• HLA haplotypes encode single nucleotide polymorphisms (SNPs) that are associated with risks after HLA-mismatched unrelated donor HCT.

  • SNPs associated with graft-versus-host disease (GVHD) are independent of those associated with relapse.

Life-threatening risks associated with HLA-mismatched unrelated donor hematopoietic cell transplantation limit its general application for the treatment of blood diseases. The increased risks might be explained by undetected genetic variation within the highly polymorphic major histocompatibility complex (MHC) region. We retrospectively assessed each of 1108 MHC region single nucleotide polymorphisms (SNPs) in 2628 patients and their HLA-mismatched unrelated donors to determine whether SNPs are associated with the risk of mortality, disease-free survival, transplant-related mortality, relapse, and acute and chronic graft-versus-host disease (GVHD). Multivariate analysis adjusted for HLA mismatching and nongenetic variables associated with each clinical end point. Twelve SNPs were identified as transplantation determinants. SNP-associated risks were conferred by either patient or donor SNP genotype or by patient-donor SNP mismatching. Risks after transplantation increased with increasing numbers of unfavorable SNPs. SNPs that influenced acute GVHD were independent of those that affected risk of chronic GVHD and relapse. HLA haplotypes differed with respect to haplotype content of (un)favorable SNPs. Outcome after HLA-mismatched unrelated donor transplantation is influenced by MHC region variation that is undetected with conventional HLA typing. Knowledge of the SNP content of HLA haplotypes provides a means to estimate risks prior to transplantation and to lower complications through judicious selection of donors with favorable MHC genetics.

The availability of >20 million registered unrelated donors worldwide is an invaluable resource for patients seeking a hematopoietic cell transplant to cure life-threatening blood disorders.1  In the United States, white patients have a 75% chance of identifying an HLA-A,-C,-B,-DRB1–matched donor, which improves to 94% if criteria are relaxed to donors with a single HLA mismatch; 31% of African-American patients have HLA-matched and 69% have HLA-mismatched donors.2  Consequently, use of donors with a limited degree of mismatch could greatly increase access of patients to transplantation as therapy. However, HLA-mismatched transplantation is associated with significantly higher risks of graft-versus-host disease (GVHD) and mortality compared with HLA-matched transplantation.3,4  These risks have limited the general use of HLA-mismatched unrelated donors.

A critical unmet need is to increase the safety of HLA-mismatched unrelated donor transplantation by understanding the immunogenetic basis of transplant-associated complications. The major histocompatibility complex (MHC) is an attractive candidate region for the discovery of clinically important human genetic variation because of the high density of genes with immune-related function.5  Single nucleotide polymorphisms (SNPs) travel with HLA genes on haplotypes and have been used to map genes within the MHC that cause disease.6,7  We tested the hypothesis that clinical outcome after HLA-mismatched unrelated donor hematopoietic cell transplantation depends on the cumulative effects of HLA mismatching and of haplotype-associated SNPs.

Study population

Patients transplanted from unrelated donors mismatched at one HLA locus (HLA-A, -C, -B, -DRB1, or -DQB1) for malignant and nonmalignant blood disorders were retrospectively studied (Table 1).8,9  HLA haplotype content for SNPs was defined in 163 HLA-A,-C,-B,-DRB1,-DQB1 homozygyous cell lines10  and HLA-matched11  and HLA-mismatched transplants. Consent from patients and donors was obtained in accordance with the Declaration of Helsinki through the Center for International Blood and Marrow Transplant Research, and the work was approved by the Institutional Review Board, Fred Hutchinson Cancer Research Center.

Table 1

Demographics of the study population

Transplants (N = 2628)
Age, y  
 Patient  
  0-19 642 (24%) 
  20-39 861 (33%) 
  40 and older 1124 (43%) 
  Unknown 1 (<1%) 
 Donor  
  18-19 14 (<1%) 
  20-39 1535 (58%) 
  40 and older 893 (34%) 
  Unknown 186 (7%) 
Year of transplantation  
 1987-1994 349 (13%) 
 1995-1999 661 (25%) 
 2000-2003 681 (26%) 
 2004-2009 937 (36%) 
Patient-donor gender  
 Male-male 910 (35%) 
 Male-female 608 (23%) 
 Female-male 562 (21%) 
 Female-female 547 (21%) 
 Unknown 1 (<1%) 
Disease/early, intermediate, late, or advanced, other or unknown, no.*  
 Acute myeloid leukemia 766 (29%)/216, 246, 296, 8 
 Acute lymphoblastic leukemia 479 (18%)/142, 221, 113, 3 
 Chronic myeloid leukemia 574 (22%)/385, 158, 31 
 Myelodysplastic syndrome 310 (12%)/67, 0, 131, 112 
 Non-Hodgkin lymphoma 150 (6%)/NA 
 Other malignancies 51 (2%)/NA 
 Nonmalignancies 185 (7%)/NA 
Patient-donor serologic status for cytomegalovirus  
 Negative-negative 816 (31%) 
 Negative-positive 799 (30%) 
 Positive-negative 412 (16%) 
 Positive-positive 556 (21%) 
 Unknown 45 (2%) 
Transplant type  
 Myeloablative 2105 (80%) 
 Reduced-intensity/nonmyeloablative 505 (19%) 
 Unknown 18 (1%) 
Source of cells  
 Bone marrow 1726 (66%) 
 Peripheral blood stem cells 902 (34%) 
GVHD prophylaxis  
 Cyclosporine with or without other agents 1244 (47%) 
 Tacrolimus with or without other agents 896 (34%) 
 T-cell depletion 329 (13%) 
 Other combinations 151 (6%) 
 Missing 8 (< 1%) 
Karnofsky performance score (%)  
 0-80 643 (24%) 
 90-100 1614 (61%) 
 Missing 371 (14%) 
Patient-donor HLA-DPB1  
 Matched 187 (7%) 
 GVH mismatch 187 (7%) 
 HVG mismatch 154 (6%) 
 Bidirectional mismatch 1025 (39%) 
 Missing 1075 (41%) 
Patient ethnicity  
 Hispanic 206 (8%) 
 Non-Hispanic  
 White 2141 (81%) 
 African American 139 (5%) 
 Asian/Pacific Islander 56 (2%) 
 Native American 10 (<1%) 
 Other 5 (<1%) 
 Unknown 71 (3%) 
Donor ethnicity  
 Hispanic 159 (6%) 
 Non-Hispanic  
 White 1834 (70%) 
 African American 128 (5%) 
 Asian/Pacific Islander 54 (2%) 
 Native American 27 (1%) 
 Other 57 (2%) 
 Unknown 369 (14%) 
HLA mismatching§  
 HLA-A 658 (25%) 
  Allele 226 
  Antigen 430 
  Unknown 
 HLA-B 351 (13%) 
  Allele 241 
  Antigen 107 
  Unknown 
 HLA-C 1006 (38%) 
  Allele 203 
  Antigen 802 
  Unknown 
 HLA-DRB1 159 (6%) 
  Allele 139 
  Antigen 19 
  Unknown 
 HLA-DQB1 454 (17%) 
  Allele 111 
  Antigen 343 
Transplants (N = 2628)
Age, y  
 Patient  
  0-19 642 (24%) 
  20-39 861 (33%) 
  40 and older 1124 (43%) 
  Unknown 1 (<1%) 
 Donor  
  18-19 14 (<1%) 
  20-39 1535 (58%) 
  40 and older 893 (34%) 
  Unknown 186 (7%) 
Year of transplantation  
 1987-1994 349 (13%) 
 1995-1999 661 (25%) 
 2000-2003 681 (26%) 
 2004-2009 937 (36%) 
Patient-donor gender  
 Male-male 910 (35%) 
 Male-female 608 (23%) 
 Female-male 562 (21%) 
 Female-female 547 (21%) 
 Unknown 1 (<1%) 
Disease/early, intermediate, late, or advanced, other or unknown, no.*  
 Acute myeloid leukemia 766 (29%)/216, 246, 296, 8 
 Acute lymphoblastic leukemia 479 (18%)/142, 221, 113, 3 
 Chronic myeloid leukemia 574 (22%)/385, 158, 31 
 Myelodysplastic syndrome 310 (12%)/67, 0, 131, 112 
 Non-Hodgkin lymphoma 150 (6%)/NA 
 Other malignancies 51 (2%)/NA 
 Nonmalignancies 185 (7%)/NA 
Patient-donor serologic status for cytomegalovirus  
 Negative-negative 816 (31%) 
 Negative-positive 799 (30%) 
 Positive-negative 412 (16%) 
 Positive-positive 556 (21%) 
 Unknown 45 (2%) 
Transplant type  
 Myeloablative 2105 (80%) 
 Reduced-intensity/nonmyeloablative 505 (19%) 
 Unknown 18 (1%) 
Source of cells  
 Bone marrow 1726 (66%) 
 Peripheral blood stem cells 902 (34%) 
GVHD prophylaxis  
 Cyclosporine with or without other agents 1244 (47%) 
 Tacrolimus with or without other agents 896 (34%) 
 T-cell depletion 329 (13%) 
 Other combinations 151 (6%) 
 Missing 8 (< 1%) 
Karnofsky performance score (%)  
 0-80 643 (24%) 
 90-100 1614 (61%) 
 Missing 371 (14%) 
Patient-donor HLA-DPB1  
 Matched 187 (7%) 
 GVH mismatch 187 (7%) 
 HVG mismatch 154 (6%) 
 Bidirectional mismatch 1025 (39%) 
 Missing 1075 (41%) 
Patient ethnicity  
 Hispanic 206 (8%) 
 Non-Hispanic  
 White 2141 (81%) 
 African American 139 (5%) 
 Asian/Pacific Islander 56 (2%) 
 Native American 10 (<1%) 
 Other 5 (<1%) 
 Unknown 71 (3%) 
Donor ethnicity  
 Hispanic 159 (6%) 
 Non-Hispanic  
 White 1834 (70%) 
 African American 128 (5%) 
 Asian/Pacific Islander 54 (2%) 
 Native American 27 (1%) 
 Other 57 (2%) 
 Unknown 369 (14%) 
HLA mismatching§  
 HLA-A 658 (25%) 
  Allele 226 
  Antigen 430 
  Unknown 
 HLA-B 351 (13%) 
  Allele 241 
  Antigen 107 
  Unknown 
 HLA-C 1006 (38%) 
  Allele 203 
  Antigen 802 
  Unknown 
 HLA-DRB1 159 (6%) 
  Allele 139 
  Antigen 19 
  Unknown 
 HLA-DQB1 454 (17%) 
  Allele 111 
  Antigen 343 

