type of leukemia, a lower risk for relapse in male recipients with female donors was only significant (P = .04) for patients with CML. Although a similar trend was observed for patients with AML and ALL, these results lacked statistical significance, perhaps because of smaller numbers of patients within each group.

Despite the beneficial GVL effect observed in male recipients with female donors, these patients had significantly reduced survival compared with all patient/donor sex combinations, suggesting that other variables, particularly GVHD, contribute to transplantation-related mortality. Indeed, after controlling for the presence of acute and chronic GVHD, the increased risk for death associated with F→M transplantation was statistically significantly diminished (data not shown). The reduction in survival in male recipients with female donors was greatest for patients with CML, presumably reflecting the lower risk for relapse in these patients, the development of effective therapy for posttransplantation relapse in the past decade, and fewer causes, other than GVHD, of non–relapse-related death.6,7,37  Our data support the choice of a male rather than a female sibling donor for a male transplant recipient, particularly for patients at low risk for relapse. However, there may be situations in which the additional GVL effect associated with using a female donor would be beneficial, and our data suggest that strategies could be developed to augment the GVL effect without GVHD, such as by the adoptive transfer of donor T cells specific for minor H antigens encoded by genes on the Y chromosome.

A potential explanation for the reduction in relapse in male recipients of transplants from female HSC donors that is not attributable to increased GVHD is the presence of minor H antigens encoded or regulated by the Y chromosome that are targets for a selective GVL effect. CD8+ T cells specific for an epitope encoded by the UTY gene and presented by HLA B8 were isolated from a male patient who did not acquire GVHD after receiving a female HLA-identical graft.20  The CD8+ UTY-specific cytotoxic T-lymphocytes (CTLs) lysed hematopoietic target cells in vitro and eliminated AML progenitors in a NOD/SCID mouse model of human leukemia but failed to lyse nonhematopoietic cells in vitro, despite the detection of lowlevel transcription of this gene in nonhematopoietic cells.20  UTY is highly polymorphic with its X homologue. Additional polymorphic UTY peptides that are predicted to bind to other common HLA molecules, including HLA A2, and are recognized by T cells from male recipients of transplants from female HSC donors have recently been described.38  Thus, a careful search for Y-chromosome–encoded minor H antigens may uncover targets that could be exploited for selective GVL activity. An alternative is that minor H antigens may be encoded by autosomal genes that are differentially expressed in male cells because of the presence of male hormones. Androgens have an effect on normal and abnormal hematopoiesis, and a variety of androgen-responsive genes have been described. Thus, the possibility that androgen-responsive genes may be overexpressed in male leukemic cells and may provide antigenic targets for donor T cells cannot be excluded. The selective GVL effect observed among recipients of F→M HSCT in this study provides the impetus for further study of potential mechanisms, including the characterization of HY antigens at the molecular level, to identify potential targets of GVL for use in the immunotherapy of leukemia and to identify those targets responsible for GVHD.

Prepublished online as Blood First Edition Paper, September 11, 2003; DOI 10.1182/blood-2003-07-2603.

Supported by the Robert Wood Johnson Foundation (S.S.B.R.), the Leukemia and Lymphoma Society (S.S.B.R., S.R.R.) the Merck/UNCF Science Initiative (S.S.B.R.), the Damon Runyon Cancer Research Foundation (E.H.W.), and the National Institutes of Health (grant CA18029; S.R.R., F.R.A., T.A.G.).

