Hematopoietic cell transplantation (HCT) can be curative for selected malignant and nonmalignant diseases.1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13  However, utilization and success of HCT are limited by several obstacles, primarily related to the importance of donor-recipient genetic match for favorable outcomes. While in most settings, best results are offered by human leukocyte antigen (HLA)14,15 –identical sibling transplantations, more than two thirds of patients awaiting HCT lack a suitable related donor. HCT with unrelated donor (URD) grafts are more frequently associated with severe graft-versus-host disease (GVHD16 ) or graft rejection.17, 18, 19  T-cell depletion of URD grafts has substantially reduced the risk of GVHD and early treatment-related mortality (TRM), but has not resulted in a concurrent survival benefit.20, 21, 22, 23, 24 

Despite the development of large international volunteer donor registries, still less than 50% of URD searches result in identification and availability of a suitably matched bone marrow (BM) graft.25  Donor searches for recipients not of Northern-European descent result in even lower success.25  These limitations have given impetus for the identification of alternative sources of hematopoietic stem cells (HSCs).

In the last decade, HCT using umbilical cord blood (UCB) grafts has increasingly been utilized, particularly for pediatric patients.26, 27, 28, 29, 30, 31, 32, 33, 34, 35  To date, it is estimated that more than 2000 unrelated umbilical cord blood transplantations (UCBTs) have been performed.26, 27, 28, 29, 30,33,35  The data indicate that UCB is a viable alternative source of HSCs and, in certain situations, may have advantages over URD marrow grafts. The aim of this review is to enumerate the advantages and disadvantages of each stem cell source and explore the issues involved in the selection process of marrow versus UCB grafts for specific patient populations.

As of 2002, more than 7 million registered volunteer marrow donors exist in more than 40 registries worldwide.36  The National Marrow Donor Program (NMDP37 ), the single largest registry, now has more than 4 million potential donors listed (R. King, NMDP, oral communication, May 2002). Still, URD marrow is not readily available for all patients.

NMDP data indicate that a median of 4 months is required to complete searches that result in a transplantation.36  Others have reported similar38,39  or considerably longer search times.40,41  An undefined number of patients succumb to their disease while awaiting identification of a suitable HLA-matched donor.42  In addition, about half of the initiated searches fail to provide a suitable donor marrow graft, even more so for ethnic/racial minority patients. Moreover, even if a donor is identified based on HLA-match criteria, not all donors are available when they are needed. Reasons for donor attrition include inability to contact the donor as a result of geographic movement or name change, loss of motivation over time, disqualification because of age or medical status, temporary unavailability, and death. The NMDP reports that about 30% of registered donors identified as potential matches are not available for further evaluation at the time of request.36 

These challenges in the donor search process are being addressed at multiple levels. Traditionally, potential donors have been serologically typed for HLA-A and HLA-B loci at recruitment. Registry studies indicate that HLA-A–, HLA-B–, and HLA-DR–typed donors are more likely to be available when needed and constitute the more often utilized pool.25,43  This has given impetus to more extensively type volunteers at the time of recruitment. As reported recently, between 16% to 52% of registered donors are HLA-A–, HLA-B–, and HLA-DR–typed in the various registries worldwide.36  Another step has been to target ethnic minorities in an effort to increase availability of rare haplotypes, although this has met with mixed success.36  The total number of donors recruited to registries has expanded rapidly in the last decade. However, the benefit of increasing registry size has perhaps reached its maximal impact. It is predicted that continuing focus on expanding the pool of registered donors, without attention to ethnicity, is likely to provide relatively little benefit.44, 45, 46 

With almost 35 years of human bone marrow transplantation (BMT) experience, several large studies have described outcomes with HLA-matched and -mismatched URD marrow transplantations. Table 1 summarizes results of selected large URD BMT series.

Table 1.

Myeloid engraftment, GVHD, and DFS with URD marrow transplantation



Reference no.
38 
51 
63 
41 
General characteristics   NMDP (1987-1990), 462 patients, malignant and nonmalignant diagnoses   IBMTR (1985-1991), leukemia only   HLA-6/6 sibling donor (n = 1224)   HLA-5/6 or 4/6, related donor (n = 340)   URD (n = 491)   Seattle (1985-1993), 88 pediatric patients, malignant and nonmalignant diagnoses   NMDP (1988-1996), 1423 patients, chronic myeloid leukemia only  
Age range, y (median)   0.3-54.5 (26.0)    1.0-57.0 (32)   1.0-53.0 (25)   1.0-56.0 (31)   0.5-17.8 (9.1)   0-20: 11%  
        21-40: 54%  
        > 40: 35%  
Myeloid engraftment,* % (median time, d)   94 (22)    99   HLA-5/6: 91   91   93 (21)   92 (20)  
     HLA-4/6: 84     
Acute GVHD II to IV, % (III-IV)   64 (47)    29 (13)   HLA-5/6: 44 (27)   HLA-6/6: 54 (35)   HLA-6/6: 83 (37)   43 (33)  
     HLA-4/6: 56 (36)   HLA-5/6: 63 (47)   HLA-5/6: 98 (62)   
Chronic GVHD, % (extensive)   55 (35)    42   HLA-5/6: 52   HLA-6/6: 62   HLA-6/6: 60   73 (60)  
     HLA-4/6: 60   HLA-5/6: 73   HLA-5/6: 69 (37)   
DFS, %   2-year DFS for leukemia, n = 352    3-year DFS   3-year DFS   3-year DFS   3-year DFS   3-year DFS:  
      Low risk = 40        Low risk: 66       HLA-5/6       HLA-6/6       CML: 75   All patients in CP: 43; < 35 years, CP, < 1 year after diagnosis: 63  
      High risk = 19        High risk: 12           Low risk: 33           Low risk: 41       Acute leukemia   
             High risk: 15           High risk: 11           Low risk ALL: 47   
         HLA-4/6       HLA-5/6           High risk ALL: 10   
             Low risk: 25           Low risk: 26           All AML: 46   

 

 

 

 
        High risk: 22
 
        High risk: 17
 
    Others: 29
 

 


Reference no.
38 
51 
63 
41 
General characteristics   NMDP (1987-1990), 462 patients, malignant and nonmalignant diagnoses   IBMTR (1985-1991), leukemia only   HLA-6/6 sibling donor (n = 1224)   HLA-5/6 or 4/6, related donor (n = 340)   URD (n = 491)   Seattle (1985-1993), 88 pediatric patients, malignant and nonmalignant diagnoses   NMDP (1988-1996), 1423 patients, chronic myeloid leukemia only  
Age range, y (median)   0.3-54.5 (26.0)    1.0-57.0 (32)   1.0-53.0 (25)   1.0-56.0 (31)   0.5-17.8 (9.1)   0-20: 11%  
        21-40: 54%  
        > 40: 35%  
Myeloid engraftment,* % (median time, d)   94 (22)    99   HLA-5/6: 91   91   93 (21)   92 (20)  
     HLA-4/6: 84     
Acute GVHD II to IV, % (III-IV)   64 (47)    29 (13)   HLA-5/6: 44 (27)   HLA-6/6: 54 (35)   HLA-6/6: 83 (37)   43 (33)  
     HLA-4/6: 56 (36)   HLA-5/6: 63 (47)   HLA-5/6: 98 (62)   
Chronic GVHD, % (extensive)   55 (35)    42   HLA-5/6: 52   HLA-6/6: 62   HLA-6/6: 60   73 (60)  
     HLA-4/6: 60   HLA-5/6: 73   HLA-5/6: 69 (37)   
DFS, %   2-year DFS for leukemia, n = 352    3-year DFS   3-year DFS   3-year DFS   3-year DFS   3-year DFS:  
      Low risk = 40        Low risk: 66       HLA-5/6       HLA-6/6       CML: 75   All patients in CP: 43; < 35 years, CP, < 1 year after diagnosis: 63  
      High risk = 19        High risk: 12           Low risk: 33           Low risk: 41       Acute leukemia   
             High risk: 15           High risk: 11           Low risk ALL: 47   
         HLA-4/6       HLA-5/6           High risk ALL: 10   
             Low risk: 25           Low risk: 26           All AML: 46   

 

 

 

 
        High risk: 22
 
        High risk: 17
 
    Others: 29
 

 

Results of selected large studies are shown.

DFS indicates disease-free survival; NMDP, National Marrow Donor Program; IBMTR, International Bone Marrow Transplant Registry; HLA-6/6, HLA matched donor; HLA-5/6 and HLA-4/6, donor-recipient mismatch for 1 or 2 HLA-A, HLA-B, or HLA-DRB1, respectively; and CP, chronic phase.

*

Myeloid engraftment was defined as absolute neutrophil count > 0.5 × 109/L, first of 3 consecutive days.

Engraftment

In a recent report of more than 5000 URD marrow transplantations facilitated by the NMDP, primary graft failure occurred in 4%.47  However, with mismatched donor grafts, this risk increases significantly.38,48, 49, 50, 51, 52  In particular, disparity for HLA-C, or more than one allele-level mismatch of class I determinants, has been associated with increased risk of graft failure.53,54  Additional factors negatively influencing probability of marrow engraftment have been the use of T-cell–depleted grafts38,50,55,56  and diseases such as chronic myeloid leukemia (CML),40,41,57  Fanconi anemia,9,58  aplastic anemia,8,59,60  and inherited metabolic storage disorders.61,62  Improved platelet engraftment has been associated with absence of severe acute GVHD and possibly higher marrow nucleated cell dose.47,63,64 

GVHD

Risk of GVHD is an important factor limiting HCT using an URD. With marrow transplantations, many studies have shown histoin-compatibility to be the greatest risk factor for the development of GVHD.52,54,63,65, 66, 67  Development of moderate to severe acute GVHD or extensive chronic GVHD is associated with diminished quality of life, as well as decreased overall survival, and is not regarded as desirable even in those with malignant disease where a graft-versus-leukemia (GVL68,69 ) effect is sought.68, 69, 70, 71  Grades II to IV GVHD is reported in 43% to 70% of phenotypically matched URD marrow transplantations, and in 63% to 95% of HLA one antigen-mismatched URD transplantations38,41,51,63,72, 73, 74, 75, 76  (Table 1). Chronic GVHD affects more than 55% of matched URD transplant recipients and as many as 80% of those receiving one antigen–mismatched URD HCTs.38,41,51,63,77  About 50% of patients with extensive chronic GVHD will die secondary to severe immune dysfunction.78 

Attempts to decrease GVHD and early TRM by the use of T-cell–depleted grafts have produced encouraging results, including in settings of mismatched unrelated and haploidentical related donors.20,79, 80, 81, 82, 83, 84, 85  However, significant concerns remain with graft failure and poor immune reconstitution resulting in opportunistic infections, posttransplantation lymphoproliferative disease, and relapse.21, 22, 23, 24  A large randomized, multicenter unrelated donor marrow transplantation trial in the United States, comparing T-cell depletion with pharmocologic GVHD prophylaxis, has recently concluded accrual.86  Analysis of this trial will include determination of GVHD prophylaxis on 3-year disease-free survival (DFS).