Data are n (%). Patients received their transplant at the Fred Hutchinson Cancer Research Center (n = 548) or at 1 of 149 other centers in the Center for International Blood and Marrow Transplant Research network (n = 2080).

GVH, graft-versus-host; HVG, host-versus-graft; NA, not applicable.

*

Disease status before transplant is categorized as early (first complete remission [CR] of acute myeloid leukemia [AML] or acute lymphoblastic leukemia [ALL], first chronic phase [CP] of CML, refractory anemia [RA] or refractory anemia with ring sideroblasts of myelodysplastic syndrome [MDS]); intermediate (second or higher CR of AML or ALL, second or higher CP or accelerated phase of CML); late or advanced (primary induction failure or first or higher relapse of AML or ALL, blast phase [or blast crisis] of CML, MDS RA with excess blasts or excess blasts in transformation); other (mainly unnamed MDS) or unknown. Other malignancies included Hodgkin lymphoma, plasma cell disorder, multiple myeloma, breast cancer, other malignancies. Non-malignancies included severe aplastic anemia, Shwachman-Diamond anemia, Diamond-Blackfan anemia, Fanconi anemia, sickle cell disease, thalassemia, inherited abnormalities of erythrocyte differentiation or function, other immune system disorders, inherited abnormality of platelets, inherited disorder of metabolism, histiocytic disorders and other nonmalignancies.

Myeloablative conditioning regimens: cyclophosphamide (Cy) with total body irradiation (TBI) given as a single dose >500 cGY or as fractionated TBI >800 cGY total; CY/etoposide (VP16)/TBI; busulfan (BU)/CY; TBI ≥ 500 cGY single dose; TBI ≥800 cGY fractionated; melphalan >150 mg/m2; BU >9 mg/kg; BU/melphalan. Reduced intensity regimens: TBI <500 cGY single dose; TBI <800 cGY fractionated; melphalan ≤ 150 mg/m2; BU ≤9 mg/kg; carmustine (BCNU)/VP16/cytarabine/melphalan (BEAM); CY/BCNU/VP16 (CBV); VP16/CY. Nonmyeloblative regimens: TBI 200 cGY; fludarabine (FLU)/TBI 200 cGY; FLU/CY; FLU/cytosine arabinoside (ARA-C).

Definitions follow the US Office of Management and Budget classification.8 

§

HLA allele and antigens were defined according to the official World Health Organization HLA Nomenclature.9  Following this nomenclature, 5 novel sequences identified in patients (A*02, A*03, B*39, B*40, and C*01) and 2 patient-donor mismatch combinations (HLA-DRB1*03:01/03:05 and HLA-B*40:08/40:11) could not be defined.

Genotyping

A total of 5135 samples, including 338 duplicates for intra- and interexperiment quality control, were genotyped for 1228 MHC region SNPs and HLA-A,-C,-B,-DRB1,-DQB1,-DPB1 alleles as previously described.11  The 338 duplicate samples had a concordance rate of 0.991. Of the 4797 unique samples, 187 (3.9%) failed genotyping or gender control, yielding 2628 transplants (1982 patient-donor pairs and 646 patients or donors) for outcomes analysis. Of the 1228 SNPs, 6 did not meet Hardy-Weinberg equilibrium at P < .001, and 114 had interexperimental failures, yielding 1108 SNPs (0.989 genotyping rate) for analysis. To define haplotype content in HLA homozygous samples, SNPs were genotyped using TaqMan chemistry.12 

Statistical analysis

The clinical end points were grades II-IV and III-IV acute GVHD, chronic GVHD, transplant-related mortality, relapse (malignancies), disease-free survival (malignancies), and survival. Before testing SNPs, we built multivariate models for each end point with appropriately selected clinical prognostic factors (Table 1). The strong linkage disequilibrium across the MHC influenced patient-donor SNP mismatch rates (Figure 1). We therefore included separate variables for HLA-A,-C,-B,-DRB1,-DQB1 antigen or allele mismatches, HLA-DPB1 mismatches, patient natural killer cell immunoglobulin–like receptor (KIR) ligands, and patient-donor KIR ligand mismatching.13-15  Assessment of these potential risk factors for outcomes was evaluated using Cox proportional hazards models. First, the proportionality assumption was tested for each factor by adding a time-dependent covariate. Factors that violated proportional hazards were adjusted through stratification. A stepwise forward/backward model selection approach was then performed to identify significant clinical prognostic risk factors at a 5% significance level. To control for HLA variation of particular alleles, we further tested association of each end point with 0, 1, or 2 copies of HLA alleles having ≥20 counts. On the basis of the multivariate clinical variable models, a second stepwise forward selection procedure was performed on the HLA allele variables with a threshold of P = .01 for entering into the preliminary models.

Figure 1

Extent of patient-donor SNP mismatching. The percent of pairs mismatched at each SNP position is shown for transplant pairs mismatched at (A) HLA-A,(B) HLA-B, (C) HLA-C, (D) HLA-DRB1, (E) HLA-DQB1, and (F) HLA-matched transplant pairs.11  Of the 1078 SNPs common to both the HLA-mismatched and the HLA-matched population, there was a higher rate of patient-donor mismatching among HLA-mismatched pairs for 96% of the 1078 SNPs compared with the HLA-matched pairs. The difference between the percent of HLA-mismatched pairs and the percent of HLA-matched pairs11  who were mismatched at each of 1078 SNP positions is graphically represented for (G) HLA-A, (H) HLA-B, (I) HLA-C, (J) HLA-DRB1, and (K) HLA-DQB1 mismatched pairs in this study. Percentiles >0 indicate a higher percent of HLA-mismatched transplants were mismatched at the SNP compared with HLA-matched pairs. Zero percentiles indicate the same percent of HLA-matched and -mismatched pairs were mismatched at the SNP position.

