Only 30% of patients who require an allogeneic hematopoietic cell transplant will have an HLA-matched sibling donor. A search for an unrelated donor will be undertaken for patients without a matched family donor. However, many patients, particularly patients of diverse racial and ethnic backgrounds, may not be able to rapidly identify a suitably matched unrelated donor. Three alternative graft sources, umbilical cord blood (UCB), haploidentical (haplo)–related donor, and mismatched unrelated donor (MMUD) are available. UCB is associated with decreased GVHD, but hematologic recovery and immune reconstitution are slow. Haplo-HCT is characterized by donor availability for transplantation and after transplantation adoptive cellular immunotherapy but may be complicated by a high risk of graft failure and relapse. A MMUD transplant may also be an option, but GVHD may be of greater concern. Phase 2 studies have documented advances in HLA typing, GVHD prophylaxis, and infection prevention, which have improved survival. The same patient evaluated in different transplant centers may be offered MMUD, UCB, or haplo-HCT depending on center preference. In this review, we discuss the rationale for donor choice and the need of phase 3 studies to help answer this important question.

Patient 1 is a 62-year-old woman of Hispanic and Native American descent with a history of breast cancer, treated with surgery and chemotherapy. She is in complete remission; 5 years later, she develops acute myelogenous leukemia (AML) with monosomy 7 karyotype. She attains a complete remission, has an Eastern Cooperative Oncology Group performance status of 1, and reduced intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation (HCT) is recommended. She has 3 siblings, but none is a HLA full (8/8 HLA- A,-B,-C,-DR) match. One sibling is a haploidentical (haplo) match. Her HLA typing is entered into the National Marrow Donor Program (NMDP), but no fully matched donor is identified. An unrelated donor with a single allele level mismatch at HLA-B is identified. Search of the umbilical cord blood (UCB) registries reveals 2 HLA 4/6 (-A,- B, -DR matches) UCB units with a combined cell dose of 5.3 × 107 nucleated cells/kg.

Most transplant physicians would agree that the only curative therapy for therapy-related AML is allogeneic HCT, and many would favor an RIC regimen for this patient. However, this patient might be offered a haplo-HCT from her sibling at one transplant center, a double UCB transplant (dUCBT) at a different center, and a mismatched unrelated transplant (MMUD) in another part of the country. All of these options have curative potential but significant mortality and morbidity. How is this patient to decide? What can her hematologists do to assist her?

Only 30% of patients will have a matched sibling donor. The NMDP was established in 1986 and now boasts more than 16 million volunteer donors.1  Today, approximately 60% of white, but only 20% to 45% of black and other minority patients, will be able to find an 8/8 matched unrelated donor (MUD) at HLA-A, -B, -C, and -DR. An estimated 5000 patients each year are candidates for MMUD, UCBT, or haplo-HCT.2  These 3 alternative graft sources have never been compared in a randomized fashion. In this review, we address the outcome data for each approach in adults with hematologic malignancies, comparing differences in GVHD, relapse, infection, second malignancies, and cost. We discuss current graft selection strategies and future developments.

UCB was first successfully transplanted into a child with Fanconi anemia in 1988.3  Expanding on the success in pediatric UCBT pioneered by Drs Kurtzberg, Gluckman, Wagner, Broxmeyer, and others, the field grew; by 2011, more than 25 000 UCBTs have been performed worldwide and more than 500 000 UCB units have been donated for public use.2,4  UCB is readily available and donors can be found for a diverse patient population.5  For example, in New York, Barker et al studied 525 patients; 56% of UCBT patients were of non-European descent, compared with 23% of MUD patients.5 

Myeloablative or high intensity transplant

Improvements in patient selection, UCB unit choice, and infection prophylaxis have led to improved UCBT outcomes. Ooi et al reported outstanding results with 5-year disease-free survival (DFS) of 60% to 70% in selected acute leukemia patients receiving myeloablative single unit UCBT (sUCBT).6  Several European centers, with a more diverse population, have reported DFS of 40% to 50%.7,8  Both HLA match and cell dose are important for UCBT outcomes; patients who receive fully matched (6/6 HLA -A, -B, -DR) UCB units have superior survival, regardless of cell dose.9  An analysis of 514 patients treated with myeloablative sUCB found a 1-year survival of 37%, with older age, more advanced disease, and limited center experience predictive for worse survival.10 

No randomized trials have compared myeloablative UCB with other graft sources, but several large retrospective studies have been published (Table 1). Takahashi et al compared 100 UCB recipients with 71 recipients of matched related donor (MRD) transplant.11  There was no difference in transplant-related mortality (TRM), relapse, or leukemia-free survival (LFS). In 2004, Laughlin et al showed comparable survival between sUCBT and MMUD, but MUD patients did better.12  In 2010, Eapen et al, reporting for the Center for International Blood and Marrow Transplant Research (CIBMTR), compared outcomes of 165 recipients of myeloablative sUCBT with 888 MUD peripheral blood stem cell (PBSC) recipients and 472 MUD bone marrow (BM) recipients. TRM was higher for the UCBT cohort, but overall survival (OS) and DFS were comparable.13 

Table 1

Selected myeloablative transplant adult studies

ReferenceDonor sourceNTRM, %OS, %DFS, %
Aversa45  Haplo 104 40 41 48 AML in CR; 46 ALL in CR; 5 no CR 
Grosso47  Haplo 27 22 48 NR 
Huang49  Haplo 250 19-51 27-73 25-71 
Laughlin12  sUCB 150 63 26 23 
Laughlin12  7/8 MMUD/BM 83 70 20 19 
Eapen13  sUCB 165 37 NR 43 
Eapen13  MMUD 406 38 NR 44 
Brunstein15  dUCB 128 34 NR 51 
Brunstein15  MMUD 52 27 NR 38 
ReferenceDonor sourceNTRM, %OS, %DFS, %
Aversa45  Haplo 104 40 41 48 AML in CR; 46 ALL in CR; 5 no CR 
Grosso47  Haplo 27 22 48 NR 
Huang49  Haplo 250 19-51 27-73 25-71 
Laughlin12  sUCB 150 63 26 23 
Laughlin12  7/8 MMUD/BM 83 70 20 19 
Eapen13  sUCB 165 37 NR 43 
Eapen13  MMUD 406 38 NR 44 
Brunstein15  dUCB 128 34 NR 51 
Brunstein15  MMUD 52 27 NR 38 

CR indicates complete remission; ALL, acute lymphoblastic leukemia; and NR, not reported.

Myeloablative transplant

dUCBT has become more popular for adults in the United States partially because of the heavier weight of Americans (∼ 10 kg heavier than Europeans and 15 kg heavier than Asians). The Minnesota group pioneered this approach, reporting a 1-year DFS of 57% and a 13% incidence of acute GVHD grade 3 or 4.14  There are no randomized studies in adults comparing sUCBT with dUCBT; however, the BMT Clinical Trials Network (CTN) is investigating this question in children. The Minnesota and Seattle groups showed comparable LFS among 536 patients receiving dUCBT, MRD, MUD, or MMUD.15  TRM was higher, but relapse was lower, in the UCB recipients.

UCBT: reduced intensity transplant

The median age of patients with AML is 68 years; therefore, an RIC or nonmyeloablative approach is an attractive option. The Eurocord group analyzed 176 hematologic malignancy patients treated with a fludarabine-based RIC sUCBT.16  DFS was 41% at one year. Extending the dUCBT experience to RIC HCT, Brunstein reported a 38% 3-year DFS for patients treated with fludarabine, cyclophosphamide, and low-dose total body irradiation (TBI; Table 2).17  Our Massachusetts General Hospital (MGH)/Dana Farber Cancer Institute (DFCI) group, using an RIC regimen of fludarabine, melphalan, and rabbit antithymocyte globulin (ATG), reported a 1-year disease-free survival of 67%.18  Subsequent studies showed a very low rate (9%) of grades 2 to 4 GVHD using sirolimus and tacrolimus for GVHD prophylaxis.19  Chen et al showed comparable survival among MUD and dUCB in patients receiving an RIC HCT at DFCI/MGH.20  TRM was highest for the UCB patients, but the relapse rate was lower, suggesting a decreased risk of relapse without an increase in GVHD.

