Donor-directed human leukocyte antigen (HLA)–specific allo-antibodies (DSAs) cause graft failure in animal models of hematopoietic stem cell transplantation (HCT). Archived pretransplantation sera from graft failure patients (n = 37) and a matched case-control cohort (n = 78) were tested to evaluate the role of DSAs in unrelated donor HCT. Controls were matched for disease, disease status, graft type, patient age, and transplantation year. Patients had acute myeloid leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, or myelodysplastic syndrome; 98% received myeloablative conditioning regimens 100% received T-replete grafts, 97% received marrow, 95% HLA-mismatched, and 97% received calcineurin-based graft-versus-host disease prophylaxis. Among the 37 failed transplantations, 9 (24%) recipients possessed DSAs against HLA-A, B, and/or DP, compared with only 1 (1%) of 78 controls. Therefore, the presence of DSAs was significantly associated with graft failure (odds ratio = 22.84; 95% confidence interval, 3.57-∞; P < .001). These results indicate that the presence of pretransplantation DSAs in recipients of unrelated donor HCT is associated with failed engraftment and should be considered in HCT donor selection.

Hematopoietic stem cell transplantation (HCT) recipients may become alloimmunized to foreign human leukocyte antigens (HLAs) through pregnancy or blood transfusions. The resulting sensitization may include antibodies directed against mismatched HLA antigens of a potential stem cell donor. Recent National Marrow Donor Program (NMDP) analyses suggest that greater than 50% of unrelated donor HCTs are mismatched for at least one classic HLA-A, B, C, or DRB1 locus.1,2  In addition, mismatching at HLA-DP is observed in approximately 88% of all unrelated donor HSCT.1,2  Engraftment failure is observed at a rate of approximately 5% in unrelated donor HCT, and donor-directed HLA alloantibodies may increase the risk.3  In a murine model of allo-sensitization, rapid graft failure was shown to result from alloimmune rejection mediated by antibody-dependent cell-mediated killing.4  Prescreening of patient serum and the identification of specific HLA antibodies could be used as part of a donor selection strategy designed to avoid a potential deleterious incompatibility.

Only a few studies have demonstrated that recipient sensitization to mismatched donor HLA antigens affects engraftment. In a study of marrow transplantations from HLA-mismatched relatives, graft failure occurred in 13 of 21 patients (62%) with a positive pretransplantation cross-match (patient serum vs donor T or B lymphocytes), compared with 31 of 501 patients (7%) with a negative cross-match (P < .001).5,6  Ottinger et al also found that a positive lymphocyte cross-match was a predictor for graft failure and poor survival after HCT from HLA-mismatched donors.7 

Although a lymphocyte cross-match is an effective tool to evaluate alloimmunization and potential donor/recipient incompatibility, the procedure is labor intensive, may detect non-HLA antibodies, and is logistically difficult for remotely located unrelated donors because of the requirement for live cells. Non-HLA antibodies may be important in HCT; however, studies have not been done that support this point unequivocally. New solid-phase antibody detection technologies can better identify HLA-specific alloantibodies and are more sensitive than cytotoxicity testing and flow cytometry.8-11  Using these methods, it may be possible to predict alloreactivity against HLA mismatches for unrelated donor recipient pairs before transplantation. Takanashi et al have reported that virtual cross-match–detected DSAs predict graft failure of unrelated umbilical cord blood transplantation.12 

We designed a case-control study to retrospectively evaluate the effect of preexisting DSAs on engraftment in unrelated donor HSCT. Thirty-seven cases with available samples were selected based on the failure to achieve engraftment after transplantation and 78 controls that engrafted were selected for comparison. Cases and controls were matched for disease, disease status, patient age, year of transplantation, conditioning regimen, and graft type. Graft failure was defined as never achieving an absolute neutrophil count more than 500 with survival beyond 28 days. Patients had acute myeloid leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, or myelodysplastic syndrome; 98% received myeloablative conditioning regimens, 100% received T-replete grafts, 97% received marrow, and 97% received calcineurin-based graft-versus-host disease prophylaxis (Table 1). Cases and controls were preferentially selected for the presence of at least one HLA mismatch at HLA-A, -B, -C, DRB1, DQB1, or DPB1 to serve as a potential allogeneic target. All HLA typing was verified using high-resolution DNA-based methods as described previously.13 