Immune reconstitution

Recovery of immune function after BMT has been characterized by several investigators, with unmanipulated as well as T-cell–depleted marrow HCTs.87, 88, 89, 90, 91  Reconstitution of T cells occurs in 2 phases: an initial expansion of mature, previously antigen-exposed T cells infused with the donor graft, and a later thymus-dependent proliferation of T cells, probably derived from HSCs. Immune recovery is impaired by the development of severe GVHD, the risk of which is particularly high after URD transplantations and in adult recipients.72, 73, 74, 75, 76  T-cell depletion can decrease the risk of GVHD; however, it is still associated with delayed restoration of T-cell subsets and increased risk of infectious complications.21, 22, 23, 24,88,89 

The immune response of the graft can also result in a GVL effect, potentially decreasing the risk of relapse in certain malignant disorders.68, 69, 70  With hematologic malignancies, both in pediatric and adult recipients, the biology of the disease and disease status at HCT have been critical factors determining risk of relapse after transplantation38, 39, 40, 41,51,57,63  (Table 1).

Survival

URD transplant recipients are at increased risk for GVHD, graft failure, infections, and decreased survival. BMT with HLA-mismatched grafts is associated with further increased morbidity and decreased survival, particularly in adult recipients48,49,63,65, 66, 67,92  (Figure 1). Moreover, as molecular techniques have developed, previously unrecognized mismatches at HLA-A, -B, and -DRB1 have been identified and shown to increase risk of graft rejection, GVHD, and mortality.53,54,67,93  Identification and study of additional transplantation antigens such as HLA-C53  and -DQB194  have suggested that outcome can be further optimized by higher levels of matching or matching across so-called ancestral haplotypes.95  However, with the extraordinary polymorphism of HLA alleles, such extensively matched URD transplantations will at best be limited to a minority of patients, particularly if allele-level matches at multiple loci are sought.

Figure 1.

Association of HLA mismatch with survival after unrelated bone marrow transplantation. Results of 211 consecutive unrelated transplantations at the University of Minnesota between 1985 and 1992. Reproduced from Davies et al92  with permission.

Figure 1.

Association of HLA mismatch with survival after unrelated bone marrow transplantation. Results of 211 consecutive unrelated transplantations at the University of Minnesota between 1985 and 1992. Reproduced from Davies et al92  with permission.

Close modal

Better DFS and overall survival with URD transplantations have been achieved when performed early in the course of the disease, both for malignant and nonmalignant diseases such as immunodeficiencies, Fanconi anemia, and inborn errors of metabolism. Notably promising results have been achieved after HLA-A–, HLA-B–, and HLA-DRB1–matched URD marrow transplantations for early CML in patients younger than 35 to 40 years40,41,57,96  (Table 1), and specific immunodeficiency states in young children.97, 98, 99  While URD transplantations are associated with inferior transplantation outcomes and survival compared with matched-related donor BMT for better prognosis leukemia, the difference in survival is less evident with poor prognosis disease.51 

The first HLA-matched sibling UCBT was performed by Gluckman et al100  in 1988 in a child with Fanconi anemia. Subsequently, reports documented the feasibility and efficacy of mismatched related and unrelated UCBT.28,101, 102, 103  By 1993, repositories of unrelated donor UCB were established in New York, Dusseldorf, and Milan.104,105  Currently, private and publicly funded cord blood banks worldwide store an estimated 70 000 cryopreserved HLA-A–, HLA-B–, and HLA-DRB1–typed units, mostly for the purpose of URD transplantations.102,104, 105, 106, 107 

As limited HLA mismatch appears to be better tolerated with UCB grafts, units are identified for most patients even with the size of current repositories. A significant advantage of UCB is the rapidity with which an acceptable HLA-matched unit, once identified, can be acquired.26,107, 108, 109  As UCB is cryopreserved, acquisition of an HLA-matched unit is quick. In addition, UCB units are typically intermediate, or high resolution typed at HLA-A, -B, and -DRB1 loci at collection, further shortening search time. In one study, the median time for a UCB unit to be identified and available was 13.5 days.108  Therefore this HSC source is particularly appealing for patients who need to proceed urgently to HCT. In addition, rescheduling the date of infusion of a cryopreserved UCB unit to fit the recipient's needs is simple.

Total nucleated cell (TNC) dose of a UCB graft has proved to be a critical determinant of engraftment and survival with UCBT. Therefore, the fixed cell content of a UCB unit represents the major limiting factor, particularly for adult recipients. However, UCB has a higher frequency of progenitor cells compared with adult peripheral blood or bone marrow.110,111  Laboratory data also suggest that UCB-derived stem cells demonstrate an increased growth/engraftment potential as evidenced by larger size of colonies formed in cultures and efficient long-term, multilineage repopulation of immunodeficient mice by human UCB cells even in the absence of exogenous human cytokine support (in contrast to results with adult human BM grafts in the same mouse model).112, 113, 114  In human HCT, while the number of nucleated cells infused with an UCB graft can frequently be one-log less than that infused with a BM graft, these features perhaps explain its comparable engraftment potential. Outcomes of human UCBT from larger series are described in the remainder of this section and in Table 2.

Table 2.

Engraftment and acute GVHD after unrelated umbilical cord blood transplantation (UCBT)



Reference no.

29 
28 
35 
General characteristics   Analysis of 562 patients receiving UCB grafts from New York Blood Center between 1992 and 1998   Multicenter (Eurocord and others) between 1988 and 1996. Results of unrelated UCBT group (n = 65) are shown   102 UCBTs at the University of Minnesota between 1994 and 2001  
Age, y (median)   82% ≤ 17   0.3-45.0 (9)   0.2-56.9 (7.4)  
  18% ≥ 18    
Donor-recipient HLA-mismatch   7% of grafts: 6/6 HLA-match   14% of grafts: 6/6 HLA-match   14% of grafts: no HLA-mm  
  39% of grafts: 1 HLA-mm   83% of grafts: 1-2 HLA-mm   84% of grafts: 1-2 HLA-mm  
  54% of grafts: 2-3 HLA-mm    
Myeloid engraftment, % (median time, d)   At day 60: 91 (28)   At day 60: 87   At day 42: 88 (23)  
Platelet engraftment, % (median time, d)   At day 180*: 85 (90)   At day 60†: 39   At day 180*: 65 (86)  
Favorable factors associated with engraftment, multivariate analysis  
  1. Higher nucleated cell dose, HLA-match, US center, and diagnosis other than chronic myeloid leukemia and aplastic anemia were associated with successful myeloid engraftment.

  2. Younger age, absence of infection after HCT, and absence of acute GVHD were associated with platelet engraftment.

 
Recovery of neutrophil count and platelet engraftment associated with the following:  
  1. Neutrophil recovery: higher CD34+ cell dose infused (> 1.7 × 105/kg).

  2. Platelet recovery: higher CD34+ cell dose infused and absence of severe acute GVHD.

  3. No association of either with HLA match was observed.

 
  
  1. Higher number of nucleated cells infused and

  2. HLA-identity.

 
 
Grade II-IV (III-IV) acute GVHD, %   6/6 HLA-match: 27 (9)   32 (20)   39 (11)  
  1 HLA-mm: 48 (22)    
  2-3 HLA-mm: 49 (25)    
Factors associated with acute GVHD
 
  1. Older age (≤ 12 vs ≥ 12).

  2. Non-US center location.

  3. HLA mismatch (0 vs ≥ 1) approached significance (P = .06), but no correlation with number of mismatches was seen.


 
  1. No association of acute GVHD with number of HLA mismatches.

  2. CMV-seronegative status was associated with a lower risk of acute GVHD.


 
No association with CD3 cell dose, HLA disparity or class of HLA-mismatch.
 


Reference no.

29 
28 
35 
General characteristics   Analysis of 562 patients receiving UCB grafts from New York Blood Center between 1992 and 1998   Multicenter (Eurocord and others) between 1988 and 1996. Results of unrelated UCBT group (n = 65) are shown   102 UCBTs at the University of Minnesota between 1994 and 2001  
Age, y (median)   82% ≤ 17   0.3-45.0 (9)   0.2-56.9 (7.4)  
  18% ≥ 18    
Donor-recipient HLA-mismatch   7% of grafts: 6/6 HLA-match   14% of grafts: 6/6 HLA-match   14% of grafts: no HLA-mm  
  39% of grafts: 1 HLA-mm   83% of grafts: 1-2 HLA-mm   84% of grafts: 1-2 HLA-mm  
  54% of grafts: 2-3 HLA-mm    
Myeloid engraftment, % (median time, d)   At day 60: 91 (28)   At day 60: 87   At day 42: 88 (23)  
Platelet engraftment, % (median time, d)   At day 180*: 85 (90)   At day 60†: 39   At day 180*: 65 (86)  
Favorable factors associated with engraftment, multivariate analysis  
  1. Higher nucleated cell dose, HLA-match, US center, and diagnosis other than chronic myeloid leukemia and aplastic anemia were associated with successful myeloid engraftment.

  2. Younger age, absence of infection after HCT, and absence of acute GVHD were associated with platelet engraftment.

 
Recovery of neutrophil count and platelet engraftment associated with the following:  
  1. Neutrophil recovery: higher CD34+ cell dose infused (> 1.7 × 105/kg).

  2. Platelet recovery: higher CD34+ cell dose infused and absence of severe acute GVHD.

  3. No association of either with HLA match was observed.

 
  
  1. Higher number of nucleated cells infused and

  2. HLA-identity.

 
 
Grade II-IV (III-IV) acute GVHD, %   6/6 HLA-match: 27 (9)   32 (20)   39 (11)  
  1 HLA-mm: 48 (22)    
  2-3 HLA-mm: 49 (25)    
Factors associated with acute GVHD
 
  1. Older age (≤ 12 vs ≥ 12).

  2. Non-US center location.

  3. HLA mismatch (0 vs ≥ 1) approached significance (P = .06), but no correlation with number of mismatches was seen.


 
  1. No association of acute GVHD with number of HLA mismatches.

  2. CMV-seronegative status was associated with a lower risk of acute GVHD.


 
No association with CD3 cell dose, HLA disparity or class of HLA-mismatch.
 

Results of 3 large peer-reviewed studies of unrelated UCBT are shown.

1, 2, or 3 HLA-mm, HLA-mismatch at 1, 2, or 3 HLA-A, -B, or -DRB1; myeloid engraftment, neutrophil count ≤ 0.5 × 109/L, first of 3 consecutive days; and platelet engraftment, ≥ 50 × 109* or ≥ 20 × 109† (untransfused) platelets/L, first of 7 days.

Engraftment

As with marrow transplantations, several factors have been identified that predict successful engraftment with UCB grafts. The single most important factor influencing time to hematopoietic recovery appears to be the nucleated cell content of the graft relative to recipient size.28,29,31,32,35  The effect of HLA mismatch on engraftment is less clear (Table 2).

In a report by Gluckman et al28  (Table 2), a graft nucleated cell dose higher than 3.7 × 107/kg was associated with shorter time to neutrophil recovery (25 vs 35 days). Wagner et al35  (Table 2), in a series of 102 unrelated UCBTs, analyzed the influence of graft CD34+ cell dose and observed significantly inferior speed and probability of engraftment with CD34+ cell dose lower than 1.7 × 105/kg. Rubinstein et al29  showed that while a step-wise increasing graft nucleated cell dose progressively shortened the time to myeloid recovery, the final cumulative incidence of myeloid recovery was similar once the nucleated cell dose exceeded 2.5 × 107/kg, suggesting that a threshold number of nucleated cells are needed for engraftment (Figure 2). Analysis of HLA mismatch in this study29  (Table 2), and a more recent review update with 861 unrelated UCBTs,115  revealed a relationship between HLA match and engraftment. The median time to neutrophil recovery with 6 antigen–matched grafts was 23 days compared with 28 days with mismatched grafts (P = .0027).115  However, no association between engraftment characteristics and number of HLA mismatches (one vs more than one HLA mismatch) was observed.115 

Figure 2.