Table 2

Selected reduced intensity adult studies

ReferenceDonor sourceNTRM, %OS, %DFS, %
Rizzieri51  Haplo 49 31 31 43 
Luznik52  Haplo 68 15 36 26 
Brunstein21  Haplo 50 62 48 
Brunstein21  dUCB 50 24 54 46 
Brunstein17  dUCB 110 19 45 38 
Chen20  dUCB 64 26 46 30 
Ho35  MMUD 33 48 30 16 
Nakamae36  MMUD/MMRD 59 36 29 28 
Koreth41  MMUD 23 78 57 
ReferenceDonor sourceNTRM, %OS, %DFS, %
Rizzieri51  Haplo 49 31 31 43 
Luznik52  Haplo 68 15 36 26 
Brunstein21  Haplo 50 62 48 
Brunstein21  dUCB 50 24 54 46 
Brunstein17  dUCB 110 19 45 38 
Chen20  dUCB 64 26 46 30 
Ho35  MMUD 33 48 30 16 
Nakamae36  MMUD/MMRD 59 36 29 28 
Koreth41  MMUD 23 78 57 

MMRD indicates mismatched related donor.

Two parallel phase 2 trials studying RIC alternative donor HCT were completed by the United States BMT CTN.21  Fifty patients were treated in each study; all patients received an RIC regimen of fludarabine, cyclophosphamide, and low-dose TBI. Nonrelapse mortality (NRM) was higher after UCBT (24% for UCB vs 7% for haplo), but the relapse rate was higher after haplo-HCT (31% for UCB vs 45% for haplo). The 1-year DFS was comparable at 46% for UCBT and 48% for haplo-HCT. The CTN is currently planning a randomized study between haplo-HCT and UCBT. Thus, retrospective RIC studies have shown comparable survival among UCBT and haplo-SCT, and UCBT and MUD.

The future of cord blood

Several centers have attempted ex vivo expansion to improve engraftment and immune recovery. The MD Anderson Cancer Center is investigating expansion of one UCB unit with mesenchymal stem cells.22,23  Encouraging results have been reported by Delaney et al using a Notch ligand-based system.24  Direct intrabone marrow injection of sUCB may speed engraftment.25  Our DFCI/MGH group is studying dimeric prostaglandin E2 in cord blood homing.26  Preliminary results indicate expedited neutrophil engraftment and preferential engraftment with the treated cord.26 

Summary of UCBT

The advantages of UCBT are the readily availability of UCB units and the low risk of GVHD and relapse. Disadvantages include the cost and the slow immune recovery, which contributes to infections and TRM.

HLA-mismatched HCT is imprecisely defined in the literature but typically involves a 1 or 2 HLA-locus mismatch at class I (HLA-A, -B, -C) and/or class II (HLA-DRB1; ± -DQ, -DP). HLA-mismatched HCT is appealing for patients lacking HLA matches as donor availability is considerably enhanced. In the NMDP registry, 68% of black patients would have an available 7 of 8 HLA-matched donor, compared with 20% with an available 8 of 8 HLA-matched donor (NMDP oral communication, 2011). However, HLA-mismatched HCT has been associated with impaired outcomes after myeloablative and RIC HCT.

Myeloablative conditioning

The Japan Marrow Donor Program (JMDP) reported higher mortality with allele or antigen-level mismatch at HLA-A and/or -B, but not HLA-C or -DRB1, although its applicability to the more diverse non-Japanese populations remained in question.27,28  A large CIBMTR retrospective study compared HLA-mismatched BM versus 8 of 8 HLA-matched (HLA-A, -B, -C, -DRB1) unrelated donor HCT and documented worse NRM, acute GVHD, and overall and DFS with increasing degree of antigen and/or allele-level HLA-mismatch.29  The 1-year survival of 8 of 8, 7 of 8, and 6 of 8 HLA-matched cohorts was 52%, 43%, and 33%, respectively, in low-risk disease, an absolute unadjusted survival difference of 9% to 10% for each HLA-locus mismatch (P < .001).

French BM registry data reported that mismatch at HLA-A, -B, -C, -DRB1, or -DQB1 was associated with significant decrement in survival.30  Reports from the International Histocompatibility Working Group also identified impaired survival with HLA-DQB1 mismatch, whereas HLA-DPB1 mismatch was associated with increased acute GVHD (P < .001) but reduced relapse risk (P = .01).31,32  There was greater survival impairment of an HLA mismatch seen in low- versus intermediate- or high-risk disease (P < .0001, P = .02, and P = .43, respectively).31  Similar outcome impairment was reported for 1-antigen HLA-mismatched (HLA-A, -B, -C, -DRB1) PBSC transplantation.33  Retrospective analyses compared outcomes after myeloablative sUCB versus MUD and MMUD BM transplantation, suggesting that sUCB outcomes were comparable with MMUD, and in one of the 2 studies, equivalent to MUD.12,34 

RIC

The impact of HLA mismatch in RIC HCT is less well defined but is associated with high rates of acute GVHD, NRM, and impaired survival after 1 or 2 locus-mismatched RIC HCT (Table 2). In a retrospective report from the DFCI, HLA-C disparity compared with HLA-matched transplantation was associated with increased grade 3 or 4 acute GVHD (33% vs 12%, P = .01), increased NRM (48% vs 16%; P = .0001), and worse 2-year OS (30% vs 51%, P = .008).35  CIBMTR data confirmed impaired survival of HLA-C antigen mismatch compared with 8 of 8 HLA-matched RIC PBSC transplantation (relative risk = 1.40, 95% CI, 1.01-1.95, P = .04).32  A prospective study of T-replete 1 or 2 locus HLA-mismatched RIC PBSC grafts and calcineurin inhibitor-based GVHD prophylaxis described similarly poor outcomes, with rates of grades 2 to 4 acute GVHD of 69%, grade 3 or 4 acute GVHD of 26%, NRM of 47%, and 2-year OS of 29%.36 

The relative impact of HLA disparity at individual loci remains controversial. JMDP data indicated higher mortality with mismatch at HLA-A and/or -B, but non-Japanese registries have reported variant results: some confirming mismatches at HLA-B or -C as better tolerated than those at HLA-A or -DRB1, but others reporting worse survival for HLA-C but not HLA-A mismatches.27,28,31,33  In addition, individual reports of the JMDP and Italian Bone Marrow Donor Registry have identified specific “high-risk” nonpermissive allele mismatches at HLA-A, -B, -C, -DR, and -DPB1 that individually increase acute GVHD risk and NRM and impair survival.37,38  The data suggest that antigen or allele-level HLA-mismatch probably has a similar deleterious impact. Although HLA-A, -B, -C, and -DRB1 are considered relevant loci, there is less certainty about HLA-DPB1 or -DQB1. Although there is a possibility of enhanced graft-versus-tumor effect with increasing HLA disparity, as suggested by the finding that MMUD outcomes may not be as impaired in higher risk disease, this finding has not been prospectively evaluated.

The future of mismatched unrelated transplantation

In the myeloablative setting, the use of a proprietary ATG (Fresenius) has been shown to reduce acute and chronic GVHD risk without impairing NRM or survival.39  In the RIC setting, however, critically dependent on immunologic graft-versus-tumor effect for cure, in vivo T-cell-depletion (eg, ATG, alemtuzumab) appears to impair disease-free survival.40  Novel T-replete RIC regimens are currently being studied in Boston, where the addition of bortezomib to a calcineurin inhibitor-based regimen has been prospectively tested in 45 patients undergoing MMUD HCT with GVHD, NRM, and survival outcomes in the range reported for HLA-matched donors.41,42 

Summary of mismatched unrelated donor transplant

The advantages of mismatched unrelated donor transplant include more available grafts, particularly for minority patients, and in some studies, a lower risk of relapse. Disadvantages include a high risk of GVHD.