Table 1

Characteristics of patients included in the study using cases (graft failure) versus controls (engraftment)

VariableGraft failure
Engraftment
No. evaluatedNo. (%)No. evaluatedNo. (%)
No. of patients  37  78 
No. of centers  22  33 
Median age, y (range) 37 35 (7-53) 78 36 (5-54) 
Age at transplantation, y 37  78  
    Less than 10  1 (3)  1 (1) 
    11-20  4 (11)  9 (12) 
    21-30  9 (24)  18 (23) 
    31-40  11 (30)  22 (28) 
    41-50  10 (27)  24 (31) 
    More than 50  2 (5)  4 (5) 
Male sex 37 17 (46) 78 45 (58) 
Karnofsky before treatment > 90% 36 26 (72) 76 51 (67) 
Overall HLA matching 37  78  
    12/12  2 (5)  4 (5) 
    11/12  4 (11)  11 (14) 
    10/12  6 (16)  26 (33) 
    9/12  11 (30)  18 (23) 
    8/12  7 (19)  8 (10) 
    7/12  5 (14)  6 (8) 
    6/12  2 (5)  5 (7) 
HLA mismatch distribution     
    HLA-A  13 (35)  25 (32) 
    HLA-B  13 (35)  25 (32) 
    HLA-C  22 (59)  38 (49) 
    HLA-DRB1  3 (8)  8 (10) 
    HLA-DQB1  10 (27)  13 (17) 
    HLA-DPB1  33 (89)  63 (81) 
Disease at transplantation 37  78  
    AML  12 (32)  26 (33) 
    ALL  2 (5)  5 (6) 
    CML  20 (54)  41 (53) 
    MDS  3 (8)  6 (8) 
Disease status at transplantation 37  78  
    Early  13 (35)  28 (36) 
    Intermediate  14 (38)  30 (38) 
    Advanced  9 (24)  19 (24) 
    Other  1 (3)  1 (1) 
Stem cell source 37  78  
    Bone marrow  36 (97)  76 (97) 
    PBSC  1 (3)  2 (3) 
Conditioning regimen 37  78  
    Myeloablative  36 (97)  76 (97) 
    Reduced intensity  1 (3)  2 (3) 
GVHD prophylaxis 37  78  
    CsA or FK506 + MTX ± other  36 (97)  73 (93) 
    CsA or FK506 ± other (no MTX)   2 (3) 
    MTX ± other  1 (3)  2 (3) 
    Other   1 (1) 
Donor/recipient sex match 37  78  
    Male/male  5 (14)  31 (40) 
    Male/female  12 (32)  20 (26) 
    Female/male  12 (32)  14 (18) 
    Female/female  8 (22)  13 (17) 
Donor/recipient CMV match 37  78  
    Negative/negative  4 (11)  29 (37) 
    Negative/positive  15 (41)  24 (31) 
    Positive/negative  10 (27)  7 (9) 
    Positive/positive  5 (14)  17 (22) 
    Unknown  3 (8)  1 (1) 
Donor median age, y (range) 37 36 (21-52) 78 35 (18-54) 
Donor age, y 37  78  
    18-29  11 (30)  21 (27) 
    30-39  12 (32)  35 (45) 
    40-49  13 (35)  14 (18) 
    50 and older  1 (3)  8 (10) 
Year of transplantation 37  78  
    1990   1 (1) 
    1991   
    1992  1 (3)  6 (8) 
    1993  3 (8)  7 (9) 
    1994  3 (8)  7 (9) 
    1995  6 (16)  5 (6) 
    1996  5 (14)  10 (13) 
    1997  4 (11)  6 (8) 
    1998  2 (5)  8 (10) 
    1999  5 (14)  8 (10) 
    2000  3 (8)  11 (14) 
    2001  5 (14)  7 (9) 
    2002   2 (3) 