Association of umbilical cord blood nucleated cell dose with speed of myeloid engraftment. Higher graft nucleated cell doses result in shorter time to neutrophil recovery. However, the 3 highest cell doses have similar Kaplan-Meier probability of achieving engraftment. Reproduced from Rubinstein et al29  with permission, for English-language use only. Copyright 1998 Massachusetts Medical Society. All rights reserved.

Figure 2.

Association of umbilical cord blood nucleated cell dose with speed of myeloid engraftment. Higher graft nucleated cell doses result in shorter time to neutrophil recovery. However, the 3 highest cell doses have similar Kaplan-Meier probability of achieving engraftment. Reproduced from Rubinstein et al29  with permission, for English-language use only. Copyright 1998 Massachusetts Medical Society. All rights reserved.

Close modal

Consequent to these studies, a consensus is emerging that UCB grafts with higher cell doses should be selected wherever possible to optimize engraftment. Particularly poor results are seen after UCBT when the nucleated cell dose infused is lower than 1.5 × 107/kg.105,116,117  These data suggest that a minimum acceptable nucleated cell dose should be 1.5 × 107 nucleated cells/kg117  to reduce time to myeloid recovery and increase probability of engraftment.

GVHD

UCBT series in the URD setting has reported 33% to 44% and 11% to 22% incidences of grades II to IV and grades III to IV acute GVHD, respectively, and 0% to 25% incidence of chronic GVHD.28,29,31,35  These results are particularly notable since the majority of unrelated UCBTs were performed using 1 to 2 HLA–mismatched grafts. It could be argued that a majority of the recipient population was young in these reports; however, the incidence of severe GVHD in adults receiving mismatched UCB grafts has also been low.33 

HLA mismatch has been the strongest risk factor for GVHD in recipients of marrow transplants. The association of histocompatibility with GVHD in the UCBT setting has only recently become apparent and remains less clear. Many UCBT series have failed to observe an influence of 0- to 3-antigen–mismatch on the risk of acute or chronic GVHD28,32,33,35  (Table 2). Eurocord studies, while observing association of HLA mismatch with risk of acute GVHD in the related UCBT setting, did not show an effect of HLA mismatch on risk of acute GVHD after unrelated UCBT.28,32  However, in a large series of unrelated UCBTs, subsequently updated to 861 patients, Rubinstein et al29  and Rubenstein and Stevens115  reported significantly higher rates of acute GVHD with mismatched grafts, although no direct association of HLA mismatch with chronic GVHD was observed.

Immune reconstitution

The goal of HCT is the establishment of donor cell immune tolerance to recipient self major histocompatibility complex (MHC) antigens, with reconstitution of effective immune surveillance as well as response to foreign antigens including tumors and pathogens. With UCB grafts, it has been hypothesized that with its lower and previously unstimulated T-cell content, the early, thymus-independent expansion of T lymphocytes may be impaired. However, preliminary analyses show that the recovery of immune-cell repertoire after UCBT is comparable with that after unmanipulated marrow HCT.30,118, 119, 120, 121, 122, 123  Natural killer (NK) cell numbers are reconstituted promptly, and the recovery of B lymphocytes and CD4+ T cells may actually be faster after UCBT.

The functional naiveté of neonatal lymphocytes and the lower risk of GVHD with UCB grafts have raised concern for a reduced GVL effect after UCBT.124  The exact relationship between GVHD and GVL is unclear,70,125,126  even though the data suggest that development of GVHD is associated with decreased risk of relapse.69,70,127,128 

Initial experience with UCBT for leukemia suggests that similar to BMT, the biology of the disease and leukemia status at transplantation are the major determinants of outcome. Wagner et al,35  in a study of 102 UCBTs (median age, 7.4 years) from a single institution, reported a 37% cumulative incidence of relapse at 2 years, with age and malignancy risk group being the 2 factors associated with risk of relapse. Also, in a Eurocord report including 60 unrelated UCBTs in children (< 15 years) with acute leukemia, disease status at transplantation was the most important factor predicting outcome.32  The 2-year incidence of relapse and 2-year probability of DFS in the poor-risk and good-risk groups were 31% versus 75% and 7% versus 40%, respectively. Notably, cell dose and HLA mismatch of the graft did not influence DFS in the latter series.

Survival

Several studies have analyzed factors influencing survival after UCBT.27, 28, 29,31, 32, 33,35  Locatelli et al32  reported outcomes in pediatric acute leukemia from the Eurocord registry. Of 60 unrelated UCB grafts, 54 were 1 to 4 loci HLA disparate, with the majority being 1 to 2 loci HLA mismatched. In univariate or multivariate analyses, number of HLA mismatches did not influence survival. Similarly Gluckman et al,28  in a report of 65 unrelated UCBTs (0-3 antigen HLA mismatched) in pediatric recipients, and Laughlin et al,33  in a study of 68 adult recipients of 0 to 3 antigen HLA-mismatched UCBTs, showed no association of degree of HLA mismatch with overall survival in the unrelated setting. Of note, however, 2 larger series, by Wagner et al35  (102 UCBTs, single institution) and Rubinstein et al29  (562 UCBTs using grafts supplied by the New York Blood Center), have observed a significant association of HLA mismatch with survival after unrelated UCBT.

Based on the UCBT experience reported thus far, UCB cell dose has been the most critical factor in determining speed of engraftment and survival after UCBT. HLA disparity is also likely a major factor, although reports are contradictory.28,29,31,32,35,115,129  The challenge for transplantation centers in selecting the optimal stem cell source for a recipient is defining the cell dose below which clinical outcomes become significantly inferior. At the University of Minnesota, local experience suggests that 1.5 × 107 nucleated cells/kg or 1.7 × 105 CD34+ cells/kg define that threshold below which outcomes are significantly poor,35  and routine use of units below this threshold is now not permitted. While patients receiving lower cell doses can engraft, risk of graft failure is exceedingly high and time to neutrophil recovery is prolonged.28,33,35,116 

Figure 3A-B shows data from the University of Minnesota, illustrating the impact of CD34+ cell dose on survival in recipients of 1 or 2 HLA–mismatched UCBTs. Survival is improved with the higher cell doses, suggesting that a higher cell dose can partially compensate for higher HLA disparity. Notably, a study from the New York Blood Center concluded that raising the nucleated cell dose by approximately 3 × 107 cells/kg may offset the negative effect of one HLA mismatch.130  As more data are collected, it will be possible to define with greater confidence an acceptable lower limit of cell dose for a given degree of HLA disparity.

Figure 3.

Association of umbilical cord blood graft CD34+ cell dose with overall survival. University of Minnesota data: 102 consecutive unrelated UCBTs between 1994 and 2001, unpublished data. Survival by CD34+ cell dose (× 105/kg) for (A) 1-antigen mismatched UCBTs, and (B) 2-antigen mismatched UCBTs.

Figure 3.

Association of umbilical cord blood graft CD34+ cell dose with overall survival. University of Minnesota data: 102 consecutive unrelated UCBTs between 1994 and 2001, unpublished data. Survival by CD34+ cell dose (× 105/kg) for (A) 1-antigen mismatched UCBTs, and (B) 2-antigen mismatched UCBTs.

Close modal

Graft TNC dose has been used as a marker of graft “potency” in the majority of UCBT studies reported thus far; although, a few studies have also defined graft CD34+ cell dose as a predictor of speed of engraftment and survival with UCBT.33,35  The TNC count of any UCB unit includes a variable number of nucleated red cells. While TNC dose has consistently correlated with transplantation outcomes, and the nucleated red cell count has been shown to correlate with progenitor cell numbers,131  a progenitor assay such as colony-forming unit (CFU) or CD34+ content of a UCB graft may be a more precise predictor of engraftment and survival with any given UCB unit.132  In addition, CFU content (after thawing) is a measure of progenitor cell viability.

The contiguous integral segment of a cryopreserved UCB unit is representative of the whole product and can be utilized to accurately estimate CFU content and viability after thawing (D. A. Wall, oral communication, October 2002). Unfortunately, results of CFU assays are susceptible to minor variations in techniques and culture conditions used, and may not be accurately reproducible between different laboratories. While, CD34+ cell content of a UCB unit at harvest is not universally available at present, the authors' bias is toward CD34+ cell content to quantify graft potency. Studies directly comparing the relative precision of TNC versus CD34+ content to predict the various outcomes after UCBT are awaited.

There are no prospective studies comparing outcomes in similar patient populations randomized to receive UCB or BM grafts. There are 3 reports31,34,133  (and 1 study in abstract form129 ) that have retrospectively compared HCT outcomes with the 2 stem cell sources (Table 3).

Table 3.

Engraftment, GVHD, and survival in series comparing UCBT with BMT


Reference no.

Age, y (median)

Donor-recipient HLA mismatch

Myeloid engraftment, % (median, d)

Platelet engraftment at day 180, % (median, d)

Acute GVHD grade II-IV, % (III-IV) [chronic GVHD]

Survival, %

Comments
Ref no. 133         
    Type of HCT and patient nos.     At day 60     3-year overall survival   
        UCBT: 113   < 1-15 (5)   HLA-identical sibling   89 (26)   86 (44)*   14 (2) [5]   64   Comparison of UCBT and BMT from HLA-identical sibling donors.  
        BMT: 2052   < 1-15 (8)   HLA-identical sibling   98 (18)   96 (24)*   24 (10) [14]   66  
  1. There was a significantly longer time to platelet recovery in the early transplantation period (< 35 days) with UCBT

  2. Risk of acute and chronic GVHD was significantly lower with UCBT

  3. No significant difference in 100-day or 3-year survival observed between UCBT or BMT groups for either malignant or nonmalignant diseases

 
Ref no. 31         
    Type of HCT and patient nos.    % with mismatched grafts   At day 60     2-year DFS (relapse rate)   
        UBMT: 262   5-12 (8)   18%: 1 HLA-mm   96 (18)   85 (29)*   56 (29) [46]   43 (39)  
  1. Significant delay of neutrophil and platelet engraftment in UCBT group

  2. Neutrophil and platelet recovery correlated with nucleated cell dose (< 3.7 vs > 3.7 × 107 cells/kg) but not with HLA-disparity for UCBT

  3. Decreased acute GVHD in the UCBT and T-UBMT groups compared with UBMT group

 
        T-UBMT: 180   6-12 (8)   40%: 1-2 HLA-mm   90 (16)   85 (29)*   19 (8) [12]   37 (47)   
        UCBT: 99   2.5-10 (6)   89%: 1-3 HLA-mm   80 (32)   90 (81)*   33 (21) [25]   31 (38)   
Ref no. 34         
    Comparison groups (patient numbers)    HLA-6/6 match   At day 45     2-year overall survival   
        UCB vs BM-MTX (n = 26 each)   (4.5) vs (4.7)   19% vs 100%   88 (29) vs 96 (22)   72 (66) vs 76 (30)†   42 (19) vs 35 (8)   53 vs 41   Matched-pair analysis.  
        UCB vs BM-TCD (n = 31 each)
 
(5.8) vs (6.8)
 
13% vs 100%
 
85 (27) vs 90 (14)
 
84 (61) vs 84 (59)†
 
36 (10) vs 35 (13)
 
52 vs 56
 
  1. No difference in graft failure among comparison groups.

  2. Neutrophil recovery was significantly delayed in UCBT group vs BM-MTX or BM-TCD.

  3. No difference in acute/chronic GVHD in comparison groups

  4. Survival at 100 days/2 years was comparable between BM and UCB pairs


 

Reference no.