Haplo-HCT is a favorable option for several reasons, including the ready availability of a related donor for HCT and for subsequent adoptive cellular immunotherapy, lack of search or cord blood banking fees, and the increased donor availability as many patients will have a parent or child that could serve as a haplo-donor. Haplo-HCT has been limited by historically high rates of graft rejection, GVHD, TRM, and poor immune reconstitution, resulting in a high incidence of serious opportunistic infection. Both myeloablative and RIC transplant strategies have been attempted, and selected experiences are summarized in Tables 1 and 2.

Myeloablative conditioning

Early attempts with myeloablative haploidentical HCT and pharmacologic GVHD prophylaxis with methotrexate (with or without cyclosporine) were complicated by high rates of graft rejection, hyperacute GVHD, and TRM.43  HLA-one antigen-mismatched HCT outcomes were not significantly different from those of HLA-matched related donor transplantation (higher incidences of GVHD but similar OS), but HLA-2/HLA-3 antigen-mismatched transplants were associated with prohibitive mortality risks.44  More recent approaches using myeloablative conditioning and vigorous ex vivo T-cell depletion have resulted in a very low incidence of acute and chronic GVHD and favorable event-free and OS probabilities for patients with acute leukemia. Aversa et al described their experience with TBI, thiotepa, fludarabine, and ATG conditioning and ex vivo T-cell depleted “megadose” peripheral blood stem cell transplantation.45  Among 104 patients with AML (N = 67) or acute lymphoblastic leukemia (ALL; N = 37), engraftment was achieved in 100 of 101 evaluable patients. Acute and chronic GVHD occurred in 8 and 5 of 70 evaluable patients, respectively. NRM probability was 38% primarily resulting from opportunistic infections.

Another myeloablative haplo-HCT strategy has involved the ex vivo induction of alloantigen specific anergy by the coculturing of host and donor BM mononuclear cells with either CTLA-4-IG or anti–B-7.1 and B7.2 antibodies. Davies et al conducted sequential trials of haplo-HCT using this ex vivo anergization strategy in 24 patients with advanced hematologic disorders.46  Grade C or D acute GVHD and chronic GVHD were seen in 5 and 1 patients, respectively. Eight of the 24 patients were disease-free at a median of 7 years after transplantation. Grosso et al47  recently described an approach involving in vivo T-cell tolerization using TBI, then donor peripheral blood T cells followed by cyclophosphamide. Patients then received ex vivo CD34-selected peripheral blood stem cells. The probabilities of grades 2 to 4 acute and chronic GVHD were 59% and 16%, respectively. NRM was 23%. OS probability was 48% at 3 years.47 

Myeloablative haplo-HCT approaches without ex vivo T-cell depletion have also been recently described. Multiple investigators have described a strategy using aggressive multiagent conditioning regimens, polyclonal ATG for in vivo T-cell depletion, and posttransplant GVHD pharmacoprophylaxis.48–50  Huang et al recently described 250 patients who received conditioning with busulfan, cytarabine, cyclophosphamide, semimustine, and ATG, and post-HCT GVHD prophylaxis with mycophenolate mofetil, cyclosporine, and methotrexate.49  A total of 249 of the 250 patients achieved full donor chimerism. Grades 2 to 4 acute and chronic GVHD incidences of 46% and 54%, respectively, were reported. Three-year LFS probabilities were excellent, especially for patients with standard-risk disease: for AML, LFS probabilities were 71% and 56% for standard- and high-risk patients, respectively; and for ALL, 60% and 25% for standard- and high-risk patients, respectively.

RIC

RIC haplo-HCT approaches have also been evaluated. Rizzieri et al administered a conditioning regimen of fludarabine, cyclophosphamide, and alemtuzumab for in vivo T-cell deletion.51  Posttransplant GVHD prophylaxis consisted of mycophenolate mofetil with or without cyclosporine. The incidence of GVHD was low with this approach, as 16% of patients developed grades 2 to 4 acute GVHD and 8% chronic GVHD. One-year relapse-free and OS probabilities were 43% and 31%, respectively. Posttransplant high-dose cyclophosphamide was used by Luznik et al to deplete alloreactive (both in the GVH and HVG direction) T cells after conditioning with low-dose TBI, fludarabine, and cyclophosphamide for a variety of hematologic malignancies.52  A 13% rate of graft failure was observed. Grades 2 to 4 acute GVHD developed in 34% of patients, and chronic GVHD developed in 25% and 5% of patients who received one or 2 doses of posttransplant cyclophosphamide, respectively. Two-year event-free survival and OS probabilities were 26% and 36%. At MGH, sequential trials initially used cyclophosphamide, equine ATG, and thymic radiation, with the later substitution of a monoclonal anti CD2 antibody (MEDI-507) for ATG for better T-cell depletion, with the intent to induce stable mixed chimerism as a platform for delayed donor lymphocyte infusion. Graft rejection and GVHD were limiting complications.53,54  Using a similar conditioning strategy at MGH with both ex vivo T-cell depletion (by CD 34+ cell selection) and in vivo T-cell depletion (using MEDI 507), we found that stable mixed chimerism was reliably achieved with minimal GVHD, and long-term survival has been achieved in approximately one-fourth of patients with chemorefractory lymphoma (B.D., K.B., S. McAfee, T.S., unpublished data, December 2011). Other fludarabine-and ATG-based RIC regimens for haplo-HCT have been reported with similar, favorable survival outcomes, particularly for standard-risk hematologic malignancies.55,56  As described above, the CTN phase 2 studies showed comparable survival for UCBT and haplo-HCT; haplo-compared with MMUD HCT has not been well studied.

Future of haploidentical transplantation

Because relapse remains a major limitation of haplo-HCT, several strategies are being developed to reduce the risk of relapse. Transfer of tumor-specific T lymphocytes or donor-derived NK cells is under investigation.57,58  Novel approaches to the prevention and treatment of infection include the use of donor-derived virus-specific T lymphocytes to treat refractory viral and fungal infections.59 

Summary of haploidentical transplantation

Haploidentical donor options are available for most patients, and there are no search or acquisition costs. Disadvantages of haploidentical HCT are the necessary technical expertise and cost of ex vivo T-cell depletion (when such methods are used), poor immune reconstitution, and high risk of relapse (after RIC approaches) after transplantation.

The 3 alternative donor sources (UCB, haplo-HCT, and MMUD) have different posttransplantation complications, as illustrated in Table 3. UCB is associated with a higher risk of infection but often a lower risk of GVHD than haplo-HCT or MMUD. Second malignancies have been reported more frequently after UCB or haplo-HCT. Cost may be higher with UCB. These sections explore the risks of the common after transplantation complications with the 3 donor sources.

Table 3

Advantages and disadvantages of alternative donor sources

UCBHaploidentical-related family memberOne locus mismatched unrelated donor
Donor availability Difficulty to find high-quality units for minorities Most patients have parent or child as donor May be lengthy/difficult to find donors, especially for minorities 
Cost US $20 000-$40 000 per cord unit Low donor acquisition costs US $20 000-25 000 
Availability of donor lymphocytes No Yes, readily available Available, but may be lengthy wait 
GVHD Low risk May be severe, especially with no T-cell depletion May be very severe 
Infection High risk, especially viral High risk Moderate risk 
Product quality High variability Low variability Low variability 
Relapse risk Moderate High, especially with some forms of T-cell depletion Moderate 
UCBHaploidentical-related family memberOne locus mismatched unrelated donor
Donor availability Difficulty to find high-quality units for minorities Most patients have parent or child as donor May be lengthy/difficult to find donors, especially for minorities 
Cost US $20 000-$40 000 per cord unit Low donor acquisition costs US $20 000-25 000 
Availability of donor lymphocytes No Yes, readily available Available, but may be lengthy wait 
GVHD Low risk May be severe, especially with no T-cell depletion May be very severe 
Infection High risk, especially viral High risk Moderate risk 
Product quality High variability Low variability Low variability 
Relapse risk Moderate High, especially with some forms of T-cell depletion Moderate 

GVHD

Acute and chronic GVHD remain important sources of morbidity and mortality after HCT and, thus, should factor heavily when faced with the choice of an alternative stem cell donor. Significant advances have been made with improved donor selection and the use of novel prophylaxis regimens for different alternative donors. Several of these studies are outlined in Table 4.