Figure 1

Extent of patient-donor SNP mismatching. The percent of pairs mismatched at each SNP position is shown for transplant pairs mismatched at (A) HLA-A,(B) HLA-B, (C) HLA-C, (D) HLA-DRB1, (E) HLA-DQB1, and (F) HLA-matched transplant pairs.11  Of the 1078 SNPs common to both the HLA-mismatched and the HLA-matched population, there was a higher rate of patient-donor mismatching among HLA-mismatched pairs for 96% of the 1078 SNPs compared with the HLA-matched pairs. The difference between the percent of HLA-mismatched pairs and the percent of HLA-matched pairs11  who were mismatched at each of 1078 SNP positions is graphically represented for (G) HLA-A, (H) HLA-B, (I) HLA-C, (J) HLA-DRB1, and (K) HLA-DQB1 mismatched pairs in this study. Percentiles >0 indicate a higher percent of HLA-mismatched transplants were mismatched at the SNP compared with HLA-matched pairs. Zero percentiles indicate the same percent of HLA-matched and -mismatched pairs were mismatched at the SNP position.

Close modal

In the single SNP analysis, each of the 1108 SNPs was tested separately for associations with the 7 clinical end points using the preliminary models. We considered 0, 1, or 2 copies of the rare SNP allele carried by each patient or donor. Each SNP was tested 3 ways: patient genotype, donor genotype, and patient-donor SNP mismatching. Samples with missing SNP genotypes were excluded. To control for the familywise error rate, the Bonferroni criterion was applied to adjust for the multiple testing of the 1108 SNPs.

We also explored predictive effects from multiple SNPs to each end point. To circumvent the problem of missing genotypes in the model selection on all the SNPs, we adopted a 2-step approach.11  For each outcome, we first selected a subset of SNPs with P < .01 from the single SNP analysis. On the basis of the preliminary models, we then performed a stepwise forward selection procedure on the selected SNPs to build the final predictive models using a threshold P = .001 for the candidate SNPs.

SNP and HLA allele frequency comparisons, and genotypic associations between HLA alleles and SNPs were tested using classical χ2 tests (or Fisher's exact tests for small cell counts). All analyses were performed using SAS version 9.2 (SAS Institute, Cary, NC). Linkage disequilibrium (D′ and r2) between SNPs was estimated using Haploview,16  PLINK version 1.07,17  and R package “Genetics” version 1.3.4.18 

Patient and donor SNP genotypes and mismatching

The minor allele frequencies of the 1108 SNPs were similar to previous reports.19  Compared with HLA-matched pairs,11  HLA-mismatched pairs were more frequently mismatched for more SNPs over longer distances across the MHC (Figure 1A-F). These results confirm the highly disparate nature of the MHC in HLA-mismatched transplant patients and donors. Of the 1078 SNPs common to both the HLA-mismatched and the HLA-matched population, there was a higher rate of patient-donor mismatching among HLA-mismatched pairs for 96% of the 1078 SNPs compared with the HLA-matched pairs. Among the HLA-mismatched pairs, the location of the mismatched SNPs depended on the HLA locus that was mismatched, indicative of the positive linkage disequilibrium characteristic of the MHC.7,20  The difference between the percent of HLA-mismatched pairs and the percent of HLA-matched pairs11  who were mismatched at each of the 1078 SNP positions shows that a higher percent of HLA-mismatched transplants were mismatched at the SNP compared with HLA-matched pairs (Figure 1G-K).

SNPs associated with clinical outcome

In the single SNP analysis, patient genotype at rs429916 was associated with survival (adjusted P = .046), disease-free survival (adjusted P = .046), and transplant-related mortality (adjusted P = .009) after Bonferroni correction. In the multi-SNP analysis, 12 SNPs were associated with transplant outcome (P < .001; Table 2; Figure 2).21  Some end points were influenced by only one SNP (survival, disease-free survival, grades II-IV acute GVHD), whereas others were affected by more than one SNP (relapse, transplant-related mortality, grades III-IV acute GVHD, chronic GVHD). Each SNP influenced clinical outcome through patient genotype, donor genotype, or patient-donor mismatching specifically as noted in Table 2. These data suggest that both patient and donor SNPs contribute to risks after HLA-mismatched transplantation.

Table 2

Summary of SNP associations

OutcomeSNPGene/location*ModelOverall PGenotype or mismatch groupNumber of eventsHR (95% CI)P
Survival rs429916 1.2 kb centromeric of HLA-DOA Patient genotype 7.48 × 10−5 CC 1254/1909  
     AC 201/296 1.11 (0.94-1.31) .23 
     AA 18/19 3.47 (1.95-6.16) 2.27 × 10−5 
Disease-free survival rs429916 1.2 kb centromeric of HLA-DOA Patient genotype 4.28 × 10−5 CC 1147/1708  
     AC 188/268 1.06 (0.88-1.26) .56 
     AA 18/19 3.75 (2.10-6.68) 7.86 × 10−6 
Relapse rs2244546 2.2 kb telomeric of HCP5 Donor genotype 6.92 × 10−4 CC 373/1611  
     CG 117/435 1.19 (0.95-1.50) .14 
     GG 16/30 2.79 (1.60-4.85) 2.87 × 10−4 
 rs986522 COL11A2, intron Donor genotype 3.20 × 10−5 CG 209/1025  
     CC 136/478 1.46 (1.16-1.85) 1.45 × 10−3 
     GG 161/573 1.62 (1.30-2.03) 2.14 × 10−5 
Transplant-related mortality rs915654 1.4 kb telomeric of LTA Patient genotype 9.93 × 10−5 AT 394/964  
     AA 142/300 1.45 (1.16-1.80) 1.16 × 10−3 
     TT 332/718 1.39 (1.17-1.64) 1.43 × 10−4 
 rs429916 1.2 kb centromeric of HLA-DOA Patient genotype 2.82 × 10−5 CC 720/1696  
     AC 134/267 1.20 (0.97-1.49) 9.67 × 10−2 
     AA 14/19 4.52 (2.31-8.86) 1.11 × 10−5 
Grades II-IV acute GVHD§ rs2242656 BAG6, intron Mismatch 3.13 × 10−4 Matched 865/1535  
     HVG 122/214 1.00 (0.82-1.22) 
     GVH 136/203 1.46 (1.21-1.77) 6.92 × 10−5 
Grades III-IV acute GVHD rs209130 3 kb telomeric of TRIM27 Mismatch 6.42 × 10−5 Matched 251/920  
     HVG 154/488 1.22 (0.99-1.50) 6.11 × 10−2 
     GVH 124/420 1.19 (0.96-1.49) .12 
     Bi 45/103 2.17 (1.56-3.01) 3.70 × 10−6 
 rs2075800 HSPA1L, Glu602Lys Patient genotype 8.37 × 10−4 GG 289/848  
     AG 214/819 0.72 (0.60-0.86) 4.01 × 10−4 
     AA 71/264 0.73 (0.56-0.95) 2.07 × 10−2 
 rs394657 NOTCH4, intron Donor 4.15 × 10−4 AA 150/626  
   genotype  AG 301/923 1.48 (1.21-1.81) 1.16 × 10−4 
     GG 123/382 1.43 (1.11-1.82) 4.75 × 10−3 
Chronic GVHD rs2523957 26.6 kb centromeric of HLA-A Mismatch 7.27 × 10−5 Matched 637/1474  
     HVG 69/159 1.21 (0.92-1.59) .18 
     GVH 79/154 1.75 (1.36-2.26) 1.73 × 10−5 
 rs3830076 240 bp telomeric of FKBPL Mismatch 1.24 × 10−4 Matched 674/1499  
     HVG 59/152 0.68 (0.50-0.92) 1.22 × 10−2 
     GVH 52/136 0.56 (0.41-0.77) 3.07 × 10−4 
 rs2071479 HLA-DOB, intron Mismatch 2.27 × 10−4 Matched 724/1648  
     HVG 24/75 0.84 (0.53-1.31) .43 
     GVH 37/64 2.16 (1.48-3.16) 7.02 × 10−5 
 rs107822 711 bp telomeric of RING1 Mismatch 3.98 × 10−4 Matched 378/911  
     HVG 183/391 1.35 (1.11-1.63) 2.17 × 10−3 
     GVH 176/400 1.15 (0.95-1.39) .16 
     Bi 48/85 1.81 (1.30-2.52) 4.95 × 10−4 
OutcomeSNPGene/location*ModelOverall PGenotype or mismatch groupNumber of eventsHR (95% CI)P
Survival rs429916 1.2 kb centromeric of HLA-DOA Patient genotype 7.48 × 10−5 CC 1254/1909  
     AC 201/296 1.11 (0.94-1.31) .23 
     AA 18/19 3.47 (1.95-6.16) 2.27 × 10−5 
Disease-free survival rs429916 1.2 kb centromeric of HLA-DOA Patient genotype 4.28 × 10−5 CC 1147/1708  
     AC 188/268 1.06 (0.88-1.26) .56 
     AA 18/19 3.75 (2.10-6.68) 7.86 × 10−6 
Relapse rs2244546 2.2 kb telomeric of HCP5 Donor genotype 6.92 × 10−4 CC 373/1611  
     CG 117/435 1.19 (0.95-1.50) .14 
     GG 16/30 2.79 (1.60-4.85) 2.87 × 10−4 
 rs986522 COL11A2, intron Donor genotype 3.20 × 10−5 CG 209/1025  
     CC 136/478 1.46 (1.16-1.85) 1.45 × 10−3 
     GG 161/573 1.62 (1.30-2.03) 2.14 × 10−5 
Transplant-related mortality rs915654 1.4 kb telomeric of LTA Patient genotype 9.93 × 10−5 AT 394/964  
     AA 142/300 1.45 (1.16-1.80) 1.16 × 10−3 
     TT 332/718 1.39 (1.17-1.64) 1.43 × 10−4 
 rs429916 1.2 kb centromeric of HLA-DOA Patient genotype 2.82 × 10−5 CC 720/1696  
     AC 134/267 1.20 (0.97-1.49) 9.67 × 10−2 
     AA 14/19 4.52 (2.31-8.86) 1.11 × 10−5 
Grades II-IV acute GVHD§ rs2242656 BAG6, intron Mismatch 3.13 × 10−4 Matched 865/1535  
     HVG 122/214 1.00 (0.82-1.22) 
     GVH 136/203 1.46 (1.21-1.77) 6.92 × 10−5 
Grades III-IV acute GVHD rs209130 3 kb telomeric of TRIM27 Mismatch 6.42 × 10−5 Matched 251/920  
     HVG 154/488 1.22 (0.99-1.50) 6.11 × 10−2 
     GVH 124/420 1.19 (0.96-1.49) .12 
     Bi 45/103 2.17 (1.56-3.01) 3.70 × 10−6 
 rs2075800 HSPA1L, Glu602Lys Patient genotype 8.37 × 10−4 GG 289/848  
     AG 214/819 0.72 (0.60-0.86) 4.01 × 10−4 
     AA 71/264 0.73 (0.56-0.95) 2.07 × 10−2 
 rs394657 NOTCH4, intron Donor 4.15 × 10−4 AA 150/626  
   genotype  AG 301/923 1.48 (1.21-1.81) 1.16 × 10−4 
     GG 123/382 1.43 (1.11-1.82) 4.75 × 10−3 
Chronic GVHD rs2523957 26.6 kb centromeric of HLA-A Mismatch 7.27 × 10−5 Matched 637/1474  
     HVG 69/159 1.21 (0.92-1.59) .18 
     GVH 79/154 1.75 (1.36-2.26) 1.73 × 10−5 
 rs3830076 240 bp telomeric of FKBPL Mismatch 1.24 × 10−4 Matched 674/1499  
     HVG 59/152 0.68 (0.50-0.92) 1.22 × 10−2 
     GVH 52/136 0.56 (0.41-0.77) 3.07 × 10−4 
 rs2071479 HLA-DOB, intron Mismatch 2.27 × 10−4 Matched 724/1648  
     HVG 24/75 0.84 (0.53-1.31) .43 
     GVH 37/64 2.16 (1.48-3.16) 7.02 × 10−5 
 rs107822 711 bp telomeric of RING1 Mismatch 3.98 × 10−4 Matched 378/911  
     HVG 183/391 1.35 (1.11-1.63) 2.17 × 10−3 
     GVH 176/400 1.15 (0.95-1.39) .16 
     Bi 48/85 1.81 (1.30-2.52) 4.95 × 10−4 