Age, y (median)

Donor-recipient HLA mismatch

Myeloid engraftment, % (median, d)

Platelet engraftment at day 180, % (median, d)

Acute GVHD grade II-IV, % (III-IV) [chronic GVHD]

Survival, %

Comments
Ref no. 133         
    Type of HCT and patient nos.     At day 60     3-year overall survival   
        UCBT: 113   < 1-15 (5)   HLA-identical sibling   89 (26)   86 (44)*   14 (2) [5]   64   Comparison of UCBT and BMT from HLA-identical sibling donors.  
        BMT: 2052   < 1-15 (8)   HLA-identical sibling   98 (18)   96 (24)*   24 (10) [14]   66  
  1. There was a significantly longer time to platelet recovery in the early transplantation period (< 35 days) with UCBT

  2. Risk of acute and chronic GVHD was significantly lower with UCBT

  3. No significant difference in 100-day or 3-year survival observed between UCBT or BMT groups for either malignant or nonmalignant diseases

 
Ref no. 31         
    Type of HCT and patient nos.    % with mismatched grafts   At day 60     2-year DFS (relapse rate)   
        UBMT: 262   5-12 (8)   18%: 1 HLA-mm   96 (18)   85 (29)*   56 (29) [46]   43 (39)  
  1. Significant delay of neutrophil and platelet engraftment in UCBT group

  2. Neutrophil and platelet recovery correlated with nucleated cell dose (< 3.7 vs > 3.7 × 107 cells/kg) but not with HLA-disparity for UCBT

  3. Decreased acute GVHD in the UCBT and T-UBMT groups compared with UBMT group

 
        T-UBMT: 180   6-12 (8)   40%: 1-2 HLA-mm   90 (16)   85 (29)*   19 (8) [12]   37 (47)   
        UCBT: 99   2.5-10 (6)   89%: 1-3 HLA-mm   80 (32)   90 (81)*   33 (21) [25]   31 (38)   
Ref no. 34         
    Comparison groups (patient numbers)    HLA-6/6 match   At day 45     2-year overall survival   
        UCB vs BM-MTX (n = 26 each)   (4.5) vs (4.7)   19% vs 100%   88 (29) vs 96 (22)   72 (66) vs 76 (30)†   42 (19) vs 35 (8)   53 vs 41   Matched-pair analysis.  
        UCB vs BM-TCD (n = 31 each)
 
(5.8) vs (6.8)
 
13% vs 100%
 
85 (27) vs 90 (14)
 
84 (61) vs 84 (59)†
 
36 (10) vs 35 (13)
 
52 vs 56
 
  1. No difference in graft failure among comparison groups.

  2. Neutrophil recovery was significantly delayed in UCBT group vs BM-MTX or BM-TCD.

  3. No difference in acute/chronic GVHD in comparison groups

  4. Survival at 100 days/2 years was comparable between BM and UCB pairs


 

All series shown are in pediatric recipients only.

HLA-mm indicates HLAmismatch; UBMT, unmanipulated (unrelated) bone marrow transplantation; BM-MTX, T-replete bone marrow transplantation with methotrexate GVHD prophylaxis; T-UBMT or BM-TCD, T-cell—depleted unrelated bone marrow transplantation; myeloid engraftment, neutrophil count ≥ 0.5 × 109/L, first of 3 consecutive days; and platelet engraftment, ≥ 20 × 109 (untransfused) platelets/L, first of 7 days* or 14 days† free of platelet transfusions.

Engraftment

In a matched pair analysis34  comparing HLA-A–, -B–, and -DRB1–matched URD marrow transplantations using either methotrexate (BM-MTX) or T-cell depletion (BM-TCD) for GVHD prophylaxis, with 0 to 3 loci HLA–mismatched UCBT (Table 3), the speed of neutrophil recovery was significantly slower after UCBT when compared with the BM-MTX group (median, 29 vs 22 days; P = .03) or the BM-TCD group (median, 27 vs 14 days; P < .01). However by day 45, the overall myeloid engraftment rate was comparable in all groups. Time to platelet engraftment, while slower with UCBT, was not statistically different from the BM-MTX group, perhaps because of small patient numbers (median, 66 days vs 30 days: UCBT vs BM-MTX [P = .12]). However, at 6 months, there was no difference in platelet engraftment. In another registry report with larger numbers (Table 3), Rocha et al31  also observed significant delay in speed of both neutrophil and platelet engraftment after UCBT when compared with URD marrow transplantation in children with acute leukemia.

GVHD

Current data indicate that HLA mismatch may be better tolerated in the UCBT setting. A registry study (Table 3) comparing outcomes of HLA-identical sibling UCBT versus HLA-identical sibling BMT in pediatric recipients observed significantly lower incidence of acute and chronic GVHD in the UCBT group.133  This study is perhaps the clearest demonstration of a difference in biologic properties between the 2 stem cell sources, as interpretation of GVHD between unrelated UCBT and BMT is often complicated by different levels of HLA histocompatibility and other recipient heterogeneity.

There are 2 other reports that have compared GVHD frequencies in unrelated donor BM and UCB recipients. In a matched-pair analysis from a single institution (Table 3), risk of acute and chronic GVHD was similar when comparing outcomes in recipients of HLA-A–, HLA-B–, and HLA-DRB1–matched unmanipulated BM and mostly 1 to 2 antigen mismatched unrelated UCB transplants.34  In another registry study (Table 3), 0 to 3 antigen mismatched UCBT transplantations were associated with a significantly lower risk of acute and chronic GVHD compared with unmanipulated, mostly HLA-matched marrow transplantations.31  Taken together these data indicate that despite the greater degrees of HLA disparity accepted in UCBT, the risk of developing acute and chronic GVHD after 1 to 2 antigen HLA-mismatched unrelated UCBT27, 28, 29, 30, 31,34,35  is similar, or even lower, than that reported with HLA-matched bone marrow transplantation.38,54,63,65,134 

The reason(s) for this lower risk of GVHD after UCBT is not clear. Functional and phenotypic immaturity of UCB lymphocytes135, 136, 137, 138, 139  and/or a reduced T-cell dose infused with UCB grafts35,105,140  may contribute to its reduced alloreactivity. Mature T cells present in the donor graft are the chief contributor to repopulating T cells in the first year after HCT.141  Large numbers of mature, antigen-specific T cells are infused in a BM graft and can initiate GVHD due to recognition of cross-reactive alloantigens.136  In contrast, characterization of the UCB αβ T-cell repertoire reveals a naive T-cell population unexposed to prior antigenic stimulation.136  It has been suggested that absence of clonal expansion in response to alloantigens by UCB T cells contributes to its enhanced tolerance.136 

Relapse and GVL effect

Current experience comparing risk of relapse after UCBT versus BMT is limited. In an analysis of HLA-matched sibling transplantations (Table 3), the 3-year survival in patients with malignant diagnosis was comparable after UCBT or BMT (P = .69). Notably, in another study of HCT for acute leukemia in children by Rocha et al31  (Table 3), a larger proportion of recipients (18%-20%) in the unmanipulated BMT (UBMT) and UCBT groups had advanced-stage leukemia compared with the T-cell–depleted BMT (T-UBMT) group (9%). Interestingly, while in both T-UBMT and UCBT groups a lower incidence of acute and chronic GVHD was noted relative to the unmanipulated BM transplant recipients, the T-UBMT group, but not the UCBT group, had an increased risk of early relapse when compared with the UBMT group (P = .02).

Overall, there is no evidence thus far to suggest a higher risk of leukemia relapse after UCBT. The observations of (1) better donor cell immune tolerance of recipient MHC antigens, evidenced by acceptable risk of GVHD despite up to 2 HLA-mismatched grafts, and (2) preserved GVL effect after UCBT might be explained by intact UCB NK cell function combined with immaturity of umbilical B and T cells. In laboratory studies, NK-like cytotoxicity has not been linked with pathogenesis of GVHD, but has been shown to mediate a GVL effect.142, 143, 144 

A recent study of HCT in mice and humans showed that infusion of donor-derived alloreactive NK cells not only provides a GVL effect, but may also protect against GVHD by targeting recipient antigen presenting cells.145  UCB contains levels of NK cells and inducible NK-like cytotoxic activity similar to those in adult peripheral blood,135,146,147  which might explain a preserved GVL effect. Prospective studies comparing similar patient populations receiving UCB or marrow graft are needed for reliable conclusions; although, it remains to be seen if such studies will be feasible or ever performed. Randomization will be difficult as UCB grafts are typically available much faster than URD marrow. Hence high-risk patients (eg, leukemia in tenuous remission) may be more likely to receive UCB transplants. Strict control of patient risk factors will be critical for any reliable comparison between UCB and marrow transplantations.

Survival

There are 3 retrospective reports that have compared survival between URD UCB and marrow HCT. A registry report comparing HLA-mismatched UCBT with unmanipulated marrow in children with leukemia (Table 3) noted a significantly higher early (< 100 days) TRM in the UCBT group; although, overall survival between 0 to 2 HLA-mismatched UCBT and mostly HLA-matched URD marrow transplantations was comparable.31  UCB recipients were more likely to have adverse prognostic factors in this study. Other groups have not reported increased early TRM with UCBT.

A second study from the International Bone Marrow Transplant registry (IBMTR), reported in abstract form, compared 296 UCBTs (94% with at least one mismatch at HLA-A, HLA-B, or HLA-DRB1) with 210 URD BMTs (62% matched at HLA-A, HLA-B, and HLA-DRB1) in children with hematologic malignancy.129  This study reported similar survival rates in recipients of BMT and 0 or 1 HLA-mismatched UCBT, with reduced GVHD in the UCB transplant recipients. In this study survival was reduced in recipients of 2 HLA-mismatched UCB transplants compared with recipients of better-matched UCB and BM transplant recipients. Also, a matched-pair case-control study (Table 3) showed comparable outcomes in recipients of HLA-A–, HLA-B–, and HLA-DRB1–matched marrow and mismatched UCB.34  Taken together, these results suggest that UCB with limited HLA mismatch is an acceptable alternative to marrow, at least in children.

BMT for acute leukemia in adults, from donors other than HLA-identical siblings, is associated with a high risk of GVHD and treatment failure.38,51,148,149  UCBT may have an advantage in adult recipients because of potentially decreased risk of GVHD. However, as cell dose is associated with survival, this considerably limits the pool of eligible UCB grafts for adult recipients.