Table 4

Risk of GVHD and relapse

ReferencenDiseasesConditioningGVHD ppxRelapse, %II-IV Acute GVHD, %Chronic GVHD, %
UCB 
    Eapen13  165 AML, ALL Various myeloablative Various 30 30 24 
    Brunstein15  128 AML, ALL, CML, MDS Flu/Cy/TBI-ablative CNI + MMF 15 60 26 
    Chen20  64 Various Flu/Mel/ATG-RIC Various 43 14 22 
Haplo 
    Aversa45  104 AML, ALL TBI, TT, Flu, ATG-myeloablative TCD 25 
    Rizzieri51  49 Various Flu/Cy/alemtuzumab-RIC CsA ± MMF NA 16 14 
    Luznik52  68 Various Flu/Cy/TBI-RIC Cy/Tacro/MMF 58 34 13 
MMUD 
    Pidala61  45 Various Various ATG/Tacro/MTX 33 64 35 
    Kroger93  158 Various Various F-ATG/CsA/MTX 27 41 41 
    Mead63  50 Various Flu/Mel/alemtuzumab CsA NA 22 39 
ReferencenDiseasesConditioningGVHD ppxRelapse, %II-IV Acute GVHD, %Chronic GVHD, %
UCB 
    Eapen13  165 AML, ALL Various myeloablative Various 30 30 24 
    Brunstein15  128 AML, ALL, CML, MDS Flu/Cy/TBI-ablative CNI + MMF 15 60 26 
    Chen20  64 Various Flu/Mel/ATG-RIC Various 43 14 22 
Haplo 
    Aversa45  104 AML, ALL TBI, TT, Flu, ATG-myeloablative TCD 25 
    Rizzieri51  49 Various Flu/Cy/alemtuzumab-RIC CsA ± MMF NA 16 14 
    Luznik52  68 Various Flu/Cy/TBI-RIC Cy/Tacro/MMF 58 34 13 
MMUD 
    Pidala61  45 Various Various ATG/Tacro/MTX 33 64 35 
    Kroger93  158 Various Various F-ATG/CsA/MTX 27 41 41 
    Mead63  50 Various Flu/Mel/alemtuzumab CsA NA 22 39 

ALL indicates acute lymphoblastic leukemia; CML, chronic myeloid leukemia; MDS, myelodysplastic syndrome; Flu, fludarabine; Cy, cyclophosphamide; Mel, melphalan; TT, thiotepa; CNI, calcineurin inhibitor; MMF, mycophenolate mofetil; TCD, T-cell depletion; CsA, cyclosporine A; Tacro, tacrolimus; MTX, methotrexate; F-ATG, Fresenius ATG; and NA, not available.

The ability to cross significant HLA barriers safely has made UCBT quite attractive. Currently, standard UCB selection criteria require matching at 4 of 6 major HLA-antigens (A, B, DRB1) with high resolution typing only routinely required for HLA-DRB1.60  Even with such a degree of HLA disparity, rates of acute and chronic GVHD after UCBT appear to be either similar or decreased compared with HCT from conventional or alternative stem cell sources. The Eapen CIBMTR study, which studied 1525 acute leukemia patients receiving myeloablative HCT, reported a significantly lower rate of both acute and chronic GVHD for sUCBT compared with unrelated PBSC recipients, and significantly decreased rates of chronic GVHD compared with PBSC or BM.13  Brunstein's myeloablative study of 536 leukemia and MDS patients found similar rates of grades 2 to 4 acute GVHD for dUCBT relative to MRD HCT, but significantly lower GVHD than MUD or MMUD patients, 26% for dUCBT, and 43% to 48% for MUD and MMUD.15 

In the reduced intensity setting, our Boston group compared RIC dUCBT (n = 64) with RIC MUD HCT (n = 221). Similar rates of grades 2 to 4 acute GVHD were observed in both groups, but patients undergoing dUCBT had a significantly lower incidence of chronic GVHD (54% MUD vs 22% dUCB, P < .0001).20  The Johns Hopkins group, using posttransplantation cyclophosphamide to achieve selective depletion of alloreactive donor T cells after haplo-RIC, reported a 34% incidence of grades 2 to 4 acute GVHD (6% grade 3 or 4) and a very low incidence of chronic GVHD.52  Recent results from the CTN phase 2 trial using this approach in 50 patients at 17 centers showed an impressively low 22% incidence of grades 2 to 4 acute GVHD (0% grade 3 or 4) and a 13% cumulative incidence of chronic GVHD at one year.21 

Several groups have reported their experience adding ATG to standard regimens for MMUD HCT. Pidala et al reported on 45 patients undergoing MMUD HCT, all of whom received ATG (thymoglobulin, Genzyme), tacrolimus, and methotrexate and showed a 1% incidence of grade 3 or 4 acute GVHD and a 19% incidence of moderate to severe chronic GVHD.61  Kim et al reported a retrospective analysis of 49 MMUD patients and showed that patients who received ATG in addition to standard tacrolimus and methotrexate had a lower incidence of grades 2 to 4 acute GVHD (8% vs 29%, P = .038) with similar incidences of chronic GVHD.62  Mead et al presented results using pre-HCT alemtuzumab and post-HCT cyclosporine in 157 RIC patients, 50 of whom were mismatched at 1 to 4 HLA antigens (64% single loci mismatch, 30% 2 loci mismatch). Their results showed similar outcomes between MUD and MMUD HCT with no evidence of any increased risk of acute or chronic GVHD.63  These studies suggest that rates of acute and chronic GVHD after MMUD HCT can be comparable to MUD and perhaps UCB if additional agents, such as alemtuzumab and ATG, are used; however, large prospective studies are needed to validate the role of ATG in MMUD HCT.

Relapse

Although significant strides have been made in reducing overall transplant-related mortality in HCT over the past 2 decades, relapse of disease remains the most common cause of failure in patients undergoing HCT.64  With lower rates of GVHD after UCBT, an initial concern was that increased rates of relapse would be observed because of a weaker graft-versus-malignancy effect. Interestingly, this has not been borne out in the several retrospective comparisons of UCBT to HCT from adult stem cell sources. In the large CIBMTR study, the cumulative incidence of disease relapse was similar despite a significantly lower rate of chronic GVHD in the UCB group.13  Several analyses have shown a decreased risk of relapse with dUCBT compared with sUCBT.65,66  In the analysis by Brunstein et al, recipients of myeloablative dUCBT had significantly lower rates of disease relapse at 5 years (15%) compared with recipients of MRD (43%), MUD (37%), and MMUD (35%).15  Donor lymphocyte infusions, however, are not available for UCBT recipients who relapse. No analysis has suggested that disease relapse is decreased in patients who are recipients of MMUD grafts relative to matched counterparts. Indeed, a large CIBMTR registry study of more than 4500 chronic myelogenous leukemia patients did not find a lower risk of relapse with greater HLA mismatching.67  Relapse rates after haplo-HCT appear to differ significantly depending on the conditioning and GVHD regimens used. More studies are needed to define whether certain disease groups benefit selectively from mismatching at specific HLA loci with adult donor stem cell sources.

In the RIC setting, analyses have shown similar rates of relapse among dUCB and conventional HCT.20,68  Preliminary analyses of RIC haplo-HCT with the Hopkins regimen suggested that patients with lymphoid diseases had less relapse than patients with myeloid diseases with especially impressive outcomes for patients with Hodgkin lymphoma.69  The presence of an HLA-DRB1 antigen mismatch in the GVH direction or the presence of 2 or more class I allele mismatches (composite of HLA-A, -B, and -C) in either direction was associated with significantly lower rates of disease relapse and improved event-free survival without excess NRM.70  Nevertheless, the relapse rate of 45% at 1-year after RIC haplo-HCT observed in the BMT CTN study raises concern that the successful prevention of GVHD in this manner may inhibit effective graft-versus-malignancy effects.21  Disease relapse is dependent on the underlying disease, disease risk, and status at the time of HCT and, thus, best determined in a prospective randomized study. Finally, the precise mechanisms of tumor immunity after HCT, which may be different depending on donor type, need to be elucidated to potentially better choose among donor types based on underlying disease.