The table summarizes the 12 SNPs that met a P < .001 in multivariate models that adjusted for SNPs other than the one of interest.

Bi, bidirectional; GVH, graft-versus-host; HVG, host-versus-graft.

*

Location of SNP according to the National Center for Biotechnology Information dbSNP database.19 

Bidirectional mismatches for SNPs rs2523957 (9 pairs), rs2242656 (10 pairs), rs3830076 (3 pairs), and rs2071479 (2 pairs) were excluded from analysis. Favorable SNP genotypes and favorable SNP (mis)matches are in bold.

Risks of relapse and disease-free survival were defined for patients with malignancies.

§

The effect of the SNP for grades II-IV acute GVHD was significant among HLA-C antigen mismatches (P = .005) and remained significant after adjusting for patient KIR ligands and patient-donor KIR ligand mismatching.

Figure 2

Twelve SNPs of clinical significance in HLA-mismatched unrelated donor transplantation. Each of the 12 SNPs having an association with grades II-IV or III-IV acute GVHD, chronic GVHD, relapse, transplant-related mortality, disease-free survival, or survival are shown on a map of the MHC on chromosome 6p21.3 (not to scale). SNPs are identified by their rs numbers. Chromosome 6 drawing modified from the National Library of Medicine, the National Center for Biotechnology Information public website.21 

Figure 2

Twelve SNPs of clinical significance in HLA-mismatched unrelated donor transplantation. Each of the 12 SNPs having an association with grades II-IV or III-IV acute GVHD, chronic GVHD, relapse, transplant-related mortality, disease-free survival, or survival are shown on a map of the MHC on chromosome 6p21.3 (not to scale). SNPs are identified by their rs numbers. Chromosome 6 drawing modified from the National Library of Medicine, the National Center for Biotechnology Information public website.21 

Close modal

Importance of the number of unfavorable SNPs

Each SNP has either a favorable or unfavorable genotype or a favorable or unfavorable (mis)match status (Table 2). The risks of relapse, transplant-related mortality, grades III-IV acute GVHD, and chronic GVHD each increased with increasing numbers of unfavorable SNPs (Figure 3). Compared with zero unfavorable SNPs for grades III-IV acute GVHD, the effect of 1, 2, and 3 unfavorable SNPs rs209130, rs2075800, and rs394657 was as follows: hazard ratio (HR), 1.58 (95% confidence interval [CI], 1.20-2.09; P = 1.2 × 10−3); HR, 2.17 (95% CI, 1.63-2.88; P = 1.0 × 10−7); and HR, 4.38 (95% CI, 2.44-7.86; P = 7.8 × 10−7), respectively (Figure 3A). The HRs of 1, 2, or ≥3 unfavorable rs2523957, rs3830076, rs2071479, and rs107822 SNPs for chronic GVHD relative to no unfavorable SNP were 1.23 (95% CI, 0.87-1.74; P = .25), 1.72 (95% CI, 1.20-2.44; P = 2.3 × 10−3), and 2.71 (95% CI, 1.80-4.08; P = 1.6 × 10−6), respectively (Figure 3B). Compared with 0 unfavorable SNP, the impact of 1 or 2 unfavorable rs2244546 and rs986522 SNPs on relapse was as follows: HR, 1.62 (95% CI, 1.34-1.97; P = 8.1 × 10−7) and HR, 2.91 (95% CI, 1.40-6.03; P = 4.1 × 10−3), respectively (Figure 3C). For transplant-related mortality, one unfavorable rs915654 or rs429916 SNP was associated with an HR of 1.40 (95% CI, 1.20-1.64; P = 2.4 × 10−5) and 2 SNPs with an HR of 6.18 (95% CI, 2.42-15.78; P = 1.4 × 10−4; Figure 3D).

Figure 3

Impact of total number of unfavorable SNP genotypes on transplant outcome. The impact of the total number of unfavorable SNP genotypes was determined for patients and donors with complete SNP genotyping for all SNPs descriptive of the end point of interest. (A) Effect of unfavorable genotypes at SNPs rs209130, rs2075800, and rs394657 on grades III-IV acute GVHD. (B) Effect of unfavorable genotypes at SNPs rs2523957, rs3830076, rs2071479, and rs107822 on chronic GVHD. (C) Effect of unfavorable genotypes at SNPs rs2244546 and rs986522 on relapse. (D) Effect of unfavorable genotypes at SNPs rs915654 and rs429916 on transplant-related mortality. (E) Effect of 12 unfavorable SNP genotypes on survival.