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
Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
.
1990
;
75
:
555
-562.
2
Weiden PL, Flournoy N, Thomas ED, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts.
N Engl J Med
.
1979
;
300
:
1068
-1073.
3
Weiden PL, Sullivan KM, Flournoy N, Storb R, Thomas ED. Antileukemic effect of chronic graft-versus-host disease: contribution to improved survival after allogeneic marrow transplantation.
N Engl J Med
.
1981
;
304
:
1529
-1533.
4
Nash RA, Pepe MS, Storb R, et al. Acute graft-versus-host disease: analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate.
Blood
.
1992
;
80
:
1838
-1845.
5
McSweeney PA, Niederwieser D, Shizuru JA, et al. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versustumor effects.
Blood
.
2001
;
97
:
3390
-3400.
6
Kolb HJ, Schattenberg A, Goldman JM, et al. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients: European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia.
Blood
.
1995
;
86
:
2041
-2050.
7
Collins RH Jr, Shpilberg O, Drobyski WR, et al. Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation.
J Clin Oncol
.
1997
;
15
:
433
-444.
8
Niederwieser D, Maris M, Shizuru JA, et al. Low-dose total body irradiation (TBI) and fludarabine followed by hematopoietic cell transplantation (HCT) from HLA-matched or mismatched unrelated donors and postgrafting immunosuppression with cyclosporine and mycophenolate mofetil (MMF) can induce durable complete chimerism and sustained remissions in patients with hematological diseases.
Blood
.
2003
;
101
:
1620
-1629.
9
Feinstein LC, Sandmaier BM, Hegenbart U, et al. Non-myeloablative allografting from human leucocyte antigen-identical sibling donors for treatment of acute myeloid leukaemia in first complete remission.
Br J Haematol
.
2003
;
120
:
281
-288.
10
Khouri IF, Keating M, Korbling M, et al. Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies.
J Clin Oncol
.
1998
;
16
:
2817
-2824.
11
Mielcarek M, Martin PJ, Leisenring W, et al. Graft-versus-host disease after nonmyeloablative versus conventional hematopoietic stem cell transplantation.
Blood
.
2003
;
102
:
756
-762.
12
Fontaine P, Roy-Proulx G, Knafo L, Baron C, Roy DC, Perreault C. Adoptive transfer of minor histocompatibility antigen-specific T lymphocytes eradicates leukemia cells without causing graft-versus-host disease.
Nat Med
.
2001
;
7
:
789
-794.
13
Truitt RL, Atasoylu AA. Contribution of CD4+ and CD8+ T cells to graft-versus-host disease and graft-versus-leukemia reactivity after transplantation of MHC-compatible bone marrow.
Bone Marrow Transplant
.
1991
;
8
:
51
-58.
14
Marijt WA, Heemskerk MH, Kloosterboer FM, et al. Hematopoiesis-restricted minor histocompatibility antigens HA-1– or HA-2–specific T cells can induce complete remissions of relapsed leukemia.
Proc Natl Acad Sci U S A
.
2003
;
100
:
2742
-2747.
15
Faber LM, van der Hoeven J, Goulmy E, et al. Recognition of clonogenic leukemic cells, remission bone marrow and HLA-identical donor bone marrow by CD8+ or CD4+ minor histocompatibility antigen-specific cytotoxic T lymphocytes.
J Clin Invest
.
1995
;
96
:
877
-883.
16
Warren EH, Gavin M, Greenberg PD, Riddell SR. Minor histocompatibility antigens as targets for T-cell therapy after bone marrow transplantation.
Curr Opin Hematol
.
1998
;
5
:
429
-433.
17
Mutis T, Goulmy E. Hematopoietic system-specific antigens as targets for cellular immunotherapy of hematological malignancies.
Semin Hematol
.
2002
;
39
:
23
-31.
18
Wang W, Meadows LR, den Haan JM, et al. Human H-Y: a male-specific histocompatibility antigen derived from the SMCY protein.
Science
.
1995
;
269
:
1588
-1590.
19