Immune reconstitution and infectious complications

Despite advances in antimicrobial therapy, severe infections remain a major cause of death after alternative donor HCT, particularly in older patients. The Spanish group compared 48 recipients of single UCBT with 144 recipients of unrelated BM or PBSC HCT.71  At 3 years, the UCBT group had a higher risk of developing an infection, but infection-related mortality (25%) was similar in the 2 groups. HLA mismatch did not affect outcome in the UCBT group but was associated with a higher mortality in the BMT/PBSC cohort. Ringden et al found a similar rate of bacteremia in UCB and MMUD patients, but TRM was higher for the MMUD patients.72  The Minnesota group has demonstrated comparable rates of cytomegalovirus infection between dUCBT and MRD transplantation.73 

Cord blood contains fewer T cells than other stem cell sources, and cord blood lymphocytes have specific immunologic characteristics, such as different response pattern to cytokines and a greater proportion of naive T cells.10  In haplo-recipients, there is more NK cell alloreactivity, and NK cell infusions have been used therapeutically after transplantation.74  In a prospective analysis of immune reconstitution in dUCBT recipients and MUD recipients from the DFCI, Jacobson et al found that CD3 recovery was significantly delayed in the dUCBT group compared with the MUD group for as long as 6 months after HCT, including naive (CD45RO) and memory (CD45RO+) CD4 T cells, regulatory (CD4CD25) T cells, and CD8 T cells.75  These unique properties of UCB may contribute to a high risk of infection reported in some studies. Novel strategies to combat infection include the use of virus-specific or trivirus-specific (adenovirus, Epstein-Barr virus, and cytomegalovirus) cytotoxic T lymphocytes.76  Donor-derived cytotoxic T lymphocytes can be used to combat viral infections in haplo-recipients.59 

Posttransplant lymphoma and other second malignancies

Second malignancies remain a devastating complication after HCT. A total of 1% of patients in a CIBMTR study of 18 000 BM patients developed posttransplantation lymphoproliferative disorder, with risk factors, including an HLA-mismatched donor and T-cell depletion.77  UCBT, which are “naturally” T cell depleted have a high incidence of posttransplantation lymphoproliferative disorder, especially with the use of ATG in an RIC regimen.78,79  Frequent monitoring for EBV and preemptive use of rituximab may reduce the risk and severity of posttransplantation lymphoproliferative disorder after UCBT and MMUD HCT.80,81  Novel approaches to posttransplantation lymphoproliferative disorder include the use of EBV or trivirus-specific cytotoxic T lymphocytes.82,83  Donor-derived second myeloid malignancies may be increased after UCBT, perhaps because of naive cells or the use of growth factors.79,84,85 

Cost and length of stay

Given the current healthcare climate, the cost for the alternative donor graft sources assumes new importance. Costs vary widely from center to center, so the number of hospital days may be a surrogate marker. The Minnesota group compared costs in the first 100 days after myeloablative and RIC transplantation using either UCB or MRD grafts.86  The median cost per day survived (not including graft acquisition) was $1016 for myeloablative MRD, $2082 for myeloablative UCB, $612 for RIC MRD, and $1156 for RIC UCB. Acquisition of 2 UCB units can cost up to $80 000 prior to the patient entering the hospital; these costs may be related to the large inventory of UCB units, of which only 10% have been used for HCT. Acquisition costs for haplo-HCT are clearly less. The CIBMTR is embarking on a retrospective comparison of length of stay among the different graft sources.

Combination of donor sources

Given the high risk of infection after UCBT, an intriguing strategy is to combine donor sources using both haplo-BM and UCB.87  The advantage of this approach is the rapid engraftment of the haplo cells to reduce the risk of early infections, followed by sustained hematopoietic engraftment of the UCB.88  Five-year DFS of 47% has been reported with this strategy.89 

In conclusion, patients with hematologic malignancies have multiple options for treatment, and multiple choices of transplant donor sources. Table 5 outlines the benefits and risks of each graft source. The field may evolve over the next 5 years so that specific patients may benefit from a particular approach; for example, patients at high risk of infection might receive an MMUD over UCBT, and patients at high risk of relapse might receive UCBT over haplo-SCT. Furthermore, specific diseases might respond differently to different donor sources. Because there are no prospective randomized comparative studies to date, we cannot promote a definitive strategy for donor selection, and clinical trial participation is encouraged. Our center performs UCBT, haplo-HCT, and MMUD HCT for patients without an 8 of 8 or 7 of 8 MRD or an 8 of 8 MUD. We use donor availability, protocol eligibility, patient age, disease, and disease status to decide on the best option for each individual patient. We do tend to favor UCBT given our research interests in this field. The field of alternative donor transplantation is moving forward as work on optimal UCB unit selection and investigation of the role of HLA antibodies, HLA-C, and KIR matching may improve results.90–92  In the haplo-HCT setting, the use of adoptive immunotherapy to decrease relapse is in progress. MMUD donor selection, with more sensitive HLA typing, will probably improve.

Table 5

Benefits and risks of each graft source

UCBMismatched unrelated donorHaploidentical transplant
Benefit Low relapse Low relapse for high-risk patients Ready availability 
Benefit Low severe GVHD Donor lymphocyte available Low cost 
Risk Cost GVHD Relapse (especially RIC) 
Risk Infection TRM Infection 
UCBMismatched unrelated donorHaploidentical transplant
Benefit Low relapse Low relapse for high-risk patients Ready availability 
Benefit Low severe GVHD Donor lymphocyte available Low cost 
Risk Cost GVHD Relapse (especially RIC) 
Risk Infection TRM Infection 

The CTN is embarking on a randomized RIC phase 3 study comparing haplo-HCT and UCB HCT. This study will answer important questions regarding the risks and benefits of these 2 donor sources. Finally, whereas this review focused on alternative donor sources, future randomized studies will be required to compare UCB, haplo, and MMUD to more conventional related and matched unrelated donor sources. We are confident that these studies will allow us to make more informed donor choices for our patients in the future.

The authors thank Drs Joseph Antin, Corey Cutler, and Robert Soiffer for their critical review of the manuscript.

Contribution: K.K.B. prepared the manuscript for publication; and all authors wrote and reviewed the manuscript.

Conflict-of-interest disclosure: J.K. acquired research funding and is on the Advisory Board of Millennium. The remaining authors declare no competing financial interests.

Correspondence: Karen Ballen, Massachusetts General Hospital, Division of Hematology/Oncology, Zero Emerson, Suite 118, Boston, MA 02114; e-mail: kballen@partners.org.

1
Ballen
 
KK
King
 
RJ
Chitphakdithai
 
P
, et al. 
The National Marrow Donor Program Experience: the first 20 years of unrelated donor stem cell transplantation.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
9 suppl
(pg. 
2
-
7
)
2
Ballen
 
KK
Spitzer
 
TR
The great debate: haploidentical or cord blood transplant.
Bone Marrow Transplant
2011
, vol. 
46
 
3
(pg. 
323
-
329
)
3
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
, vol. 
321
 
17
(pg. 
1174
-
1178
)
4
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
, vol. 
335
 
3
(pg. 
157
-
166
)
5
Barker
 
JN
Cyam
 
CE
Kernan
 
NA
, et al. 
Availability of cord blood extends allogeneic hematopoietic stem cell transplant access to racial and ethnic minorities.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
11
(pg. 
1541
-
1548
)
6
Ooi
 
J
Takahashi
 
S
Tomonari
 
A
, et al. 
Unrelated cord blood transplantation after myeloablative conditioning in adults with acute myelogenous leukemia.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
12
(pg. 
1341
-
1347
)
7
Sanz
 
J
Sanz
 
MA
Saavedra
 
S
, et al. 
Cord blood transplantation from unrelated donors in adults with high-risk acute myeloid leukemia.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
1
(pg. 
86
-
94
)
8
Rocha
 
V
Gluckman
 
E
Improving outcomes of cord blood transplantation: HLA matching, cell dose, and other graft-and transplantation-related factors.
Br J Haematol
2009
, vol. 
147
 