Figure 3

Impact of total number of unfavorable SNP genotypes on transplant outcome. The impact of the total number of unfavorable SNP genotypes was determined for patients and donors with complete SNP genotyping for all SNPs descriptive of the end point of interest. (A) Effect of unfavorable genotypes at SNPs rs209130, rs2075800, and rs394657 on grades III-IV acute GVHD. (B) Effect of unfavorable genotypes at SNPs rs2523957, rs3830076, rs2071479, and rs107822 on chronic GVHD. (C) Effect of unfavorable genotypes at SNPs rs2244546 and rs986522 on relapse. (D) Effect of unfavorable genotypes at SNPs rs915654 and rs429916 on transplant-related mortality. (E) Effect of 12 unfavorable SNP genotypes on survival.

Close modal

To test the hypothesis that some combinations of 2 or more SNPs may be more detrimental than others, we performed a model selection on the 12 SNPs as defined in Table 2. Two SNPs were associated with survival: rs429916 (P < .0001) and rs915654 (P = .0048). Among the remaining 10 SNPs, combinations of ≥2 unfavorable SNPs conferred similar mortality compared with 0 to 1 unfavorable SNPs (HR, 1.01; 95% CI, 0.88-1.16; P = .86). In contrast, among 885 patients with ≥1 unfavorable rs429916 or rs915654 SNP, the HR of mortality was 1.22 (95% CI, 1.07-1.39; P = .0038) compared with patients with 0 to 1 unfavorable SNPs. These results suggest that mortality is influenced mainly by rs429916 and rs915654.

The selection of donors with favorable SNPs is likely to improve outcomes for patients. For example, transplantation of donors with favorable SNPs for relapse (rs2244546CC and rs986522CG) and for grades III-IV acute GVHD (rs394657AA) is anticipated to lower risks of both complications. Among 331 patients transplanted from donors with rs2244546CC, rs986522CG, and rs394657AA, the HR of relapse was 0.66 (95% CI, 0.49-0.88; P = 4.1 × 10−3), and the HR of grades III-IV acute GVHD was 0.78 (95% CI, 0.61-1.0; P = .05) compared with transplants with all other donor genotypes. These data suggest that the selection of donors with this particular combination of favorable SNP genotypes may help to lower relapse and severe acute GVHD.

SNPs and HLA

All models were adjusted for HLA mismatching to identify SNPs that influence transplant outcome. In turn, patients transplanted from HLA-DQB1 antigen-mismatched donors had lower risks of grades III-IV acute GVHD and transplant-related mortality and improved disease-free survival and survival compared with HLA-C antigen-mismatched transplants (supplemental Table 1), consistent with previous observations.4  Because HLA and SNPs each conferred independent risks, we tested the hypothesis that SNPs and HLA mismatching have additive effects. Among the 790 HLA-C antigen mismatches, the raw proportion of patients who developed grades III-IV acute GVHD was 22% with 0, 31% with 1, 40% with 2, and 45% with 3 unfavorable SNPs rs209130, rs2075800, and rs394657. Similar trends were observed among HLA-DQB1 antigen mismatches for rs209130, rs2075800, and rs394657: 10% of patients with 0, 20% with 1, 33% with 2, and 50% with 3 unfavorable SNPs developed grades III-IV acute GVHD. Therefore, the presence of ≥2 unfavorable SNPs for severe GVHD in HLA-DQB1–mismatched pairs confers risks that are as high as those in HLA-C–mismatched pairs with fewer unfavorable SNPs. In HLA-C– or -DQB1-mismatched transplantation, GVHD risks are conferred independently by the HLA mismatch and by SNP-associated effects. These data suggest that, at a minimum, information on HLA-C and -DQB1 matching and SNPs are needed to assess GVHD risks in individual patients.

Additive effects were most significant among HLA-B (P = 8.58 × 10−4) and HLA-DQB1 (P = .005) antigen mismatches for transplant-related mortality and for DRB1 alleles for grades III-IV acute GVHD (P = .001). The negative effects retained their significance regardless of patient KIR ligands or patient-donor KIR ligand mismatching.13-15  Considering all 12 SNPs, we found cumulative negative effects on survival with each additional unfavorable SNP (Figure 3E, log-rank P = .003). Compared with 0 to 1 unfavorable SNP, the hazards of mortality associated with 2, 3, 4, 5, 6, and ≥7 unfavorable SNPs were as follows: HR, 1.28 (95% CI, 0.84−1.96; P = .25); HR, 1.43 (95% CI, 0.96-2.2; P = .08); HR, 1.68 (95% CI, 1.13-2.50; P = 9.91 × 10−3); HR, 1.86 (95% CI, 1.24-2.79; P = 2.50 × 10−3); HR, 1.63 (95% CI, 1.05-2.53; P = .03); and HR, 2.40 (95% CI, 1.39-4.14; P = 1.7 × 10−3), respectively. These data demonstrate that survival depends on the balance of favorable and unfavorable SNPs that affect relapse and nonrelapse complications and suggest that transplantation of HLA-mismatched donors with fewer unfavorable SNPs will lower risks after transplantation.

If specific patient-donor HLA mismatches are more likely to be associated with specific favorable SNPs, then this information could be used to select among HLA-mismatched donors. Patient-donor mismatching occurred most frequently at HLA-A, -C and -DQB1. The 2 most common HLA-A mismatches, A*02:01 vs A*02:05 and A*03:01 vs A*03:02, had similar frequencies of favorable SNPs at all 12 positions. The 2 most common HLA-C mismatches, C*03:03 vs C*03:04 (n = 96) and C*01:02 vs C*02:02 (n = 46), differed significantly for favorable SNP rs3830076 for chronic GVHD: 58.8% of C*03:03/03:04 mismatches had a favorable SNP rs3830076 in contrast to 26.1% of C*01:02/02:02 mismatches (P = .0003). The 2 most common HLA-DQB1 mismatches, DQB1*03:01 vs DQB1*03:02 (n = 104), and DQB1*02:02 vs DQB1*03:03 (n = 45), differed significantly for favorable SNP rs2075800 for grades III-IV acute GVHD: 74% of DQB1*03:01/03:02 mismatches had a favorable SNP rs2075800 compared with 35.6% of DQB1*02:02/03:03 mismatches (P < .0001). These observations suggest that HLA mismatches differ from one another for favorable SNPs carried on their haplotypes. A more complete understanding of HLA mismatches associated with favorable SNPs may offer a new approach for selecting HLA-mismatched donors in the future.

Acute and chronic GVHD

Chronic GVHD increases morbidity and lowers the quality of life after HLA-mismatched unrelated donor transplantation.22  Although acute GVHD is a known risk factor, patients can develop chronic GVHD without prior acute GVHD; the absence of acute GVHD, therefore, is not a safeguard against chronic GVHD. Because acute and chronic GVHD have different SNP associations, we tested the hypothesis that the SNPs associated with chronic GVHD risk may depend on the presence or absence of acute GVHD. We examined the risk of chronic GVHD using a Cox proportional hazard model wherein grades III-IV acute GVHD was treated as a time-dependent variable. Patients who developed grades III-IV acute GVHD had a higher risk of developing chronic GVHD compared with patients with no acute GVHD (HR, 1.78; 95% CI, 1.49-2.12; P = 1.23 × 10−10). As expected, none of the risk SNPs for acute GVHD influenced the risk of chronic GVHD. However, among the 4 risk SNPs for chronic GVHD, patient-donor mismatching at SNPs rs2523957, rs2071479, and rs3830076 remained independent risk factors for chronic GVHD (P = 3.5 × 10−4, P = 3.9 × 10−4, and P = 1.9 × 10−3, respectively) regardless of grades III-IV acute GVHD. SNP rs107822 (P = .019) was no longer a significant risk factor for chronic GVHD after adjusting for acute GVHD (Table 2), suggesting that its effect on chronic GVHD may depend on the development of acute GVHD. A landmark analysis was also performed on 1630 patients who were evaluable at day 100 after transplant without chronic GVHD. SNPs rs2523957, rs2071479, and rs3830076 remained risk factors for chronic GVHD after adjustment for acute GVHD (P = .013, P = .011, and P = .0043, respectively), whereas SNP rs107822 was no longer significant (P = .085). These results suggest that efforts to match patients and donors at SNPs rs2523957, rs2071479, and rs3830076 may help to lower the risk of chronic GVHD independent of acute GVHD.