Median follow-up of survivors, mo 72 (70-74) 21 65 (38-158) 
VariableGraft failure
Engraftment
No. evaluatedNo. (%)No. evaluatedNo. (%)
No. of patients  37  78 
No. of centers  22  33 
Median age, y (range) 37 35 (7-53) 78 36 (5-54) 
Age at transplantation, y 37  78  
    Less than 10  1 (3)  1 (1) 
    11-20  4 (11)  9 (12) 
    21-30  9 (24)  18 (23) 
    31-40  11 (30)  22 (28) 
    41-50  10 (27)  24 (31) 
    More than 50  2 (5)  4 (5) 
Male sex 37 17 (46) 78 45 (58) 
Karnofsky before treatment > 90% 36 26 (72) 76 51 (67) 
Overall HLA matching 37  78  
    12/12  2 (5)  4 (5) 
    11/12  4 (11)  11 (14) 
    10/12  6 (16)  26 (33) 
    9/12  11 (30)  18 (23) 
    8/12  7 (19)  8 (10) 
    7/12  5 (14)  6 (8) 
    6/12  2 (5)  5 (7) 
HLA mismatch distribution     
    HLA-A  13 (35)  25 (32) 
    HLA-B  13 (35)  25 (32) 
    HLA-C  22 (59)  38 (49) 
    HLA-DRB1  3 (8)  8 (10) 
    HLA-DQB1  10 (27)  13 (17) 
    HLA-DPB1  33 (89)  63 (81) 
Disease at transplantation 37  78  
    AML  12 (32)  26 (33) 
    ALL  2 (5)  5 (6) 
    CML  20 (54)  41 (53) 
    MDS  3 (8)  6 (8) 
Disease status at transplantation 37  78  
    Early  13 (35)  28 (36) 
    Intermediate  14 (38)  30 (38) 
    Advanced  9 (24)  19 (24) 
    Other  1 (3)  1 (1) 
Stem cell source 37  78  
    Bone marrow  36 (97)  76 (97) 
    PBSC  1 (3)  2 (3) 
Conditioning regimen 37  78  
    Myeloablative  36 (97)  76 (97) 
    Reduced intensity  1 (3)  2 (3) 
GVHD prophylaxis 37  78  
    CsA or FK506 + MTX ± other  36 (97)  73 (93) 
    CsA or FK506 ± other (no MTX)   2 (3) 
    MTX ± other  1 (3)  2 (3) 
    Other   1 (1) 
Donor/recipient sex match 37  78  
    Male/male  5 (14)  31 (40) 
    Male/female  12 (32)  20 (26) 
    Female/male  12 (32)  14 (18) 
    Female/female  8 (22)  13 (17) 
Donor/recipient CMV match 37  78  
    Negative/negative  4 (11)  29 (37) 
    Negative/positive  15 (41)  24 (31) 
    Positive/negative  10 (27)  7 (9) 
    Positive/positive  5 (14)  17 (22) 
    Unknown  3 (8)  1 (1) 
Donor median age, y (range) 37 36 (21-52) 78 35 (18-54) 
Donor age, y 37  78  
    18-29  11 (30)  21 (27) 
    30-39  12 (32)  35 (45) 
    40-49  13 (35)  14 (18) 
    50 and older  1 (3)  8 (10) 
Year of transplantation 37  78  
    1990   1 (1) 
    1991   
    1992  1 (3)  6 (8) 
    1993  3 (8)  7 (9) 
    1994  3 (8)  7 (9) 
    1995  6 (16)  5 (6) 
    1996  5 (14)  10 (13) 
    1997  4 (11)  6 (8) 
    1998  2 (5)  8 (10) 
    1999  5 (14)  8 (10) 
    2000  3 (8)  11 (14) 
    2001  5 (14)  7 (9) 
    2002   2 (3) 
Median follow-up of survivors, mo 72 (70-74) 21 65 (38-158) 