2
(pg. 
262
-
274
)
9
Barker
 
JN
Scaradavou
 
A
Stevens
 
CE
Combined total effect of total nucleated cell dose and HLA-match on transplant outcomes in 1061 cord blood recipients with hematologic malignancies.
Blood
2010
, vol. 
115
 
9
(pg. 
1843
-
1849
)
10
Cohen
 
YC
Scaradavou
 
A
Stevens
 
CE
, et al. 
Factors affecting mortality following myeloablative cord blood transplantation in adults: a pooled analysis of three international registries.
Bone Marrow Transplant
2011
, vol. 
46
 
1
(pg. 
70
-
76
)
11
Takahashi
 
S
Ooi
 
J
Tomanari
 
A
, et al. 
Comparative single-institute analysis of cord blood transplantation from related donors with bone marrow or peripheral blood stem cell transplants from related donors in adult patients with hematologic malignancies and myeloablative conditioning regimen.
Blood
2007
, vol. 
109
 
3
(pg. 
1322
-
1330
)
12
Laughlin
 
MJ
Eapen
 
M
Rubinstein
 
P
, et al. 
Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia.
N Engl J Med
2004
, vol. 
351
 
22
(pg. 
2265
-
2275
)
13
Eapen
 
M
Rocha
 
V
Sanz
 
G
, et al. 
Effect of graft source of unrelated donor hematopoietic stem-cell transplantation in adults with acute leukemia: a retrospective analysis.
Lancet
2010
, vol. 
11
 
7
(pg. 
653
-
660
)
14
Barker
 
JN
Weisdorf
 
DF
DeFor
 
TE
, et al. 
Transplantation of 2 partially HLA-matched UCB units to enhance engraftment in adults with hematologic malignancy.
Blood
2005
, vol. 
105
 
3
(pg. 
1343
-
1347
)
15
Brunstein
 
CG
Gutman
 
JA
Weisdorf
 
DJ
, et al. 
Allogeneic hematopoietic cell transplantation for hematologic malignancy: relative risks and benefits of double umbilical cord blood.
Blood
2010
, vol. 
116
 
22
(pg. 
4693
-
4699
)
16
Rocha
 
V
Mohty
 
M
Gluckman
 
E
Rio
 
B
Reduced-intensity conditioning regimens before unrelated cord blood transplantation in adults with acute leukemia and other hematologic malignancies.
Curr Opin Oncol
2009
, vol. 
21
 
suppl 1
(pg. 
S31
-
S34
)
17
Brunstein
 
CG
Barker
 
JN
Weisdorf
 
DJ
, et al. 
Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease.
Blood
2007
, vol. 
110
 
8
(pg. 
3064
-
3070
)
18
Ballen
 
KK
Spitzer
 
TR
Yeap
 
BY
, et al. 
Double unrelated reduced-intensity umbilical cord blood transplantation in adults.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
1
(pg. 
82
-
89
)
19
Cutler
 
C
Stevenson
 
K
Kim
 
HT
, et al. 
Double umbilical cord blood transplantation with reduced intensity conditioning and sirolimus based GVHD prophylaxis.
Bone Marrow Transplant
2011
, vol. 
46
 
5
(pg. 
659
-
667
)
20
Chen
 
YB
Aldridge
 
J
Kim
 
HT
, et al. 
Reduced-intensity conditioning stem cell transplantation: comparison of double unbilical cord blood and unrelated donor grafts [published online ahead of print October 19, 2011].
Biol Blood Marrow Transplant
 
21
Brunstein
 
CG
Fuchs
 
EJ
Carter
 
SJ
, et al. 
Alternative donor transplantation after reduced intensity conditioning: results of parallel phase 2 trials using partially HLA-mismatched related bone marrow or unrelated double umbilical cord blood grafts.
Blood
2011
, vol. 
118
 
2
(pg. 
282
-
288
)
22
Robinson
 
SN
Simmons
 
PJ
Yang
 
H
Alousi
 
AM
deLima
 
M
Sphall
 
EJ
Mesenchymal stem cells in ex vivo cord blood expansion.
Best Pract Res Clin Hematol
2011
, vol. 
24
 
1
(pg. 
83
-
92
)
23
Shpall
 
EJ
Bollard
 
CM
Brunstein
 
C
, et al. 
Novel cord blood transplant therapies.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
1 suppl
(pg. 
S39
-
S45
)
24
Delaney
 
C
Heimfeld
 
S
Brashem-Stein
 
C
, et al. 
Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution.
Nat Med
2010
, vol. 
16
 
2
(pg. 
232
-
236
)
25
Frassoni
 
F
Gualandi
 
F
Podesta
 
M
, et al. 
Direct intrabone transplant of unrelated cord-blood cells in acute leukemia: a phase I/II study.
Lancet Oncol
2008
, vol. 
9
 
9
(pg. 
831
-
839
)
26
Cutler
 
CS
Desponts
 
C
Robbins
 
D
, et al. 
Ex vivo treatment of hematopoietic stem cells with 16,16 dimethyl prostaglandin E2 (FT 1050) improves engraftment and immune reconstitution.
Biol Blood Marrow Transplant
2011
, vol. 
17
 pg. 
198a
 
27
Sasazuki
 
T
Juji
 
T
Morishima
 
Y
, et al. 
Effect of matching of class I HLA alleles on clinical outcome after transplantation of hematopoietic stem cells from an unrelated donor: Japan Marrow Donor Program.
N Engl J Med
1998
, vol. 
339
 
17
(pg. 
1177
-
1185
)
28
Morishima
 
Y
Sasazuki
 
T
Inoko
 
H
, et al. 
The clinical significance of human leukocyte antigen (HLA) allele compatibility in patients receiving a marrow transplant from serologically HLA-A, HLA-B, and HLA-DR matched unrelated donors.
Blood
2002
, vol. 
99
 
11
(pg. 
4200
-
4206
)
29
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
)
30
Loiseau
 
P
Busson
 
M
Balere
 
ML
, et al. 
HLA association with hematopoietic stem cell transplantation outcome: the number of mismatches at HLA-A, -B, -C, -DRB1, or -DQB1 is strongly associated with overall survival.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
8
(pg. 
965
-
974
)
31
Petersdorf
 
EW
Gooley
 
T
Malkki
 
M
Horowitz
 
M
Clinical significance of donor-recipient HLA matching on survival after myeloablative hematopoietic cell transplantation from unrelated donors.
Tissue Antigens
2007
, vol. 
69
 
suppl 1
(pg. 
25
-
30
)
32
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
)
33
Woolfrey
 
A
Klein
 
JP
Haagenson
 
M
, et al. 
HLA-C antigen mismatch is associated with worse outcome in unrelated donor peripheral blood stem cell transplantation.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
6
(pg. 
885
-
892
)
34
Rocha
 
V
Labopin
 
M
Sanz
 
G
, et al. 
Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia.
N Engl J Med
2004
, vol. 
351
 
22
(pg. 
2276
-
2285
)
35
Ho
 
VT
Kim
 
HT
Liney
 
D
, et al. 
HLA-C mismatch is associated with inferior survival after unrelated donor non-myeloablative hematopoietic stem cell transplantation.
Bone Marrow Transplant
2006
, vol. 
37
 
9
(pg. 
845
-
850
)
36
Nakamae
 
H
Storer
 
BE
Storb
 
R
, et al. 
Low-dose total body irradiation and fludarabine conditioning for HLA class I-mismatched donor stem cell transplantation and immunologic recovery in patients with hematologic malignancies: a multicenter trial.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
3
(pg. 
384
-
394
)
37
Kawase
 
T
Morishima
 
Y
Matsuo
 
K
, et al. 
High-risk HLA allele mismatch combinations responsible for severe acute graft-versus-host disease and implication for its molecular mechanism.
Blood
2007
, vol. 
110
 
7
(pg. 
2235
-
2241
)
38
Crocchiolo
 
R
Zino
 
E
Vago
 
L
, et al. 
Nonpermissive HLA-DPB1 disparity is a significant independent risk factor for mortality after unrelated hematopoietic stem cell transplantation.
Blood
2009
, vol. 
114
 