GVHD and relapse

GVHD is not always undesirable. The graft-versus-leukemia effect describes the lower risk of relapse in patients with clinical GVHD compared with patients without GVHD.23-26  For individual patients, however, the development of clinical GVHD does not guarantee the patient will be relapse free, nor does the absence of GVHD necessarily predict that relapse is inevitable. To identify SNPs informative for relapse with or without GVHD, we used a Cox proportional hazard model for relapse with time-dependent variables for acute and chronic GVHD. Chronic but not acute GVHD was significantly associated with a lower risk for relapse (HR, 0.62; 95% CI, 0.47-0.82; P < .0008). None of the 8 risk SNPs associated with acute or chronic GVHD met a P < .01 significance level; however, SNPs rs2244546 and rs986522 remained predictive of relapse. Compared with transplants with 0 unfavorable SNPs for relapse, transplants with 1 or 2 unfavorable SNPs had an HR of 1.65 (95% CI, 1.36-2.00; P < .0001). A landmark analysis was performed on 1121 patients evaluable at day 100 without relapse. The effects of SNPs rs2244546 (P = .0054) and rs986522 (P = .0022) on relapse remained similar regardless of the presence or absence of acute GVHD. These results suggest that avoidance of donors with unfavorable SNPs for relapse may help to lower relapse independent of GVHD.

Haplotype content

Of the 12 SNPs, 3 (rs915654, rs2075800, and rs429916) were associated with transplant outcome through the patient’s genotype. We hypothesized that these 3 SNPs define extended HLA haplotypes. Three biallelic SNPs yield 8 possible SNP haplotypes (Table 3). Some haplotypes displayed SNP haplotype diversity, whereas others were unique (supplemental Table 2). These data demonstrate that individuals with the same tissue type may have different extended haplotypes defined by their linked SNPs. Because the total number of favorable rs915654-rs2075800-rs429916 haplotypes depends on both the individual haplotype content and the combination of parental haplotypes, 2 individuals with the same HLA tissue type may have different numbers of favorable SNPs (Figure 4). Therefore, the consequences of HLA-SNP haplotype diversity leads to heterogeneity of transplant risks among patients with the same tissue type.

Table 3

HLA-SNP haplotype content

SNP haplotype
International Histocompatibility Workshop (IHW) reference cell number10 
rs915654*rs2075800rs429916HLA haplotype
A*11:01-C*04:01-B*35:01-DRB1*01:01-DQB1*05:01 09006 
A*02:01-C*05:01-B*44:02-DRB1*04:01-DQB1*03:01 09090 
A*68:02-C*04:01-B*53:01-DRB1*15:03-DQB1*06:02 09010 
A*02:01-C*01:02-B*27:05-DRB1*01:01-DQB1*05:01 09004 
A*02:01-C*14:02-B*51:01-DRB1*08:03-DQB1*03:01 09070 
A*31:01-C*04:01-B*35:01-DRB1*04:01-DQB1*03:01 09025 
A*01:01-C*07:01-B*08:01-DRB1*03:01-DQB1*02:01 09022, 09023, 09086, 09088 
A*03:01-C*07:02-B*07:02-DRB1*15:01-DQB1*06:02 09013, 09017, 09081, 09318 
SNP haplotype
International Histocompatibility Workshop (IHW) reference cell number10 
rs915654*rs2075800rs429916HLA haplotype
A*11:01-C*04:01-B*35:01-DRB1*01:01-DQB1*05:01 09006 
A*02:01-C*05:01-B*44:02-DRB1*04:01-DQB1*03:01 09090 
A*68:02-C*04:01-B*53:01-DRB1*15:03-DQB1*06:02 09010 
A*02:01-C*01:02-B*27:05-DRB1*01:01-DQB1*05:01 09004 
A*02:01-C*14:02-B*51:01-DRB1*08:03-DQB1*03:01 09070 
A*31:01-C*04:01-B*35:01-DRB1*04:01-DQB1*03:01 09025 
A*01:01-C*07:01-B*08:01-DRB1*03:01-DQB1*02:01 09022, 09023, 09086, 09088 
A*03:01-C*07:02-B*07:02-DRB1*15:01-DQB1*06:02 09013, 09017, 09081, 09318 

A total of 163 homozygous individuals were characterized for 12 SNPs of clinical significance. The table shows one representative haplotype for each of the 3 SNP haplotypes defined by rs915654, rs2075800, and rs429916 which were each associated with transplant outcome through the patient’s genotype. A full listing of the 12 SNPs among homozygous individuals is provided in supplemental Table 2.

*

SNP rs915654 was PCR-amplified from 50 ng genomic DNA (0.42 μM of CAGCTCCAACCCCTCTAACA forward and CCTGCTGATACCCTCCAAAG reverse primers; 1× Apex Hot Start Master Mix [Genesee Scientific]) following 15 min at 95°C activation; 30 s at 95°C denaturation, 30 s at 60°C annealing, 2 min at 72°C extension for 30 cycles; 7 min at 72°C extension, and 4°C hold. Amplified products were sequenced with BigDye Terminator v.3.1 Cycle Sequencing kits and the 3730XL DNA analyzer (Applied Biosystems, Foster City, CA).

IHW09070, IHW09086, and IHW09088 were heterozygous at rs429916.

Figure 4

Patient-derived risks are defined by haplotypes. A patient’s haplotypes can be used to assess risks associated with rs915654 (transplant-related mortality), rs2075800 (grades III-IV acute GVHD), and rs429916 (survival, disease-free survival, and transplant-related mortality). The number of favorable genotypes among these three positions depends on the combination of the patient’s maternal and paternal haplotypes. Illustrated are patients whose haplotypes contribute 3, 2, 1, or 0 favorable genotypes at the 3 SNP positions. The patient with ab haplotypes has the TA-GA-CC haplotype at SNPs rs915654, rs2075800, and rs429916, respectively; these genotypes are all favorable (Table 2). The patient with bc haplotypes has 2 favorable (rs2075800AA and rs429916CC) and 1 unfavorable (rs915654AA) genotypes. The patient with de haplotypes has 1 favorable (rs429916CC) and 2 unfavorable (rs915654TT and rs2075800GG) genotypes. The patient with fg haplotypes has all unfavorable genotypes. The total number of favorable SNPs is defined as the combination of the maternal and paternal haplotypes. For example, the presence of HLA-A3-B7-DR15 (a haplotype) with HLA-A2-B44-DR4 (b haplotype) yields 3 favorable (rs915654, rs2075800, and rs429916) genotypes, but the same HLA-A2-B44-DR4 (b haplotype) with HLA-A3-B7-DR11 (c haplotype) yields 2 favorable genotypes. Two individuals with the same HLA tissue type may have different numbers of favorable SNPs because of haplotype diversity. For example, an individual with the HLA-A1,29-B8,44-DR3,7 tissue type can encode either TT-GG-CC (1 favorable SNP; haplotypes de) or TT-GG-AA (0 favorable SNPs; haplotypes fg) as a result of haplotypic diversity (Table 3; supplemental Table 2). Although a patient’s haplotypes cannot be modified, knowledge of the haplotypes can be used for risk assessment, and risks can be lowered through the avoidance of donors with unfavorable genotypes or SNP mismatching.

Figure 4

Patient-derived risks are defined by haplotypes. A patient’s haplotypes can be used to assess risks associated with rs915654 (transplant-related mortality), rs2075800 (grades III-IV acute GVHD), and rs429916 (survival, disease-free survival, and transplant-related mortality). The number of favorable genotypes among these three positions depends on the combination of the patient’s maternal and paternal haplotypes. Illustrated are patients whose haplotypes contribute 3, 2, 1, or 0 favorable genotypes at the 3 SNP positions. The patient with ab haplotypes has the TA-GA-CC haplotype at SNPs rs915654, rs2075800, and rs429916, respectively; these genotypes are all favorable (Table 2). The patient with bc haplotypes has 2 favorable (rs2075800AA and rs429916CC) and 1 unfavorable (rs915654AA) genotypes. The patient with de haplotypes has 1 favorable (rs429916CC) and 2 unfavorable (rs915654TT and rs2075800GG) genotypes. The patient with fg haplotypes has all unfavorable genotypes. The total number of favorable SNPs is defined as the combination of the maternal and paternal haplotypes. For example, the presence of HLA-A3-B7-DR15 (a haplotype) with HLA-A2-B44-DR4 (b haplotype) yields 3 favorable (rs915654, rs2075800, and rs429916) genotypes, but the same HLA-A2-B44-DR4 (b haplotype) with HLA-A3-B7-DR11 (c haplotype) yields 2 favorable genotypes. Two individuals with the same HLA tissue type may have different numbers of favorable SNPs because of haplotype diversity. For example, an individual with the HLA-A1,29-B8,44-DR3,7 tissue type can encode either TT-GG-CC (1 favorable SNP; haplotypes de) or TT-GG-AA (0 favorable SNPs; haplotypes fg) as a result of haplotypic diversity (Table 3; supplemental Table 2). Although a patient’s haplotypes cannot be modified, knowledge of the haplotypes can be used for risk assessment, and risks can be lowered through the avoidance of donors with unfavorable genotypes or SNP mismatching.