Meadows L, Wang W, den Haan JM, et al. The HLA-A*0201-restricted H-Y antigen contains a posttranslationally modified cysteine that significantly affects T cell recognition.
Immunity
.
1997
;
6
:
273
-281.
20
Warren EH, Gavin MA, Simpson E, et al. The human UTY gene encodes a novel HLA-B8–restricted H-Y antigen.
J Immunol
.
2000
;
164
:
2807
-2814.
21
Vogt MH, de Paus RA, Voogt PJ, Willemze R, Falkenburg JH. DFFRY codes for a new human male-specific minor transplantation antigen involved in bone marrow graft rejection.
Blood
.
2000
;
95
:
1100
-1105.
22
Vogt MH, van den Muijsenberg JW, Goulmy E, et al. The DBY gene codes for an HLA-DQ5-restricted human male-specific minor histocompatibility antigen involved in graft-versus-host disease.
Blood
.
2002
;
99
:
3027
-3032.
23
Dickinson AM, Wang XN, Sviland L, et al. In situ dissection of the graft-versus-host activities of cytotoxic T cells specific for minor histocompatibility antigens.
Nat Med
.
2002
;
8
:
410
-414.
24
Mutis T, Gillespie G, Schrama E, Falkenburg JH, Moss P, Goulmy E. Tetrameric HLA class I-minor histocompatibility antigen peptide complexes demonstrate minor histocompatibility antigen-specific cytotoxic T lymphocytes in patients with graft-versus-host disease.
Nat Med
.
1999
;
5
:
839
-842.
25
Bonnet D, Warren EH, Greenberg PD, Dick JE, Riddell SR. CD8+ minor histocompatability antigen-specific cytotoxic T lymphocyte clones eliminate human acute myeloid leukemia stem cells.
Proc Natl Acad Sci U S A
.
1999
;
96
:
8639
-8644.
26
Flowers ME, Pepe MS, Longton G, et al. Previous donor pregnancy as a risk factor for acute graft-versus-host disease in patients with aplastic anaemia treated by allogeneic marrow transplantation.
Br J Haematol
.
1990
;
74
:
492
-496.
27
Atkinson K, Farrell C, Chapman G, Downs K, Penny R, Biggs J. Female marrow donors increase the risk of acute graft-versus-host disease: effect of donor age and parity and analysis of cell subpopulations in the donor marrow inoculum.
Br J Haematol
.
1986
;
63
:
231
-239.
28
Gratwohl A, Hermans J, Niederwieser D, van Biezen A, van Houwelingen HC, Apperley J. Female donors influence transplant-related mortality and relapse incidence in male recipients of sibling blood and marrow transplants.
Hematol J
.
2001
;
2
:
363
-370.
29
Przepiorka D, Weisdorf D, Martin P, et al. 1994 Consensus Conference on Acute GVHD Grading.
Bone Marrow Transplant
.
1995
;
15
:
825
-828.
30
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
.
1981
;
57
:
267
-276.
31
Sullivan KM, Weiden PL, Storb R, 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
-1728.
32
Porter DL, Antin JH. The graft-versus-leukemia effects of allogeneic cell therapy.
Annu Rev Med
.
1999
;
50
:
369
-386.
33
Korngold R, Leighton C, Manser T. Graft-versus-myeloid leukemia responses following syngeneic and allogeneic bone marrow transplantation.
Transplantation
.
1994
;
58
:
278
-287.
34
Murata M, Warren EH, Riddell SR. A human minor histocompatibility antigen resulting from differential expression due to a gene deletion.
J Exp Med
.
2003
;
197
:
1279
-1289.
35
Gratwohl A, Brand R, Apperley J, et al. Graft-versus-host disease and outcome in HLA-identical sibling transplantations for chronic myeloid leukemia.
Blood
.
2002
;
100
:
3877
-3886.
36
Ringden 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
.
2000
;
111
:
1130
-1137.
37
Radich JP, Gooley T, Bensinger W, et al. HLA-matched related hematopoietic cell transplantation for chronic-phase CML using a targeted busulfan and cyclophosphamide preparative regimen.
Blood
.
2003
;
102
:
31
-35.
38
Randolph SB, Warren EH, Riddell SR. Identification of novel minor histocompatibility antigens encoded by the Y chromosome gene, UTY [abstract].
Blood
.
2002
;
100
:
615a
.
Sign in via your Institution