AML indicates acute myeloid leukemia; ALL, acute lymphoblastic leukemia; CML, chronic myelogenous leukemia; MDS, myelodysplastic syndrome; and MTX, methotrexate.

Pretransplantation recipient serum samples were obtained from the NMDP Research Repository. All surviving recipients included in this analysis were retrospectively contacted and provided informed consent for participation in the NMDP research program. Research was approved and conducted under supervision of the NMDP Institutional Review Board. A modeling process was used as previously described to adjust for any bias introduced by exclusion of nonconsenting survivors.1 

The sera were tested in 2 different laboratories using solid-phase microparticle methods with 10% tested in duplicate for quality control purposes. HLA antibody screening was initially performed on all samples by flow cytometry using FlowPRA (One Lambda, Inc). All samples with a positive panel reactive antibody (PRA) were further evaluated using the Luminex-based LABScreen single antigen assay or single antigen flow beads (One Lambda Inc) to determine HLA specificities. Samples were considered positive for specific HLA antigens based on a background-adjusted mean fluorescence intensity of more than 2000 for Luminex-tested samples or a mean fluorescence channel shift of more than 40 for single antigen flow bead assays. DSA assessments were performed by comparing the HLA antibody profile with the mismatched HLA antigens in the graft-rejection direction.

Conditional logistic regression analysis was performed to compare the PRA and DSA positivity rates between the graft failure cases and engrafted controls. Cox regression models were developed to test the impact of HLA-C mismatches, cytomegalovirus (CMV), and cell dose on graft failure rates, as these were not included in the original matching schema.

The PRA testing found that 37% of all patients carried preexisting HLA class I or II antibodies before transplantation. Forty-three percent of graft failure cases and 32% of engraftment controls possessed a positive PRA, and the distribution by class I and II is noted in Table 2. There was no significant difference in PRA positivity rates between the 2 groups (χ2, P = .221) or an association found between PRA and graft failure using conditional logistic regression (data not shown).

Table 2

Results of univariate analysis comparing the rates of panel-reactive antibody positivity and donor-specific HLA antibody positivity between graft failure cases and engrafted controls

Class I positive, no. (%)Class II positive, no. (%)Class I and/or II positive, no. (%)P
Panel-reactive antibody results    > .221 
    Cases (n = 37) 16 (43) 10 (27) 16 (43)  
    Controls (n = 78) 21 (27) 13 (17) 25 (32)  
Donor-specific HLA antibodies    < .001 
    Cases (n = 37)     
 A: 1 (3) DP: 6 (16) 9 (24)  
 B: 4 (11) DR: 0 (0)   
 C: 0 (0) DQ: 0 (0)   
    Controls (n = 78)     
 A: 1 (1) DP: 1 (1) 1 (1)  
 B: 0 (0) DR: 0 (0)   
 C: 0 (0) DQ: 0 (0)   
Class I positive, no. (%)Class II positive, no. (%)Class I and/or II positive, no. (%)P
Panel-reactive antibody results    > .221 
    Cases (n = 37) 16 (43) 10 (27) 16 (43)  
    Controls (n = 78) 21 (27) 13 (17) 25 (32)  
Donor-specific HLA antibodies    < .001 
    Cases (n = 37)     
 A: 1 (3) DP: 6 (16) 9 (24)  
 B: 4 (11) DR: 0 (0)   
 C: 0 (0) DQ: 0 (0)   
    Controls (n = 78)     
 A: 1 (1) DP: 1 (1) 1 (1)  
 B: 0 (0) DR: 0 (0)   
 C: 0 (0) DQ: 0 (0)   

Screening for the presence of DSAs in the PRA-positive graft failure cases and controls found that 8.7% were positive. Among the cases, 24% (9 of 37) carried DSAs, 3 with class I only, 4 with class II only, and 2 with both class I and II specific antibodies (Table 2). In the control group, only one case was positive for DSA (class I and II). The DSA positivity rates were significantly different between the groups (χ2, P < .001). The conditional logistic regression analysis found that the presence of class I (odds ratio [OR] = 11.34; 95% confidence interval [CI], 1.49-∞; P = .017), class II (OR = 12.00; 95% CI, 1.46-551.97; P = .014), or either class I or II (OR = 22.84; 95% CI, 3.57-∞; P < .001) DSAs in the recipients before transplantation was significantly associated with graft failure (Table 3).

Table 3

Results of conditional logistic regression analysis evaluating the association of DSA directed against HLA class I and/or II and graft failure

Odds ratio95% confidence intervalP
Class I DSA 11.34 1.49-∞ .017 
Class II DSA 12.00 1.46-551.97 .014 
Class I and/or II DSA 22.84 3.57-∞ < .001 
Odds ratio95% confidence intervalP
Class I DSA 11.34 1.49-∞ .017 
Class II DSA 12.00 1.46-551.97 .014 
Class I and/or II DSA 22.84 3.57-∞ < .001 

Further analyses were conducted using Cox regression models to evaluate a limited set of covariates not accounted for in the case-control matching, including patient CMV status, cell dose, and HLA-C match, because these variables had been associated with graft failure in a previous study of unrelated donor transplantations.14  Cell dose (< 2 × 109/kg) and CMV status (recipient positive) were independently predictive of engraftment (P = .01 and .03, respectively). No effect was observed for HLA-C match (P = .84). The presence of antidonor HLA class I or II antibodies was still predictive of engraftment failure when adjustment was made for either cell dose (OR = 15.49; 95% CI, 2.06-697.83; P = .002) or CMV status (OR = 7.94; 95% CI, 0.97-367.84; P = .05).