Laughlin et al33  reported the first major adult UCBT series from 5 US centers. The recipients (N = 68) weighed a median of 69.2 kg (range, 40.9-115.5 kg), and 54 had a hematologic cancer, of which 50 were considered intermediate/high risk. Of the grafts, 97% were 1 to 3 antigen HLA–mismatched with their recipient. The median nucleated cell dose infused was 2.1 × 107 cells/kg of body weight (range, 1-6.3 × 107 cells/kg). Engraftment frequency was similar to pediatric series with an estimated probability of myeloid recovery by day 42 of 90% (median time to engraftment, 28 days). Time to neutrophil count higher than 0.5 × 109/L was associated with nucleated cell dose (< 1.87 × 107 vs > 1.87 × 107; P = .003). Despite a high frequency of HLA-mismatched grafts, the probabilities of grades II to IV and grades III to IV GVHD were 60% and 20%, respectively. Of 68 recipients, 19 (18 disease-free [26%]) were alive at 22 months of follow-up. Speed of myeloid recovery and DFS were linked with higher nucleated cell and CD34+ cell dose. There was no significant association between the kinetics of myeloid recovery, graft failure, or acute GVHD with extent of HLA mismatch. Of note, the risk of severe acute and chronic GVHD was lower than typically reported after URD marrow transplantations.38,40,57,150 

In another report151  of 22 adult patients weighing 41 to 85 kg, who received unrelated UCBT with TNC dose ranging from 1.01 to 4.96 × 107/kg, all 20 patients who survived longer than 30 days showed myeloid engraftment at a median of 22 days. One patient who received the lowest cell dose experienced secondary graft failure. Also, in a separate Eurocord report of 42 adult recipients of UCBT,116  median time to neutrophil recovery (> 0.5 × 109/L) was 35 days, and no patient who received less than 1.0 × 107 nucleated cells/kg survived. These results combined once again underscore the importance of graft cell dose relative to recipient size for optimal results after UCBT and support a minimum nucleated cell dose limit of 1.5 × 107 nucleated cells/kg.

Several strategies to increase the nucleated/CD34+ cell dose of the graft are being investigated, including ex vivo expansion of UCB hematopoietic cells.152,153  The hematopoietic cells in UCB have an immense capacity to expand ex vivo in the presence of an appropriate cytokine cocktail.111,154,155  However, while the yield of more mature hematopoietic progenitor cells is extensive, it is not yet clear if actual expansion of the pluripotent, long-term repopulating HSCs is possible or required.111,154,155  It will be important however, to demonstrate that HSCs are at least maintained, and safety studies are currently under way.

Alternative approaches to optimize outcomes are being explored, such as multiunit UCB transplantation,156  UCBT combined with infusion of mesenchymal stem cells, or UCBT after a nonmyeloablative preparative regimen.157  Barker et al158  reported short-term outcomes in 8 adults (median weight, 81 kg) with advanced hematologic malignancy using 2 unrelated 1 to 2 HLA-mismatched UCB grafts. Initials results are encouraging; however, further study is required to show if double UCBT is associated with improved engraftment and survival compared with single UCBT in larger-sized recipients.

As noted earlier, there are a number of logistic and biologic differences between the 2 stem cell sources, which may make one more advantageous over the other. A comparison of issues involved in the selection of BM versus UCB grafts is outlined in Table 4.

Table 4.

Comparison of issues involved in URD marrow and UCB graft selection




Marrow graft

UCB graft
Available pool   Living, volunteer donors, currently ≈ 7 million   Preharvested, cryopreserved units, currently ≈ 70 000  
Typical donor HLA-type information in database   HLA-A, -B ± -DRB1   HLA-A, -B, and -DRB1  
Acceptable donor-recipient HLA match   HLA-A, -B (serologic), and -DRB1 match, with 1 mismatch permitted for recipients < 36 y   4 of 6 loci match at HLA-A, -B, and -DRB1  
Median search time   Longer, ≈ 4 mo   Shorter, < 1 mo  
Major limiting factors to graft acquisition   HLA-match; donor attrition/availability   Fixed UCB cell content (especially for larger sized recipients)  
Ease of rearranging date of stem cell infusion   Can be difficult   Easy  
Potential for second HSC graft or DLI from same donor   Yes   No  
Potential for disease transmission to recipient    
    CMV/EBV   Yes   No  
    Congenital disease*  No   Yes  
Risk to donor
 
Uncommon (anesthesia related, surgical complication, emergent blood transfusion)
 
None
 



Marrow graft

UCB graft
Available pool   Living, volunteer donors, currently ≈ 7 million   Preharvested, cryopreserved units, currently ≈ 70 000  
Typical donor HLA-type information in database   HLA-A, -B ± -DRB1   HLA-A, -B, and -DRB1  
Acceptable donor-recipient HLA match   HLA-A, -B (serologic), and -DRB1 match, with 1 mismatch permitted for recipients < 36 y   4 of 6 loci match at HLA-A, -B, and -DRB1  
Median search time   Longer, ≈ 4 mo   Shorter, < 1 mo  
Major limiting factors to graft acquisition   HLA-match; donor attrition/availability   Fixed UCB cell content (especially for larger sized recipients)  
Ease of rearranging date of stem cell infusion   Can be difficult   Easy  
Potential for second HSC graft or DLI from same donor   Yes   No  
Potential for disease transmission to recipient    
    CMV/EBV   Yes   No  
    Congenital disease*  No   Yes  
Risk to donor
 
Uncommon (anesthesia related, surgical complication, emergent blood transfusion)
 
None
 

HSC indicates hematopoietic stem cells; DLI, donor lymphocyte infusion.

*

Such as hereditary spherocytosis, thalassemia, or infant leukemia.

For URD BMT, suitable donors have previously been accepted as HLA matched at HLA-A and HLA-B loci by serology, and at HLA-DRB1 by molecular techniques, with one mismatch allowed by some centers in recipients younger than 36 years.49,63,92,159,160  Additionally, allele-level matching at HLA-A, HLA-B, and HLA-C is increasingly being sought, in conjunction with matching at additional class II loci. This practice may, however, further restrict availability of suitably matched donors. Cell dose is generally not a limiting factor in identifying a marrow donor, although data indicate improved survival when higher cell doses are obtained.4,161, 162, 163 

For UCBT, cell dose is the most critical determinant of outcome, and currently, 4 to 6 antigen HLA-matched grafts are considered acceptable. While the minimum acceptable UCB graft cell dose is yet to be unanimously agreed upon, a minimum threshold of 1.5 × 107 nucleated cells/kg or 1.7 × 105 CD34+ cells/kg has been suggested.35,117  Among 0 to 2 antigen HLA-mismatched grafts, current data suggest that for the same cell dose, survival is superior with better-matched grafts; although, the negative effect of HLA mismatch can be at least partially overcome by a higher cell dose.35,130  Hence, higher cell doses are even more important with HLA-mismatched grafts. While it has become obvious that both HLA match and cell dose must be considered in the algorithm for UCB graft selection, future studies are awaited to delineate their relative importance.

When URD HCT is indicated, initiating a simultaneous search of UCB and marrow donor registries is justifiable, as it will maximize the chance of finding an acceptable source of HSCs, particularly for children. When both a suitable UCB and marrow graft are available on preliminary search, the urgency for HCT and recipient's diagnosis may dictate the best stem cell source for the specific patient. The time required to acquire an identified UCB graft is significantly shorter. Also, since UCB grafts are banked, rescheduling the date of HSC infusion is simple. Hence, in situations in which the patient requires urgent transplantation, or when the patient's disease or medical status may require rescheduling the date of transplantation at short notice, an UCB graft has obvious advantages.

In comparison with BMT, there is relatively less experience with UCBT. However, sufficient data are becoming available to suggest at least comparable efficacy between HLA-matched marrow and UCB for pediatric patients with acute leukemia.28,29,31,32,34,35  Some investigators have also reported encouraging preliminary results after UCBT in young children (< 2.5 years) with leukemia, immunodeficiency, or inborn errors of metabolism.164,165  Of note, available UCB grafts for children this age typically deliver a high TNC dose.

Experience with UCBT for most nonmalignant disorders such as aplastic anemia, and for adults with hematologic malignancies, is more limited at present. In diagnoses in which HCT is associated with higher rates of graft rejection, or in which need for donor lymphocytes may arise after transplantation (eg, in chronic myeloid leukemia [CML]), marrow grafts may be a better option. Moreover, in certain diseases such as CML in chronic phase (< 40 years) or Wiskott-Aldrich syndrome in young children, disease-free survival after HLA-A–, HLA-B–, and HLA-DRB1–matched URD BMT is very good.40,57,96,98,99  For such patients, at present, UCB might be considered, but only if HLA-A–, HLA-B–, and HLA-DRB1–matched URD marrow is not available.

With certain inherited diseases, such as mucopolysaccharidoses or X-linked cerebral adrenoleukodystrophy, the disease manifestations can be rapidly progressive, potentially limiting the usefulness of HCT.11, 12, 13  Hence, the decision to wait for a marrow graft needs to be carefully weighed against the disease stage at presentation and the predicted rate of progression. In adult patients, while cell dose restricts the use of UCBT, results with URD BMT for acute leukemia are sufficiently poor to warrant investigation of UCB in an attempt to reduce nonrelapse mortality.38,51,148,149  A suggested approach to select an URD graft is shown in Figure 4.

Figure 4.

Approach to unrelated donor search for HCT. HLA-mm indicates HLA mismatch; TNC, total nucleated cells; and *, see “Conclusions and Recommendations.”

Figure 4.

Approach to unrelated donor search for HCT. HLA-mm indicates HLA mismatch; TNC, total nucleated cells; and *, see “Conclusions and Recommendations.”

Close modal

Efforts to more rapidly identify and increase the availability of allele-level HLA-matched URD marrow grafts are ongoing by marrow donor registries worldwide. Alternatively, the majority of UCB searches are likely to identify a 0 to 2 antigen–mismatched graft for pediatric and many adult patients, even within the currently accessible UCB pool. With growing awareness and public interest, the number of UCB units being harvested and stored is rapidly expanding. Also, programs aimed at increasing the pool of uncommon haplotypes by recruiting ethnic/racial minority donors might meet with higher success for UCB “donors” compared with marrow donors. In the future, with a larger pool of stored UCB units, and with possibly increased representation of minority haplotypes, there will be an increased likelihood of identifying UCB units with 0 to 1 HLA mismatch. Unfortunately, currently a UCB search requires initiation of separate queries with individual cord blood banks, and different banks have diverse policies on methods of HLA typing and cell content assessment at harvest. This can make comparisons between different search reports difficult. The development of a unified system that will allow for a single search, with access to all available UCB units worldwide that have been stored by banks using uniform standards, will make the search process faster, more accurate, and comparisons of results after UCBT more reliable.

Prepublished online as Blood First Edition Paper, January 9, 2003; DOI 10.1182/blood-2002-08-2510.

Supported in part by Children's Cancer Research Fund (J.N.B., J.E.W., S.M.D.), and the National Institute of Health (P01 CA 21737 [J.E.W.], N01 HB 67139 [J.E.W.], and N01 HB 47095 [J.E.W., S.M.D.]).

S.M.D. and J.E.W. contributed equally to this paper.

The authors would like to acknowledge and thank the National Marrow Donor Program (NMDP) for providing registry data. We thank the staff at the University of Minnesota Blood and Marrow Transplant Program for their assistance in the investigation of the clinical role of UCBT. The authors also acknowledge Dr Norma K. C. Ramsay for her critical review of the manuscript and helpful suggestions.