Close modal

HLA-mismatched unrelated donor transplantation is a high-risk procedure. We found that clinical outcome after HLA-mismatched transplantation depends on undetected haplotype-linked SNPs that have synergistic effects with HLA mismatching. HLA haplotypes differ with respect to their SNP content. Genetic diversity of HLA-SNP haplotypes leads to heterogeneity of risks even among transplants with the same HLA mismatch. These observations indicate that the overall success of HLA-mismatched unrelated donor transplantation can be improved for future patients through a pretransplant definition of patients’ haplotypes and patient and donor SNP genotypes. The avoidance of donors with multiple unfavorable SNPs may help to lower mortality. A patient’s haplotypes cannot be modified, but if the patient carries high-risk variants, then these patients may benefit from alternative approaches for managing and preventing GVHD. Hence, a paradigm that considers the MHC region genetics of both the patient and the donor may aid in increasing the safety of HLA-mismatched unrelated donor transplantation.

We previously identified two SNPs, rs887464 and rs2281389, that play a role in HLA-matched unrelated donor transplantation.11  In the current study, only rs2281389 showed a suggestive effect for acute GVHD (P = .04). There are several possible reasons for our findings. HLA-mismatched pairs were genetically different from HLA-matched pairs, in that they had higher frequencies of SNP mismatching (Figure 1) and were more ethnically and genetically diverse, encoding 159 unique HLA genotypes that were not present in the HLA-matched population. More importantly, the 2 populations differed significantly for demographic characteristics that influence transplant outcome: patient age, year of transplantation, patient-donor gender, disease diagnosis, GVHD prophylaxis, DPB1 mismatching, and patient-donor ethnicity (P < .0001; Table 1). These observations underscore the importance of the use of cohorts that have similar HLA match status and other demographic variables for validation of the 12 SNPs in the future.

Delineation of the HLA-SNP haplotypes provides a first approximation of genetic diversity among several common North American white haplotypes and contrasts sharply with more conserved SNP haplotypes in other populations.27  Two of the 1108 SNPs in this study were evaluated in Japanese transplants, where mismatching at SNP rs1799964 but not the recipient genotype at SNP rs2075800 was associated with grade IV acute GVHD.28  In our data, mismatching at SNP rs1799964 yielded P = .0054 for grade IV acute GVHD, which did not reach significance after Bonferroni adjustment. The 2 studies evaluated transplants with different ethnicities and known HLA haplotype diversity (North American vs Japanese),27  HLA match status (single mismatches vs all match grades) and study design (SNP discovery vs candidate gene approach). Future studies with sufficiently large numbers of ethnically diverse populations are required to fully evaluate the impact of SNPs in populations with different clinical and demographic characteristics.

SNP haplotype diversity has important clinical implications. Patients with the same tissue type may have different extended HLA-SNP haplotypes that give rise to different numbers of favorable SNPs and different risks (Figure 4). Although a patient’s inherited HLA haplotypes might set the stage for variable degrees of protection or risk, transplant-associated risks can be lessened by the judicious avoidance of donors with unfavorable SNP genotypes or mismatching. Central to the global effort to increase the sharing of donors internationally1  is the need for more complete information on haplotype content of clinically relevant MHC variation in ethnically diverse populations.

When the only option for transplantation is an HLA-mismatched unrelated donor, the criteria for selecting the donor with the least risky HLA mismatch include consideration of the specific HLA locus that is mismatched.29  If certain HLA mismatches are associated with favorable SNPs, then both HLA and SNP haplotypes could aid clinicians in donor selection. We explored whether certain HLA mismatches are more likely to be associated with favorable SNPs. We found that the 2 most frequent HLA-C and HLA-DQB1 mismatches had significantly different frequencies of favorable SNPs for chronic GVHD and acute GVHD, respectively. Interestingly, C*03:03/03:04 is an “allele” mismatch, which, as a group, has historically been associated with lower posttransplant risks compared with “antigen” mismatches such as C*01:01/02:02.3,4  Hence, the features that define a “permissible” HLA mismatch might be highly complex and involve polymorphisms that extend well beyond the physical boundaries of the HLA locus itself. These hypotheses should be amenable to examination in a much larger cohort of HLA-mismatched transplant pairs in the future.

The biological mechanisms that lead to the development of chronic GVHD in the absence of prior acute GVHD are unknown. We observed that patients mismatched at SNPs rs2523957 and rs107822 had a higher risk of chronic GVHD that did not depend on acute GVHD, suggesting that efforts to prospectively match patients and donors at these 2 SNPs will lower risks of chronic GVHD. Regardless of whether patients developed chronic GVHD or not, the risk of relapse depended on the presence of unfavorable SNPs for relapse. These observations may help to explain, in part, why some patients relapse despite clinical GVHD, whereas other patients are spared of both GVHD and relapse. The SNP associations permit further investigation into the genetic basis of GVHD-(in)dependent relapse and afford new possibilities for pretransplant risk assessment and for optimizing individualized treatment of patients.25  Our data may also provide a means to explore GVHD that occurs after autologous or syngeneic transplantation,30  particularly for SNP rs2075800, which was associated with GVHD through the patient’s genotype.

SNPs provide clues to the candidate genes and mechanisms involved in transplant-associated complications. The identification of 2 SNPs within the HLA-DOA and -DOB genes provides strong evidence for a role for antigen processing and presentation in immune responses in HLA-mismatched transplantation.31  The rs2075800G/A SNP defines a glutamic acid/lysine substitution at residue 602 of the heat shock protein-70 A1L molecule. We hypothesize a possible role for the differential binding of peptides by heat shock protein-A1L molecules and/or stimulation of cytokines in GVHD.32  The second SNP marker for grades III-IV acute GVHD, rs394657, resides within the NOTCH4 gene intron and is in positive linkage disequilibrium with nonsynonymous substitutions. Sequence polymorphism of NOTCH4 receptors could influence the inflammatory nature of acute GVHD through altered ligand-receptor binding and production of TNF-α, IFN-γ, IL-4, and IL-17.33  Alternatively, SNP rs394657 might influence GVHD through its role as a putative expression quantitative locus for HLA-DQA1,34  the gene that encodes the DQα chain of the HLA-DQ heterodimer. Differential DQα expression may have consequences for alloantigen recognition in GVHD.

In conclusion, HLA and non-HLA MHC factors contribute to the high morbidity and mortality after HLA-mismatched unrelated donor transplantation. Knowledge of HLA-SNP haplotype-associated risks may provide clinicians with an approach for increasing the safety of HLA-mismatched transplantation. In the future, validation of the SNPs from this study will be feasible with a larger HLA-mismatched transplant experience. Systematic evaluation of HLA haplotypes in other HLA-mismatched settings including haploidentical related and cord blood transplantation may facilitate the investigation of the MHC barrier in transplantation.

The online version of this article contains a data supplement.

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

The authors thank Dr John Klein and Dr Ted Gooley for helpful discussions on biostatistical methods, Gary Schoch for database assistance, and Charlie Du, Mark Gatterman, and Dawn Moran for outstanding technical support.

This work was supported by National Institutes of Health grants AI069197 (E.W.P., M.M., S.R.S.H., M.D.H., M.M.H., T.W.) from the National Institute of Allergy and Infectious Diseases; CA100019 (E.W.P., M.M.), CA18029 (E.W.P., M.M.), and CA76518 (M.M.H.) from the National Cancer Institute; Health Resources and Services Administration grant HHSH234200637015C (M.M.H.), and Office of Naval Research grants N00014-10-1-0204 and N00014-1-1-0339 (S.R.S.H., M.M.H., T.W.).

National Institutes of Health

Contribution: E.W.P. developed the hypotheses and designed the study; E.W.P., M.M., S.R.S., M.D.H., and M.M.H. provided HLA and clinical data; M.M. managed DNA genotyping and data; T.W. performed the statistical analysis; E.W.P., M.M., and T.W. analyzed the data; and all authors contributed to the preparation of the paper and approved the final manuscript.