7
(pg. 
1437
-
1444
)
39
Finke
 
J
Bethge
 
WA
Schmoor
 
C
, et al. 
Standard graft-versus-host disease prophylaxis with or without anti-T-cell globulin in haematopoietic cell transplantation from matched unrelated donors: a randomized, open-label, multicentre phase 3 trial.
Lancet Oncol
2009
, vol. 
10
 
9
(pg. 
855
-
864
)
40
Soiffer
 
RJ
Lerademacher
 
J
Ho
 
V
, et al. 
Impact of immune modulation with anti-T-cell antibodies on the outcome of reduced-intensity allogeneic hematopoietic stem cell transplantation for hematologic malignancies.
Blood
2011
, vol. 
117
 
25
(pg. 
6963
-
6970
)
41
Koreth
 
J
Stevenson
 
KE
Kim
 
HT
, et al. 
Bortezomib, tacrolimus, and methotrexate for prophylaxis of graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation from HLA-mismatched unrelated donors.
Blood
2009
, vol. 
114
 
18
(pg. 
3956
-
3959
)
42
Koreth
 
J
Stevenson
 
EK
Kim
 
HT
, et al. 
A prospective phase I/II trial of bortezomib-based graft-versus-host-disease prophylaxis in HLA-mismatched unrelated donor reduced-intensity conditioning hematopoietic stem cell transplantation: encouraging safety, efficacy, and survival [abstract].
Biol Blood Marrow Transplant
2011
, vol. 
17
 
2
(pg. 
S165
-
S166
)
43
Powles
 
RL
Morgenstern
 
GR
Kay
 
HE
, et al. 
Mismatched family donors for bone-marrow transplantation as treatment for acute leukaemia.
Lancet
1983
, vol. 
1
 
8325
(pg. 
612
-
615
)
44
Beatty
 
PG
Clift
 
RA
Mickelson
 
EM
, et al. 
Marrow transplantation from related donors other than HLA-identical siblings.
N Engl J Med
1985
, vol. 
313
 
13
(pg. 
765
-
771
)
45
Aversa
 
F
Terenzi
 
A
Tabilio
 
A
, et al. 
Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse.
J Clin Oncol
2005
, vol. 
23
 
15
(pg. 
3447
-
3454
)
46
Davies
 
JK
Gribben
 
JG
Brennan
 
LL
Yuk
 
D
Nadler
 
LM
Guinan
 
EC
Outcome of alloanergized haploidentical bone marrow transplantation after ex vivo costimulatory blockade: results of 2 phase 1 studies.
Blood
2008
, vol. 
112
 
6
(pg. 
2232
-
2241
)
47
Grosso
 
D
Carabasi
 
M
Filicko-O'Hara
 
, et al. 
A 2-step approach to myeloablative haploidentical stem cell transplantation: a phase 1/2 trial performed with optimized T-cell dosing.
Blood
2011
, vol. 
118
 
17
(pg. 
4732
-
4739
)
48
Ogawa
 
H
Ikegame
 
K
Yoshihara
 
S
, et al. 
Unmanipulated HLA 2-3 antigen mismatched (haploidentical) stem cell transplantation using nonmyeloablative conditioning.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
10
(pg. 
1073
-
1084
)
49
Huang
 
X-J
Liu
 
D-H
Liu
 
K-Y
Treatment of acute leukemia with unmanipulated HLA-mismatched/haploidentical blood and bone marrow transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
2
(pg. 
257
-
265
)
50
Wang
 
HX
Yan
 
HM
Wang
 
ZD
, et al. 
Haploidentical hematopoietic stem cell transplantation in hematologic malignancies with G-CSF mobilized bone marrow plus peripheral blood stem cell grafts without T cell depletion: a single center report of 29 cases [published online ahead of print December 5, 2011].
Leuk Lymphoma
 
51
Rizzieri
 
DA
Koh
 
LP
Long
 
GD
, et al. 
Partially matched, nonmyeloablative allogeneic transplantation: clinical outcomes and immune reconstitution.
J Clin Oncol
2007
, vol. 
25
 
6
(pg. 
690
-
697
)
52
Luznik
 
L
O'Donnell
 
PV
Symons
 
HJ
, et al. 
HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
6
(pg. 
641
-
650
)
53
Sykes
 
M
Preffer
 
M
McAfee
 
SL
, et al. 
Lasting mixed lymphohematopoietic chimerism is achievable in adult humans following non-myeloablative therapy and HLA-mismatched donor bone marrow transplantation.
Lancet
1999
, vol. 
353
 (pg. 
1755
-
1756
)
54
Spitzer
 
TR
McAfee
 
SL
Dey
 
BR
, et al. 
Nonmyeloablative haploidentical stem-cell transplantation using anti-CD2 monoclonal antibody (MEDI-507)-based conditioning for refractory hematologic malignancies.
Transplantation
2003
, vol. 
75
 
10
(pg. 
1748
-
1751
)
55
Lee
 
KH
Lee
 
JH
Lee
 
JH
, et al. 
Reduced intensity conditioning therapy with busulfan, fludarabine, and antithymocyte globulin for HLA-haplodidentical hematopoietic cell transplantation in acute leukemia and myelodysplastic syndromes.
Blood:
2011
, vol. 
118
 
9
(pg. 
2609
-
2617
)
56
Ciurea
 
SO
Saliba
 
R
Rondon
 
G
, et al. 
Reduced-intensity conditioning using fludarabine, melphalan and thiotepa for adult patients undergoing haploidentical SCT.
Bone Marrow Transplant
2010
, vol. 
45
 
3
(pg. 
429
-
436
)
57
Bleakley
 
M
Otterud
 
BE
Richards
 
JL
, et al. 
Leukemia-associated minor histocompatibility antigen discovery using T-cell clones isolated by an invitro stimulation of naive CD8+ T cells.
Blood
2010
, vol. 
115
 
23
(pg. 
4923
-
4833
)
58
Rezvani
 
K
Barrett
 
AJ
Characterizing and optimizing immune responses to leukemia antigens after allogeneic stem cell transplantation.
Best Pract Res Clin Haematol
2008
, vol. 
21
 
3
(pg. 
437
-
453
)
59
Leen
 
AM
Christin
 
A
Myers
 
GD
, et al. 
Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein-Barr virus infections after haploidentical and matched unrelated stem cell transplantation.
Blood
2009
, vol. 
114
 
19
(pg. 
4283
-
4292
)
60
Barker
 
JN
Byam
 
C
Scaradavou
 
A
How I treat: the selection and acquisition of unrelated cord blood grafts.
Blood
2011
, vol. 
117
 
8
(pg. 
2332
-
2339
)
61
Pidala
 
J
Tomblyn
 
M
Nishihori
 
T
, et al. 
ATG prevents severe graft-versus-host disease in mismatched unrelated donor hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
8
(pg. 
1237
-
1244
)
62
Kim
 
H
Min
 
WS
Cho
 
SB
, et al. 
Successful prevention of acute graft-versus-host disease using low-dose anti-thymocyte globulin after mismatched, unrelated hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
6
(pg. 
704
-
717
)
63
Mead
 
AJ
Thomson
 
KJ
Morris
 
EC
, et al. 
HLA-mismatched unrelated donors are a viable alternate graft source for allogeneic transplantation following alemtuzumab-based reduced-intensity conditioning.
Blood
2010
, vol. 
115
 
25
(pg. 
5147
-
5153
)
64
Gooley
 
TA
Chien
 
JW
Pergam
 
SA
, et al. 
Reduced mortality after allogeneic hematopoietic-cell transplantation.
N Engl J Med
2010
, vol. 
363
 
22
(pg. 
2091
-
2101
)
65
Verneris
 
MR
Brunstein
 
CG
Barker
 
J
, et al. 
Relapse risk after umbilical cord blood transplantation: enhanced graft-versus-leukemia effect in recipients of 2 units.
Blood
2009
, vol. 
114
 
19
(pg. 
4293
-
4299
)
66
Rodrigues
 
CA
Sanz
 
G
Brunstein
 
CG
, et al. 
Analysis of risk factors for outcomes after unrelated cord blood transplantation in adults with lymphoid malignancies: a study by the Eurocord-Netcord and lymphoma working party of the European group for blood and marrow transplantation.
J Clin Oncol
2009
, vol. 
27
 