1
Thomas E, Storb R, Clift RA, et al. Bone-marrow transplantation (first of two parts).
N Engl J Med
.
1975
;
292
:
832
-843.
2
Thomas ED, Buckner CD, Banaji M, et al. One hundred patients with acute leukemia treated by chemotherapy, total body irradiation, and allogeneic marrow transplantation.
Blood
.
1977
;
49
:
511
-533.
3
Blume KG, Beutler E, Bross KJ, et al. Bone-marrow ablation and allogeneic marrow transplantation in acute leukemia.
N Engl J Med
.
1980
;
302
:
1041
-1046.
4
Bortin MM, Gale RP, Kay HE, Rimm AA. Bone marrow transplantation for acute myelogenous leukemia: factors associated with early mortality.
JAMA
.
1983
;
249
:
1166
-1175.
5
Speck B, Bortin MM, Champlin R, et al. Allogeneic bone-marrow transplantation for chronic myelogenous leukaemia.
Lancet
.
1984
;
1
:
665
-668.
6
Walters MC, Patience M, Leisenring W, et al. Bone marrow transplantation for sickle cell disease.
N Engl J Med
.
1996
;
335
:
369
-376.
7
Lucarelli G, Galimberti M, Polchi P, et al. Bone marrow transplantation in patients with thalassemia.
N Engl J Med
.
1990
;
322
:
417
-421.
8
Storb R, Thomas ED, Buckner CD, et al. Allogeneic marrow grafting for treatment of aplastic anemia.
Blood
.
1974
;
43
:
157
-180.
9
Gluckman E, Auerbach AD, Horowitz MM, et al. Bone marrow transplantation for Fanconi anemia.
Blood
.
1995
;
86
:
2856
-2862.
10
Buckley RH. Bone marrow reconstitution in primary immunodeficiency.
Clin Immunol Principles Practice
.
1995
;
2
:
1813
-1830.
11
Shapiro E, Krivit W, Lockman L, et al. Long-term effect of bone-marrow transplantation for childhood-onset cerebral X-linked adrenoleukodystrophy.
Lancet
.
2000
;
356
:
713
-718.
12
Krivit W, Aubourg P, Shapiro E, Peters C. Bone marrow transplantation for globoid cell leukodystrophy, adrenoleukodystrophy, metachromatic leukodystrophy, and Hurler syndrome.
Curr Opin Hematol
.
1999
;
6
:
377
-382.
13
Peters C, Shapiro EG, Krivit W. Hurler syndrome: past, present, and future.
J Pediatr
.
1998
;
133
:
7
-9.
14
Amos DB. Human histocompatibility locus HL-A.
Science
.
1968
;
159
:
659
-660.
15
Bodmer WF. HLA: what's in a name? A commentary on HLA nomenclature development over the years.
Tissue Antigens
.
1997
;
49
:
293
-296.
16
Ferrara JL, Deeg HJ. Graft-versus-host disease.
N Engl J Med
.
1991
;
324
:
667
-674.
17
Horowitz SD, Bach FH, Groshong T, Hong R, Yunis EJ. Treatment of severe combined immuno-deficiency with bone-marrow from an unrelated, mixed-leucocyte-culture-non-reactive donor.
Lancet
.
1975
;
2
:
431
-433.
18
Lohrmann HP, Dietrich M, Goldmann SF, et al. Bone marrow transplantation for aplastic anaemia from a HL-A and MLC-identical unrelated donor.
Blut
.
1975
;
31
:
347
-354.
19
Speck B, Zwaan FE, van Rood JJ, Eernisse JG. Allogeneic bone marrow transplantation in a patient with aplastic anemia using a phenotypically HL-A-identifcal unrelated donor.
Transplantation
.
1973
;
16
:
24
-28.
20
Green A, Clarke E, Hunt L, et al. Children with acute lymphoblastic leukemia who receive T-cell-depleted HLA mismatched marrow allografts from unrelated donors have an increased incidence of primary graft failure but a similar overall transplant outcome.
Blood
.
1999
;
94
:
2236
-2246.
21
Hale G, Waldmann H. Control of graft-versus-host disease and graft rejection by T cell depletion of donor and recipient with Campath-1 antibodies: results of matched sibling transplants for malignant diseases.
Bone Marrow Transplant
.
1994
;
13
:
597
-611.
22
Novitzky N, Thomas V, Hale G, Waldmann H. Ex vivo depletion of T cells from bone marrow grafts with CAMPATH-1 in acute leukemia: graft-versus-host disease and graft-versus-leukemia effect.
Transplantation
.
1999
;
67
:
620
-626.
23
Papadopoulos EB, Carabasi MH, Castro-Malaspina H, et al. T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: freedom from relapse in the absence of graft-versus-host disease.
Blood
.
1998
;
91
:
1083
-1090.
24
Antin JH, Bierer BE, Smith BR, et al. Selective depletion of bone marrow T lymphocytes with anti-CD5 monoclonal antibodies: effective prophylaxis for graft-versus-host disease in patients with hematologic malignancies.
Blood
.
1991
;
78
:
2139
-2149.
25
Confer DL. Unrelated marrow donor registries.
Curr Opin Hematol
.
1997
;
4
:
408
-412.
26
Wagner JE, Rosenthal J, Sweetman R, et al. Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: analysis of engraftment and acute graft-versus-host disease.
Blood
.
1996
;
88
:
795
-802.
27
Kurtzberg J, Laughlin M, Graham ML, et al. Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients.
N Engl J Med
.
1996
;
335
:
157
-166.
28
Gluckman E, Rocha V, Boyer-Chammard A, et al. Outcome of cord-blood transplantation from related and unrelated donors: Eurocord Transplant Group and the European Blood and Marrow Transplantation Group.
N Engl J Med
.
1997
;
337
:
373
-381.
29
Rubinstein P, Carrier C, Scaradavou A, et al. Outcomes among 562 recipients of placental-blood transplants from unrelated donors.
N Engl J Med
.
1998
;
339
:
1565
-1577.
30
Thomson BG, Robertson KA, Gowan D, et al. Analysis of engraftment, graft-versus-host disease, and immune recovery following unrelated donor cord blood transplantation.
Blood
.
2000
;
96
:
2703
-2711.
31
Rocha V, Cornish J, Sievers EL, et al. Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia.
Blood
.
2001
;
97
:
2962
-2971.
32
Locatelli F, Rocha V, Chastang C, et al. Factors associated with outcome after cord blood transplantation in children with acute leukemia: Eurocord-Cord Blood Transplant Group.
Blood
.
1999
;
93
:
3662
-3671.
33
Laughlin MJ, Barker J, Bambach B, et al. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors.
N Engl J Med
.
2001
;
344
:
1815
-1822.
34
Barker JN, Davies SM, DeFor T, et al. Survival after transplantation of unrelated donor umbilical cord blood is comparable to that of human leukocyte antigen-matched unrelated donor bone marrow: results of a matched-pair analysis.
Blood
.
2001
;
97
:
2957
-2961.
35
Wagner JE, Barker JN, DeFor TE, et al. Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival.
Blood
.
2002
;
100
:
1611
-1618.
36
Howe CW, Radde-Stepaniak T. Hematopoietic cell donor registries. In: Thomas E, Blume KG, Forman SJ, eds.
Hematopoietic Cell Transplantation
, 2nd ed. Malden, MA: Blackwell Science;
1999
:
503
-514.
37
McCullough J, Hansen J, Perkins H, Stroncek D, Bartsch G. The National Marrow Donor Program: how it works, accomplishments to date.
Oncology (Huntingt)
.
1989
;
3
:
63
-68,72; discussion 72-74.
38
Kernan NA, Bartsch G, Ash RC, et al. Analysis of 462 transplantations from unrelated donors facilitated by the National Marrow Donor Program.
N Engl J Med
.
1993
;
328
:
593
-602.
39
Davies SM, Wagner JE, Shu XO, et al. Unrelated donor bone marrow transplantation for children with acute leukemia.
J Clin Oncol
.
1997
;
15
:
557
-565.
40
McGlave P, Bartsch G, Anasetti C, et al. Unrelated donor marrow transplantation therapy for chronic myelogenous leukemia: initial experience of the National Marrow Donor Program.
Blood
.
1993
;
81
:
543
-550.
41
McGlave PB, Shu XO, Wen W, et al. Unrelated donor marrow transplantation for chronic myelogenous leukemia: 9 years' experience of the national marrow donor program.
Blood
.
2000
;
95
:
2219
-2225.
42
Davies SM, Ramsay NK, Weisdorf DJ. Feasibility and timing of unrelated donor identification for patients with ALL.
Bone Marrow Transplant
.
1996
;
17
:
737
-740.
43
Dupont B. Immunology of hematopoietic stem cell transplantation: a brief review of its history.
Immunol Rev
.
1997
;
157
:
5
-12.
44
Beatty PG, Dahlberg S, Mickelson EM, et al. Probability of finding HLA-matched unrelated marrow donors.
Transplantation
.
1988
;
45
:
714
-718.
45
Sonnenberg FA, Eckman MH, Pauker SG. Bone marrow donor registries: the relation between registry size and probability of finding complete and partial matches.
Blood
.
1989
;
74
:
2569
-2578.
46
Takahashi K, Juji T, Miyazaki H. Determination of an appropriate size of unrelated donor pool to be registered for HLA-matched bone marrow transplantation.
Transfusion
.
1989
;
29
:
311
-316.
47
Davies SM, Kollman C, Anasetti C, et al. Engraftment and survival after unrelated-donor bone marrow transplantation: a report from the National Marrow Donor Program.
Blood
.
2000
;
96
:
4096
-4102.
48
Davies SM, Ramsay NK, Haake RJ, et al. Comparison of engraftment in recipients of matched sibling of unrelated donor marrow allografts.
Bone Marrow Transplant
.
1994
;
13
:
51
-57.
49
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
;
320
:
197
-204.
50
Ash RC, Horowitz MM, Gale RP, et al. Bone marrow transplantation from related donors other than HLA-identical siblings: effect of T cell depletion.
Bone Marrow Transplant
.
1991
;
7
:
443
-452.
51
Szydlo R, Goldman JM, Klein JP, et al. Results of allogeneic bone marrow transplants for leukemia using donors other than HLA-identical siblings.
J Clin Oncol
.
1997
;
15
:
1767
-1777.
52
Beatty PG, Clift RA, Mickelson EM, et al. Marrow transplantation from related donors other than HLA-identical siblings.
N Engl J Med
.
1985
;
313
:
765
-771.
53
Petersdorf EW, Longton GM, Anasetti C, et al. Association of HLA-C disparity with graft failure after marrow transplantation from unrelated donors.
Blood
.
1997
;
89
:
1818
-1823.
54
Petersdorf EW, Gooley TA, Anasetti C, et al. Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient.
Blood
.
1998
;
92
:
3515
-3520.
55
Trigg ME, Billing R, Sondel PM, et al. Clinical trial depleting T lymphocytes from donor marrow for matched and mismatched allogeneic bone marrow transplants.
Cancer Treat Rep
.
1985
;
69
:
377
-386.
56
Kernan NA, Bordignon C, Heller G, et al. Graft failure after T-cell-depleted human leukocyte antigen identical marrow transplants for leukemia: I. Analysis of risk factors and results of secondary transplants.
Blood
.
1989
;
74
:
2227
-2236.
57
Hansen JA, Gooley TA, Martin PJ, et al. Bone marrow transplants from unrelated donors for patients with chronic myeloid leukemia.
N Engl J Med
.
1998
;
338
:
962
-968.
58