Interestingly, antibodies against HLA-DP were quite prominent and were present in 60% of antibody-positive failures (Table 2). HLA-DP mismatching has previously been implicated as affecting graft-versus-host disease and lower relapse rates in the T-cell replete myeloablative bone marrow transplantation setting, suggesting its ability to act as a transplantation antigen.1,15  This suggests that, even though matching for HLA-DP may not be important for overall survival, if a patient has HLA antibodies directed against the mismatched DP type of the donor, there may be an increased risk for graft failure. Although it is possible that a higher cell dose, such as provided by a peripheral blood stem cell graft, could help overcome the engraftment barrier posed by DSAs, there are no clinical data available to define a cell dose threshold. Thus, HLA antibody screening is indicated for potential HCT recipients. Moreover, if HLA antibody is present, a thorough assessment of antibody specificity and donor mismatches is warranted before transplantation.

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 Center for International Blood and Marrow Transplant Research is supported by the National Cancer Institute, the National Heart, Lung and Blood Institute, and the National Institute of Allergy and Infectious Diseases (Public Health Service Grant/Cooperative Agreement U24-CA76518); the National Heart, Lung and Blood Institute and National Cancer Institute (Grant/Cooperative Agreement 5U01HL069294); the Health Resources and Services Administration (Department of Health and Human Services; contract HHSH234200637015C); the Office of Naval Research (grants N00014-06-1-0704 and N00014-08-1-0058); and grants from AABB; Aetna; American Society for Blood and Marrow Transplantation; Amgen Inc; anonymous donation to the Medical College of Wisconsin; Astellas Pharma US Inc; Baxter International Inc; Bayer HealthCare Pharmaceuticals; Be the Match Foundation; Biogen IDEC; BioMarin Pharmaceutical Inc; Biovitrum AB; BloodCenter of Wisconsin; Blue Cross and Blue Shield Association; Bone Marrow Foundation; Canadian Blood and Marrow Transplant Group; CaridianBCT; Celgene Corporation; CellGenix GmbH; Centers for Disease Control and Prevention; Children's Leukemia Research Association; ClinImmune Labs; CTI Clinical Trial and Consulting Services; Cubist Pharmaceuticals; Cylex Inc; CytoTherm; DOR = BioPharma Inc; Dynal Biotech, an Invitrogen Company; Eisai Inc; Enzon Pharmaceuticals Inc; European Group for Blood and Marrow Transplantation; Gamida Cell Ltd; GE Healthcare; Genentech Inc; Genzyme Corporation; Histogenetics Inc; HKS Medical Information Systems; Hospira Inc; Infectious Diseases Society of America; Kiadis Pharma; Kirin Brewery Co Ltd; Leukemia & Lymphoma Society; Merck & Company; Medical College of Wisconsin; MGI Pharma Inc; Michigan Community Blood Centers; Millennium Pharmaceuticals Inc; Miller Pharmacal Group; Milliman USA Inc; Miltenyi Biotec Inc; National Marrow Donor Program; Nature Publishing Group; New York Blood Center; Novartis Oncology; Oncology Nursing Society; Osiris Therapeutics Inc; Otsuka America Pharmaceutical Inc; Pall Life Sciences; PDL BioPharma Inc; Pfizer Inc; Pharmion Corporation; Saladax Biomedical Inc; Schering Corporation; Society for Healthcare Epidemiology of America; StemCyte Inc; StemSoft Software Inc; Sysmex America Inc; Teva Pharmaceutical Industries; THERAKOS Inc; Thermogenesis Corporation; Vidacare Corporation; Vion Pharmaceuticals Inc; ViraCor Laboratories; ViroPharma Inc; and Wellpoint Inc.

The views expressed in this article do not reflect the official policy or position of the National Institutes of Health, the Department of the Navy, the Department of Defense, or any other agency of the US government.