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

Correspondence: Effie W. Petersdorf, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave North, D4-115, Seattle, WA 98109; e-mail: epetersd@fhcrc.org.

1
Foeken
 
LM
Green
 
A
Hurley
 
CK
, et al. 
Donor Registries Working Group of the World Marrow Donor Association (WMDA)
Monitoring the international use of unrelated donors for transplantation: the WMDA annual reports.
Bone Marrow Transplant
2010
, vol. 
45
 
5
(pg. 
811
-
818
)
2
Petz
 
LD
Spellman
 
SS
Gragert
 
L
Broxmeyer
 
HE
The underutilization of cord blood transplantation: extent of the problem, causes, and methods improvement.
Cord Blood: Biology, Transplantation, Banking, and Regulation
2011
Bethesda, MD
AABB Press
(pg. 
557
-
584
)
3
Petersdorf
 
EW
Hansen
 
JA
Martin
 
PJ
, et al. 
Major-histocompatibility-complex class I alleles and antigens in hematopoietic-cell transplantation.
N Engl J Med
2001
, vol. 
345
 
25
(pg. 
1794
-
1800
)
4
Lee
 
SJ
Klein
 
J
Haagenson
 
M
, et al. 
High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation.
Blood
2007
, vol. 
110
 
13
(pg. 
4576
-
4583
)
5
Trowsdale
 
J
The MHC, disease and selection.
Immunol Lett
2011
, vol. 
137
 
1-2
(pg. 
1
-
8
)
6
International HapMap 3 Consortium, Altshuler DM, Gibbs RA, et al. Integrating common and rare genetic variation in diverse human populations. Nature. 2010;467(7311):52-58.
7
Miretti
 
MM
Walsh
 
EC
Ke
 
X
, et al. 
A high-resolution linkage-disequilibrium map of the human major histocompatibility complex and first generation of tag single-nucleotide polymorphisms.
Am J Hum Genet
2005
, vol. 
76
 
4
(pg. 
634
-
646
)
8
Baker
 
KS
Davies
 
SM
Majhail
 
NS
, et al. 
Race and socioeconomic status influence outcomes of unrelated donor hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
12
(pg. 
1543
-
1554
)
9.
HLA Informatics Group, The Anthony Nolan Trust. HLA nomenclature. Available at: http://hla.alleles.org/wmda/index.html. Accessed May 1, 2012.
10
Mickelson
 
E
Hurley
 
C
Ng
 
J
, et al. 
 
Cell and Gene Bank and reference cell panels. In: Hansen J, ed. Immunobiology of the Human MHC. Seattle, WA: International Histocompatibility Working Group Press; 2006:523-553.
11
Petersdorf
 
EW
Malkki
 
M
Gooley
 
TA
, et al. 
 
MHC resident variation affects risks after unrelated donor hematopoietic cell transplantation. Sci Transl Med. 2012;4(144):144ra101.
12
Livak
 
KJ
Marmaro
 
J
Todd
 
JA
Towards fully automated genome-wide polymorphism screening.
Nat Genet
1995
, vol. 
9
 
4
(pg. 
341
-
342
)
13
Ruggeri
 
L
Capanni
 
M
Urbani
 
E
, et al. 
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
Science
2002
, vol. 
295
 
5562
(pg. 
2097
-
2100
)
14
Hsu
 
KC
Keever-Taylor
 
CA
Wilton
 
A
, et al. 
Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes.
Blood
2005
, vol. 
105
 
12
(pg. 
4878
-
4884
)
15
Cooley
 
S
Weisdorf
 
DJ
Guethlein
 
LA
, et al. 
Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia.
Blood
2010
, vol. 
116
 
14
(pg. 
2411
-
2419
)
16
Barrett
 
JC
Fry
 
B
Maller
 
J
, et al. 
Haploview: analysis and visualization of LD and haplotype maps.
Bioinformatics
2005
, vol. 
21
 
2
(pg. 
263
-
265
)
17
Purcell
 
S
Neale
 
B
Todd-Brown
 
K
, et al. 
PLINK: a tool set for whole-genome association and population-based linkage analyses.
Am J Hum Genet
2007
, vol. 
81
 
3
(pg. 
559
-
575
)
18.
Warnes
 
G
 
Gorjanc G, Leisch F, Man M. Package R “Genetics” 1.3.4. Available at: http://cran.r-project.org/web/packages/genetics/. Accessed August 20, 2008
19.
National Center for Biotechnology Information. dbSNP database. Available at: http://www.ncbi.nlm.nih.gov/projects/SNP/. Accessed June 10, 2012.
20
Horton
 
R
Wilming
 
L
Rand
 
V
, et al. 
Gene map of the extended human MHC.
Nat Rev Genet
2004
, vol. 
5
 
12
(pg. 
889
-
899
)
21
National Library of Medicine
 
National Center for Biotechnology information public website. Available at: http://www.ncbi.nlm.nih.gov/projects/mapview/maps.cgi?taxid=9606&chr=6. Accessed October 25, 2012.
22
Martin
 
PJ
Appelbaum
 
FR
Forman
 
SJ
Negrin
 
RS
, et al. 
Overview of hematopoietic cell transplantation.
Thomas’ Hematopoietic Cell Transplantation
2008
4th Ed
West Sussex, United Kingdom
Wiley-Blackwell
(pg. 
131
-
144
)
23
Horowitz
 
MM
Gale
 
RP
Sondel
 
PM
, et al. 
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
1990
, vol. 
75
 
3
(pg. 
555
-
562
)
24
Ferrara
 
JL
Levine
 
JE
Reddy
 
P
, et al. 
Graft-versus-host disease.
Lancet
2009
, vol. 
373
 
9674
(pg. 
1550
-
1561
)
25
Kolb
 
H-J
Graft-versus-leukemia effects of transplantation and donor lymphocytes.
Blood
2008
, vol. 
112
 
12
(pg. 
4371
-
4383
)
26
Ringdén
 
O
Labopin
 
M
Gorin
 
NC
, et al. 
Acute Leukaemia Working Party of the European Group for Blood and Marrow Transplantation
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
, vol. 
111
 
4
(pg. 
1130
-
1137
)
27
Morishima
 
S
Ogawa
 
S
Matsubara
 
A
, et al. 
Japan Marrow Donor Program
Impact of highly conserved HLA haplotype on acute graft-versus-host disease.
Blood
2010
, vol. 
115
 
23
(pg. 
4664
-
4670
)
28
Harkensee
 
C
Oka
 
A
Onizuka
 
M
, et al. 
Japan Marrow Donor Program
Single nucleotide polymorphisms and outcome risk in unrelated mismatched hematopoietic stem cell transplantation: an exploration study.
Blood
2012
, vol. 
119
 
26
(pg. 
6365
-
6372
)
29
Spellman
 
SR
Eapen
 
M
Logan
 
BR
, et al. 
National Marrow Donor Program
Center for International Blood and Marrow Transplant Research
A perspective on the selection of unrelated donors and cord blood units for transplantation.
Blood
2012
, vol. 
120
 
2
(pg. 
259
-
265
)
30
Hood
 
AF
Vogelsang
 
GB
Black
 
LP
, et al. 
Acute graft-vs-host disease. Development following autologous and syngeneic bone marrow transplantation.
Arch Dermatol
1987
, vol. 
123
 
6
(pg. 
745
-
750
)
31
Busch
 
R
Rinderknecht
 
CH
Roh
 
S
, et al. 
Achieving stability through editing and chaperoning: regulation of MHC class II peptide binding and expression.
Immunol Rev
2005
, vol. 
207
 
1
(pg. 
242
-
260
)
32
Fourie
 
AM
Peterson
 
PA
Yang
 
Y
Characterization and regulation of the major histocompatibility complex-encoded proteins Hsp70-Hom and Hsp70-1/2.
Cell Stress Chaperones
2001
, vol. 
6
 
3
(pg. 
282
-
295
)
33
Zhang
 
Y
Sandy
 
AR
Wang
 
J
, et al. 
Notch signaling is a critical regulator of allogeneic CD4+ T-cell responses mediating graft-versus-host disease.
Blood
2011
, vol. 
117
 
1
(pg. 
299
-
308
)
34
SNP and CNV annotation database (SCAN). Available at: http://www.scandb.org/newinterface/about.html. Accessed June 12, 2012.

Supplemental data

Sign in via your Institution