MacMillan ML, Auerbach AD, Davies SM, et al. Haematopoietic cell transplantation in patients with Fanconi anaemia using alternate donors: results of a total body irradiation dose escalation trial.
Br J Haematol
.
2000
;
109
:
121
-129.
59
Storb R, Thomas ED, Weiden PL, et al. Aplastic anemia treated by allogeneic bone marrow transplantation: a report on 49 new cases from Seattle.
Blood
.
1976
;
48
:
817
-841.
60
Storb R, Thomas ED, Buckner CD, et al. Allogeneic marrow grafting for treatment of aplastic anemia: a follow-up on long-term survivors.
Blood
.
1976
;
48
:
485
-490.
61
Peters C, Balthazor M, Shapiro EG, et al. Outcome of unrelated donor bone marrow transplantation in 40 children with Hurler syndrome.
Blood
.
1996
;
87
:
4894
-4902.
62
Peters C, Shapiro EG, Anderson J, et al. Hurler syndrome: II. Outcome of HLA-genotypically identical sibling and HLA-haploidentical related donor bone marrow transplantation in fifty-four children: the Storage Disease Collaborative Study Group.
Blood
.
1998
;
91
:
2601
-2608.
63
Balduzzi A, Gooley T, Anasetti C, et al. Unrelated donor marrow transplantation in children.
Blood
.
1995
;
86
:
3247
-3256.
64
Sierra J, Storer B, Hansen JA, et al. Transplantation of marrow cells from unrelated donors for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching, and marrow cell dose.
Blood
.
1997
;
89
:
4226
-4235.
65
Anasetti C, Beatty PG, Storb R, et al. Effect of HLA incompatibility on graft-versus-host disease, relapse, and survival after marrow transplantation for patients with leukemia or lymphoma.
Hum Immunol
.
1990
;
29
:
79
-91.
66
Beatty PG, Anasetti C, Hansen JA, et al. Marrow transplantation from unrelated donors for treatment of hematologic malignancies: effect of mismatching for one HLA locus.
Blood
.
1993
;
81
:
249
-253.
67
Petersdorf EW, Longton GM, Anasetti C, et al. The significance of HLA-DRB1 matching on clinical outcome after HLA-A, B, DR identical unrelated donor marrow transplantation.
Blood
.
1995
;
86
:
1606
-1613.
68
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.
69
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.
70
Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
.
1990
;
75
:
555
-562.
71
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.
72
Gale RP, Bortin MM, van Bekkum DW, et al. Risk factors for acute graft-versus-host disease.
Br J Haematol
.
1987
;
67
:
397
-406.
73
Martin PJ, Schoch G, Fisher L, et al. A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment.
Blood
.
1990
;
76
:
1464
-1472.
74
Weisdorf D, Haake R, Blazar B, et al. Treatment of moderate/severe acute graft-versus-host disease after allogeneic bone marrow transplantation: an analysis of clinical risk features and outcome.
Blood
.
1990
;
75
:
1024
-1030.
75
Ratanatharathorn V, Nash RA, Przepiorka D, et al. Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation.
Blood
.
1998
;
92
:
2303
-2314.
76
Nash RA, Antin JH, Karanes C, et al. Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors.
Blood
.
2000
;
96
:
2062
-2068.
77
Sullivan KM, Agura E, Anasetti C, et al. Chronic graft-versus-host disease and other late complications of bone marrow transplantation.
Semin Hematol
.
1991
;
28
:
250
-259.
78
Atkinson K, Horowitz MM, Gale RP, et al. Risk factors for chronic graft-versus-host disease after HLA-identical sibling bone marrow transplantation.
Blood
.
1990
;
75
:
2459
-2464.
79
Aversa F, Tabilio A, Velardi A, et al. Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype.
N Engl J Med
.
1998
;
339
:
1186
-1193.
80
Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment of T-cell-depleted haploidentical “three-loci” incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells to bone marrow inoculum.
Blood
.
1994
;
84
:
3948
-3955.
81
Soiffer RJ, Fairclough D, Robertson M, et al. CD6-depleted allogeneic bone marrow transplantation for acute leukemia in first complete remission.
Blood
.
1997
;
89
:
3039
-3047.
82
Drobyski WR, Hessner MJ, Klein JP, et al. T-cell depletion plus salvage immunotherapy with donor leukocyte infusions as a strategy to treat chronicphase chronic myelogenous leukemia patients undergoing HLA-identical sibling marrow transplantation.
Blood
.
1999
;
94
:
434
-441.
83
Hale G, Zhang MJ, Bunjes D, et al. Improving the outcome of bone marrow transplantation by using CD52 monoclonal antibodies to prevent graft-versus-host disease and graft rejection.
Blood
.
1998
;
92
:
4581
-4590.
84
Sehn LH, Alyea EP, Weller E, et al. Comparative outcomes of T-cell-depleted and non-T-cell-depleted allogeneic bone marrow transplantation for chronic myelogenous leukemia: impact of donor lymphocyte infusion.
J Clin Oncol
.
1999
;
17
:
561
-568.
85
Martin PJ, Rowley SD, Anasetti C, et al. A phase I-II clinical trial to evaluate removal of CD4 cells and partial depletion of CD8 cells from donor marrow for HLA-mismatched unrelated recipients.
Blood
.
1999
;
94
:
2192
-2199.
86
Wagner J, Thompson J, Carter S, Jensen L, Kernan N. Impact of graft-versus-host disease prophylaxis on 3 year disease free survival: results of a multi-center, randomized phase II-III trial comparing T-cell depletion/cyclosporine and methotrexate/cyclosporine in recipients of unrelated donor bone marrow [abstract].
Blood
.
2002
;
100
:
75a
87
Lum LG. The kinetics of immune reconstitution after human marrow transplantation.
Blood
.
1987
;
69
:
369
-380.
88
Small TN, Papadopoulos EB, Boulad F, et al. Comparison of immune reconstitution after unrelated and related T-cell-depleted bone marrow transplantation: effect of patient age and donor leukocyte infusions.
Blood
.
1999
;
93
:
467
-480.
89
Godthelp BC, van Tol MJ, Vossen JM, van Den Elsen PJ. T-cell immune reconstitution in pediatric leukemia patients after allogeneic bone marrow transplantation with T-cell-depleted or unmanipulated grafts: evaluation of overall and antigen-specific T-cell repertoires.
Blood
.
1999
;
94
:
4358
-4369.
90
Atkinson K. Reconstruction of the haemopoietic and immune systems after marrow transplantation.
Bone Marrow Transplant
.
1990
;
5
:
209
-226.
91
Parkman R, Weinberg KI. Immunological reconstitution following bone marrow transplantation.
Immunol Rev
.
1997
;
157
:
73
-78.
92
Davies SM, Shu XO, Blazar BR, et al. Unrelated donor bone marrow transplantation: influence of HLA A and B incompatibility on outcome.
Blood
.
1995
;
86
:
1636
-1642.
93
Rufer N, Tiercy JM, Breur-Vriesendorp B, et al. Histoincompatibilities in ABDR-matched unrelated donor recipient combinations.
Bone Marrow Transplant
.
1995
;
16
:
641
-646.
94
Petersdorf EW, Longton GM, Anasetti C, et al. Definition of HLA-DQ as a transplantation antigen.
Proc Natl Acad Sci U S A
.
1996
;
93
:
15358
-15363.
95
La Nasa G, Giardini C, Argiolu F, et al. Unrelated donor bone marrow transplantation for thalassemia: the effect of extended haplotypes.
Blood
.
2002
;
99
:
4350
-4356.
96
Spencer A, Szydlo RM, Brookes PA, et al. Bone marrow transplantation for chronic myeloid leukemia with volunteer unrelated donors using ex vivo or in vivo T-cell depletion: major prognostic impact of HLA class I identity between donor and recipient.
Blood
.
1995
;
86
:
3590
-3597.
97
Filipovich AH, Shapiro RS, Ramsay NK, et al. Unrelated donor bone marrow transplantation for correction of lethal congenital immunodeficiencies.
Blood
.
1992
;
80
:
270
-276.
98
Lenarsky C, Weinberg K, Kohn DB, Parkman R. Unrelated donor BMT for Wiskott-Aldrich syndrome.
Bone Marrow Transplant
.
1993
;
12
:
145
-147.
99
Filipovich A. Stem cell transplantation from unrelated donors for correction of primary immunodeficiencies.
Organ Bone Marrow Transplant
.
1996
;
16
:
377
-393.
100
Gluckman E, Broxmeyer HA, Auerbach AD, et al. Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical-cord blood from an HLA-identical sibling.
N Engl J Med
.
1989
;
321
:
1174
-1178.
101
Wagner JE, Kernan NA, Steinbuch M, Broxmeyer HE, Gluckman E. Allogeneic sibling umbilicalcord-blood transplantation in children with malignant and non-malignant disease.
Lancet
.
1995
;
346
:
214
-219.
102
Rubinstein P, Taylor PE, Scaradavou A, et al. Unrelated placental blood for bone marrow reconstitution: organization of the placental blood program.
Blood Cells
.
1994
;
20
:
587
-596.
103
Kurtzberg J, Graham M, Casey J, et al. The use of umbilical cord blood in mismatched related and unrelated hemopoietic stem cell transplantation.
Blood Cells
.
1994
;
20
:
275
-283.
104
Wagner JE. Allogeneic cord blood transplantation. In: Winter JN, ed.
Blood Stem Cell Transplantation
. Boston, MA: Kluwer Academic Publishers;
1997
:
187
-216.
105
Gluckman E. Current status of umbilical cord blood hematopoietic stem cell transplantation.
Exp Hematol
.
2000
;
28
:
1197
-1205.
106
Silberstein LE, Jefferies LC. Placental-blood banking—a new frontier in transfusion medicine.
N Engl J Med
.
1996
;
335
:
199
-201.
107
Rubinstein P, Rosenfield RE, Adamson JW, Stevens CE. Stored placental blood for unrelated bone marrow reconstitution.
Blood
.
1993
;
81
:
1679
-1690.
108
Barker J, Krepski T, De For TE, Wagner J, Weisdorf D. Searching for unrelated donor hematopoietic stem cell grafts: availability and speed of umblical cord blood versus bone marrow.
Biol Blood Marrow Transplant
.
2002
;
8
:
257
-260.
109
Rubinstein P. Placental blood-derived hematopoietic stem cells for unrelated bone marrow reconstitution.
J Hematother
.
1993
;
2
:
207
-210.
110
Broxmeyer HE, Douglas GW, Hangoc G, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells.
Proc Natl Acad Sci U S A
.
1989
;
86
:
3828
-3832.
111
Broxmeyer HE, Hangoc G, Cooper S, et al. Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation in adults.
Proc Natl Acad Sci U S A
.
1992
;
89
:
4109
-4113.
112
Lapidot T, Pflumio F, Doedens M, et al. Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice.
Science
.
1992
;
255
:
1137
-1141.
113
Vormoor J, Lapidot T, Pflumio F, et al. Immature human cord blood progenitors engraft and proliferate to high levels in severe combined immunodeficient mice.
Blood
.
1994
;
83
:
2489
-2497.
114