National Institutes of Health

Contribution: S.S., R.B., and C.A. participated in the conception and design of this study and wrote the paper; S.S., S.F., M.H., and J.K. selected and defined the study population; R.B. and S.R.-B. performed laboratory testing; M.H. and C.V.-G. prepared the dataset for analysis and performed univariate assessments; J.K. performed the multivariate analysis; and S.F., M.H., S.R.-B., C.V.-G., and J.K. reviewed the manuscript.

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

Correspondence: Stephen Spellman, Scientific Services, National Marrow Donor Program, 3001 Broadway St NE, Ste 100, Minneapolis, MN 55413; e-mail: sspellma@nmdp.org.

1
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
)
2
Flomenberg
 
N
Baxter-Lowe
 
LA
Confer
 
D
, et al. 
Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome.
Blood
2004
, vol. 
104
 
7
(pg. 
1923
-
1930
)
3
Karanes
 
C
Nelson
 
GO
Chitphakdithai
 
P
, et al. 
Twenty years of unrelated donor hematopoietic cell transplantation for adult recipients facilitated by the National Marrow Donor Program.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
9 suppl
(pg. 
8
-
15
)
4
Taylor
 
PA
Ehrhardt
 
MJ
Roforth
 
MM
, et al. 
Preformed antibody, not primed T cells, is the initial and major barrier to bone marrow engraftment in allosensitized recipients.
Blood
2007
, vol. 
109
 
3
(pg. 
1307
-
1315
)
5
Anasetti
 
C
Amos
 
D
Beatty
 
PG
, et al. 
Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma.
N Engl J Med
1989
, vol. 
320
 
4
(pg. 
197
-
204
)
6
Anasetti
 
C
Velardi
 
A
Blume
 
KG
Forman
 
SJ
Applebaum
 
JR
Hematopoietic cell transplantation from HLA partially matched related donors.
Thomas' Hematopoietic Cell Transplantation
2004
3rd edition
Boston, MA
Blackwell Science
(pg. 
1116
-
1131
)
7
Ottinger
 
HD
Rebmann
 
V
Pfeiffer
 
KA
, et al. 
Positive serum crossmatch as predictor for graft failure in HLA-mismatched allogeneic blood stem cell transplantation.
Transplantation
2002
, vol. 
73
 
8
(pg. 
1280
-
1285
)
8
Bray
 
RA
Nickerson
 
PW
Kerman
 
RH
Gebel
 
HM
Evolution of HLA antibody detection.
Immunol Res
2004
, vol. 
29
 
1–3
(pg. 
41
-
53
)
9
Bray
 
R
Nolan
 
JD
Larsen
 
C
, et al. 
Transplanting the sensitized patient: the Emory Wlgorithm.
Am J Transplant
2006
, vol. 
6
 
10
(pg. 
2307
-
2315
)
10
Zachary
 
AA
Leffell
 
MS
Detecting and monitoring human leukocyte antigen-specific antibodies.
Hum Immunol
2008
, vol. 
69
 
10
(pg. 
591
-
604
)
11
Patel
 
AM
Pancoska
 
C
Mulgaonkar
 
S
Weng
 
FL
Renal transplantation in patients with pretransplant donor-specific antibodies and negative flow cytometry crossmatches.
Am J Transplant
2007
, vol. 
7
 
10
(pg. 
2371
-
2377
)
12
Takanashi
 
M
Fujiwara
 
K
Tanaka
 
H
Satake
 
M
Nakajima
 
K
The impact of HLA antibodies on engraftment of unrelated cord blood transplants.
Transfusion
2008
, vol. 
48
 
4
(pg. 
791
-
793
)
13
Spellman
 
S
Setterholm
 
M
Maiers
 
M
, et al. 
Advances in the selection of HLA-compatible donors: refinements in HLA typing and matching over the first 20 years of the National Marrow Donor Program Registry.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
9 suppl
(pg. 
37
-
44
)
14
Petersdorf
 
EW
Longton
 
GM
Anasetti
 
C
, et al. 
Association of HLA-C disparity with graft failure after marrow transplantation from unrelated donors.
Blood
1997
, vol. 
89
 
5
(pg. 
1818
-
1823
)
15
Shaw
 
BE
Gooley
 
TA
Malkki
 
M
, et al. 
The importance of HLA-DPB1 in unrelated donor hematopoietic cell transplantation.
Blood
2007
, vol. 
110
 
13
(pg. 
4560
-
4566
)
Sign in via your Institution