2
(pg. 
256
-
263
)
67
Arora
 
M
Weisdorf
 
DJ
Spellman
 
SR
, et al. 
HLA-identical sibling compared with 8/8 matched and mismatched unrelated donor bone marrow transplant for chronic phase chronic myeloid leukemia.
J Clin Oncol
2009
, vol. 
27
 
10
(pg. 
1644
-
1652
)
68
Majhail
 
NS
Weisdorf
 
DJ
Wagner
 
JE
, et al. 
Comparable results of umbilical cord blood and HLA matched sibling donor hematopoietic stem cell transplant after reduced-intensity preparative regimen for advanced Hodgkin's lymphoma.
Blood
2006
, vol. 
107
 
9
(pg. 
3804
-
3807
)
69
Burroughs
 
LM
O'Donnell
 
PY
Sandmaier
 
BM
, et al. 
Comparison of outcomes of HLA-matched related, unrelated, or HLA haploidentical related hematopoietic cell transplantation following nonmyeloablative conditioning for relapsed or refractory Hodgkin's lymphoma.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
11
(pg. 
1279
-
1287
)
70
Kasamon
 
YL
Luznik
 
L
Leffell
 
MS
, et al. 
Nonmyeloablative HLA-haploidentical bone marrow transplantation with high-dose posttransplantation cyclophosphamide: effect of HLA disparity on outcome.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
4
(pg. 
482
-
489
)
71
Parody
 
R
Martino
 
R
Rovira
 
M
, et al. 
Severe infections after unrelated donor allogeneic hematopoietic stem cell transplantation in adults: comparison of cord blood transplantation with peripheral blood and bone marrow transplantation.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
7
(pg. 
734
-
748
)
72
Ringden
 
O
Okas
 
M
Uhlin
 
M
, et al. 
Unrelated cord blood and mismatched unrelated volunteer donor transplants, two alternatives in patients who lack an HLA-identical donor.
Bone Marrow Transplant
2008
, vol. 
42
 
10
(pg. 
643
-
648
)
73
Walker
 
CM
van Burik
 
JH
De For
 
TE
Cytomegalovirus infection after allogeneic transplantation: comparison of cord blood with peripheral blood and marrow graft sources.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
9
(pg. 
1106
-
1115
)
74
Zhao
 
XY
Chang
 
YJ
Huang
 
XJ
Conflicting impact of alloreactive NK cells on transplantation outcomes after haploidentical transplantation: do the reconstitution kinetics of natural killer cells create these differences?
Biol Blood Marrow Transplant
2011
, vol. 
17
 
10
(pg. 
1436
-
1442
)
75
Jacobson
 
CA
Turki
 
AT
McDonough
 
SM
, et al. 
Immune reconstitution after double umbilical cord blood stem cell transplantation: comparison with unrelated peripheral blood stem cell transplantation [published online ahead of print August 29, 2011].
Biol Blood Marrow Transplant
 
76
Heslop
 
HE
Slobod
 
KS
Pule
 
MA
, et al. 
Long-term outcomes of EBV specific T cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients.
Blood
2010
, vol. 
115
 
5
(pg. 
925
-
935
)
77
Curtis
 
RE
Travis
 
LB
Rowlings
 
PA
, et al. 
Risk of lymphoproliferative disorders after bone marrow transplantation: a multi-institutional study.
Blood
1999
, vol. 
94
 
7
(pg. 
2208
-
2216
)
78
Brunstein
 
CG
Weisdorf
 
DJ
DeFor
 
T
, et al. 
Marked increased risk of Epstein-Barr virus-related complications with the addition of anti-thymocyte globulin to a nonmyeloablative conditioning prior to unrelated umbilical cord blood transplantation.
Blood
2006
, vol. 
108
 
8
(pg. 
2874
-
2880
)
79
Ballen
 
KK
Cutler
 
C
Yeap
 
BY
, et al. 
Donor-derived second hematologic malignancies after cord blood transplantation.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
7
(pg. 
1025
-
1031
)
80
Blaes
 
AH
Cao
 
Q
Wagner
 
JE
, et al. 
Monitoring and preemptive rituximab therapy for Epstein-Barr virus reactivation after anti-thymocyte globulin containing nonmyeloablative conditioning for umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
2
(pg. 
287
-
291
)
81
Dominietto
 
A
Tedone
 
E
Soracca
 
M
, et al. 
In vivo B-cell depletion with rituximab for alternative donor hematopoietic SCT.
Bone Marrow Transplant
2012
, vol. 
47
 
1
(pg. 
101
-
106
)
82
Heslop
 
HE
How I treat EBV lymphoproliferation.
Blood
2009
, vol. 
114
 
19
(pg. 
4002
-
4008
)
83
Haque
 
T
Wilkie
 
GM
Jones
 
MM
, et al. 
Allogeneic cytotoxic T-cell therapy for EBV-positive posttransplantation lymphoproliferative disease: results of a phase 2 multicenter clinical trial.
Blood
2007
, vol. 
110
 
4
(pg. 
1123
-
1131
)
84
Fraser
 
CJ
Hirsch
 
BA
Dayton
 
V
, et al. 
First report of donor cell-derived acute leukemia as a complication of umbilical cord blood transplantation.
Blood
2005
, vol. 
106
 
13
(pg. 
4377
-
4380
)
85
Greaves
 
M
Cord blood donor cell leukemia in recipients.
Leukemia
2006
, vol. 
20
 
9
(pg. 
1633
-
1634
)
86
Majhail
 
NS
Mothkuri
 
JM
Brunstein
 
CG
Weisdorf
 
DF
Costs of hematopoietic cell transplantation: comparison of umbilical cord blood and matched related donor transplantation and the impact of posttransplant complications
Biol Blood Marrow Transplant
2009
, vol. 
15
 
5
(pg. 
564
-
573
)
87
Sebrango
 
A
Vicuna
 
I
De Laiglesia
 
A
, et al. 
Hematopoietic transplants combining a single unrelated cord blood unit and mobilized hematopoietic stem cells from an adult HLA-mismatched third party donor: comparable results to transplants from HLA-identical related donors in adults with acute leukemia and myelodysplastic syndrome.
Best Pract Res Clin Haematol
2010
, vol. 
23
 
2
(pg. 
259
-
274
)
88
Liu
 
H
Rich
 
ES
Godley
 
L
, et al. 
Reduced-intensity conditioning with combined haploidentical and cord blood transplantation results in rapid engraftment, low GVHD, and durable remissions.
Blood
2011
, vol. 
118
 
24
(pg. 
6438
-
6445
)
89
Bautista
 
G
Cabrera
 
R
Regidor
 
C
, et al. 
Cord blood transplants supported by co-infusion of mobilized hematopoietic stem cells from a third party donor.
Bone Marrow Transplant
2009
, vol. 
43
 
5
(pg. 
365
-
373
)
90
Boo
 
M
Ballen
 
K
Maiers
 
M
Cord blood unit access and selection: 2010 and beyond. Best practices and emerging trends in cord blood unit selection.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
1 suppl
(pg. 
S46
-
S51
)
91
Cutler
 
C
Kim
 
HT
Sun
 
L
, et al. 
Donor-specific anti-HLA antibodies predict outcome in double umbilical cord blood transplantation.
Blood
2011
, vol. 
118
 
25
(pg. 
6691
-
6697
)
92
Eapen
 
M
Klein
 
JP
Sanz
 
G
, et al. 
Effect of donor-recipient HLA matching at HLA A, B, C, and DR B1 on outcomes after umbilical-cord blood transplantation for leukemia and myelodysplastic syndrome: a retrospective analysis.
Lancet Oncol
2011
, vol. 
12
 
13
(pg. 
1214
-
1221
)
93
Kroger
 
N
Zabelina
 
T
Binder
 
T
, et al. 
HLA-mismatched unrelated donors as an alternative graft source for allogeneic stem cell transplantation after anti-thymocyte globulin-containing conditioning regimens.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
4
(pg. 
454
-
462
)
Sign in via your Institution