Hogan CJ, Shpall EJ, McNulty O, et al. Engraftment and development of human CD34(+)-enriched cells from umbilical cord blood in NOD/LtSz-scid/scid mice.
Blood
.
1997
;
90
:
85
-96.
115
Rubinstein P, Stevens CE. Placental blood for bone marrow replacement: the New York Blood Center's program and clinical results.
Baillieres Best Pract Res Clin Haematol
.
2000
;
13
:
565
-584.
116
Rocha V, Chastang C, Laporte JP, et al. Unrelated umblical cord blood transplants in adults with hematologic malignancies [abstract].
Blood
.
1998
;
92
:
144a
.
117
Gluckman E. Hematopoietic stem-cell transplants using umbilical-cord blood.
N Engl J Med
.
2001
;
344
:
1860
-1861.
118
Locatelli F, Maccario R, Comoli P, et al. Hematopoietic and immune recovery after transplantation of cord blood progenitor cells in children.
Bone Marrow Transplant
.
1996
;
18
:
1095
-1101.
119
Giraud P, Thuret I, Reviron D, et al. Immune reconstitution and outcome after unrelated cord blood transplantation: a single paediatric institution experience.
Bone Marrow Transplant
.
2000
;
25
:
53
-57.
120
Comoli P, Locatelli F, Moretta A, et al. Human alloantigen-specific anergic cells induced by a combination of CTLA4-Ig and CsA maintain anti-leukemia and anti-viral cytotoxic responses.
Bone Marrow Transplant
.
2001
;
27
:
1263
-1273.
121
Niehues T, Rocha V, Filipovich AH, et al. Factors affecting lymphocyte subset reconstitution after either related or unrelated cord blood transplantation in children—a Eurocord analysis.
Br J Haematol
.
2001
;
114
:
42
-48.
122
Moretta A, Maccario R, Fagioli F, et al. Analysis of immune reconstitution in children undergoing cord blood transplantation.
Exp Hematol
.
2001
;
29
:
371
-379.
123
Talvensaari K, Clave E, Douay C, et al. A broad T-cell repertoire diversity and an efficient thymic function indicate a favorable long-term immune reconstitution after cord blood stem cell transplantation.
Blood
.
2002
;
99
:
1458
-1464.
124
Linch DC, Brent L. Marrow transplantation: can cord blood be used?
Nature
.
1989
;
340
:
676
.
125
Butturini A, Gale RP. The role of T-cells in preventing relapse in chronic myelogenous leukemia.
Bone Marrow Transplant
.
1987
;
2
:
351
-354.
126
Butturini A, Bortin MM, Gale RP. Graft-versus-leukemia following bone marrow transplantation.
Bone Marrow Transplant
.
1987
;
2
:
233
-242.
127
Gale RP, Horowitz MM, Ash RC, et al. Identical-twin bone marrow transplants for leukemia.
Ann Intern Med
.
1994
;
120
:
646
-652.
128
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.
129
Rubinstein P, Kurtzberg J, Loberiza FR, et al. Comparison of unrelated cord blood and unrelated bone marrow transplants for leukemia in children: a collaborative study of the New York Blood Center and the International Bone Marrow Transplant Registry [abstract].
Blood
.
2001
;
98
:
814a
.
130
Rubinstein P, Carrier C, Carpenter C, et al. Graft selection in unrelated placental/umbilical cord blood (PCB) transplantation: influence and weight of HLA match and cell dose on engraftment and survival [abstract].
Blood
.
2000
;
96
:
588a
.
131
Stevens CE, Gladstone J, Taylor PE, et al. Placental/umbilical cord blood for unrelated-donor bone marrow reconstitution: relevance of nucleated red blood cells.
Blood
.
2002
;
100
:
2662
-2664.
132
Migliaccio AR, Adamson JW, Stevens CE, et al. Cell dose and speed of engraftment in placental/umbilical cord blood transplantation: graft progenitor cell content is a better predictor than nucleated cell quantity.
Blood
.
2000
;
96
:
2717
-2722.
133
Rocha V, Wagner JE Jr, Sobocinski KA, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling: Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources.
N Engl J Med
.
2000
;
342
:
1846
-1854.
134
Hongeng S, Krance RA, Bowman LC, et al. Outcomes of transplantation with matched-sibling and unrelated-donor bone marrow in children with leukaemia.
Lancet
.
1997
;
350
:
767
-771.
135
Harris DT, Schumacher MJ, Locascio J, et al. Phenotypic and functional immaturity of human umbilical cord blood T lymphocytes.
Proc Natl Acad Sci U S A
.
1992
;
89
:
10006
-10010.
136
Garderet L, Dulphy N, Douay C, et al. The umbilical cord blood alphabeta T-cell repertoire: characteristics of a polyclonal and naive but completely formed repertoire.
Blood
.
1998
;
91
:
340
-346.
137
Madrigal JA, Cohen SB, Gluckman E, Charron DJ. Does cord blood transplantation result in lower graft-versus-host disease? It takes more than two to tango.
Hum Immunol
.
1997
;
56
:
1
-5.
138
Takahashi N, Imanishi K, Nishida H, Uchiyama T. Evidence for immunologic immaturity of cord blood T cells: cord blood T cells are susceptible to tolerance induction to in vitro stimulation with a superantigen.
J Immunol
.
1995
;
155
:
5213
-5219.
139
Risdon G, Gaddy J, Horie M, Broxmeyer HE. Alloantigen priming induces a state of unresponsiveness in human umbilical cord blood T cells.
Proc Natl Acad Sci U S A
.
1995
;
92
:
2413
-2417.
140
Ho VT, Soiffer RJ. The history and future of T-cell depletion as graft-versus-host disease prophylaxis for allogeneic hematopoietic stem cell transplantation.
Blood
.
2001
;
98
:
3192
-3204.
141
Roux E, Helg C, Dumont-Girard F, et al. Analysis of T-cell repopulation after allogeneic bone marrow transplantation: significant differences between recipients of T-cell depleted and unmanipulated grafts.
Blood
.
1996
;
87
:
3984
-3992.
142
Granberg C, Hirvonen T. Cell-mediated lympholysis by fetal and neonatal lymphocytes in sheep and man.
Cell Immunol
.
1980
;
51
:
13
-22.
143
Moretta A, Comoli P, Montagna D, et al. High frequency of Epstein-Barr virus (EBV) lymphoblastoid cell line-reactive lymphocytes in cord blood: evaluation of cytolytic activity and IL-2 production.
Clin Exp Immunol
.
1997
;
107
:
312
-320.
144
Moretta A, Locatelli F, Mingrat G, et al. Characterisation of CTL directed towards non-inherited maternal alloantigens in human cord blood.
Bone Marrow Transplant
.
1999
;
24
:
1161
-1166.
145
Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
Science
.
2002
;
295
:
2097
-2100.
146
Han P, Hodge G, Story C, Xu X. Phenotypic analysis of functional T-lymphocyte subtypes and natural killer cells in human cord blood: relevance to umbilical cord blood transplantation.
Br J Haematol
.
1995
;
89
:
733
-740.
147
Umemoto M, Azuma E, Hirayama M, et al. Two cytotoxic pathways of natural killer cells in human cord blood: implications in cord blood transplantation.
Br J Haematol
.
1997
;
98
:
1037
-1040.
148
Weisdorf DJ. Bone marrow transplantation for acute lymphocytic leukemia (ALL).
Leukemia
.
1997
;
11
(suppl 4):
S20
-S22.
149
Madrigal JA, Scott I, Arguello R, et al. Factors influencing the outcome of bone marrow transplants using unrelated donors.
Immunol Rev
.
1997
;
157
:
153
-166.
150
Schiller G, Feig SA, Territo M, et al. Treatment of advanced acute leukaemia with allogeneic bone marrow transplantation from unrelated donors.
Br J Haematol
.
1994
;
88
:
72
-78.
151
Sanz GF, Saavedra S, Planelles D, et al. Standardized, unrelated donor cord blood transplantation in adults with hematologic malignancies.
Blood
.
2001
;
98
:
2332
-2338.
152
Shpall EJ, Quinones R, Hami L, et al. Transplantation of cancer patients receiving high dose chemotherapy with ex-vivo expanded cord blood cells [abstract].
Blood
.
1998
;
92
:
646a
.
153
Stiff P, Pecora A, Parthasarathy M, et al. Umblical cord blood transplants in adults using a combination of unexpanded and ex-vivo expanded cells: preliminary clinical observations [abstract].
Blood
.
1998
;
92
:
646a
.
154
Ruggieri L, Heimfeld S, Broxmeyer HE. Cytokine-dependent ex vivo expansion of early subsets of CD34+ cord blood myeloid progenitors is enhanced by cord blood plasma, but expansion of the more mature subsets of progenitors is favored.
Blood Cells
.
1994
;
20
:
436
-454.
155
Hows JM, Bradley BA, Marsh JC, et al. Growth of human umbilical-cord blood in longterm haemopoietic cultures.
Lancet
.
1992
;
340
:
73
-76.
156
Barker JN, Weisdorf DJ, Wagner JE. Creation of a double chimera after the transplantation of umbilical-cord blood from two partially matched unrelated donors.
N Engl J Med
.
2001
;
344
:
1870
-1871.
157
Rizzieri DA, Long GD, Vredenburgh JJ, et al. Successful allogeneic engraftment of mismatched unrelated cord blood following a nonmyeloablative preparative regimen.
Blood
.
2001
;
98
:
3486
-3488.
158
Barker J, Weisdorf D, De For TE, et al. Impact of multiple unit unrelated donor cord blood transplantation in adults: preliminary analysis of safety and efficacy [abstract].
Blood
.
2001
;
98
:
666a
.
159
Beatty PG, Hansen JA, Longton GM, et al. Marrow transplantation from HLA-matched unrelated donors for treatment of hematologic malignancies.
Transplantation
.
1991
;
51
:
443
-447.
160
Ottinger HD, Albert E, Arnold R, et al. German consensus on immunogenetic donor search for transplantation of allogeneic bone marrow and peripheral blood stem cells.
Bone Marrow Transplant
.
1997
;
20
:
101
-105.
161
Sierra J, Storer B, Hansen JA, et al. Unrelated donor marrow transplantation for acute myeloid leukemia: an update of the Seattle experience.
Bone Marrow Transplant
.
2000
;
26
:
397
-404.
162
Niederwieser D, Pepe M, Storb R, Loughran TP Jr, Longton G. Improvement in rejection, engraftment rate and survival without increase in graft-versus-host disease by high marrow cell dose in patients transplanted for aplastic anaemia.
Br J Haematol
.
1988
;
69
:
23
-28.
163
Mavroudis D, Read E, Cottler-Fox M, et al. CD34+ cell dose predicts survival, posttransplant morbidity, and rate of hematologic recovery after allogeneic marrow transplants for hematologic malignancies.
Blood
.
1996
;
88
:
3223
-3229.
164
Kurtzberg J, Scaradavou M, Wagner J, et al. Banked umbilical cord blood is an excellent source of donor hematopoietic stem cells for infants with malignant and nonmalignant conditions lacking a related donor [abstract].
Blood
.
2000
;
96
:
587a
.
165
Staba S, Martin P, Ciocci G, Allison-Thacker J, Kurtzberg J. Correction of Hurler syndrome with unrelated umbilical cord blood transplantation [abstract].
Blood
.
2001
;
98
:
667a
.
Sign in via your Institution