Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 1.
Published in final edited form as: Biol Blood Marrow Transplant. 2012 Feb 16;18(8):1265–1272. doi: 10.1016/j.bbmt.2012.01.019

UNRELATED DONOR CORD BLOOD TRANSPLANTATION FOR CHILDREN WITH SEVERE SICKLE CELL DISEASE: RESULTS OF ONE COHORT FROM THE PHASE II STUDY FROM THE BLOOD AND MARROW TRANSPLANT CLINICAL TRIALS NETWORK (BMT CTN)

Naynesh R Kamani 1, Mark C Walters 2, Shelly Carter 3, Victor Aquino 4, Joel A Brochstein 5, Sonali Chaudhury 6, Mary Eapen 7, Brian M Freed 8, Michael Grimley 9, John E Levine 10, Brent Logan 7, Theodore Moore 11, Julie Panepinto 7, Suhag Parikh 12, Michael A Pulsipher 13, Jane Sande 1, Kirk R Schultz 14, Stephen Spellman 15, Shalini Shenoy 16
PMCID: PMC3618440  NIHMSID: NIHMS384639  PMID: 22343376

Abstract

The Sickle Cell Unrelated donor Transplant trial (SCURT trial) of the Blood and Marrow Transplant Clinical Trials Network (BMT CTN) is a phase II study of the toxicity and efficacy of unrelated donor hematopoietic cell transplantation in children with severe sickle cell disease (SCD) using a reduced intensity conditioning regimen. Here we report the results for the cord blood cohort of this trial. Eight children with severe SCD underwent unrelated donor cord blood transplantation (CBT) following alemtuzumab, fludarabine and melphalan. Cyclosporine or tacrolimus and mycophenolate mofetil were administered for graft-versus-host disease (GVHD) prophylaxis. Donor/recipient HLA match status was 6/6 (n=1) or 5/6 (n=7), based on low/intermediate resolution molecular typing at HLA -A, -B, and high resolution typing at -DRB1. Median recipient age was 13.7 years (range 7.4 to 16.2 years), and median weight was 35.0 kg (range 25.2 to 90.2 kg). The median pre-cryopreservation total nucleated cell dose was 6.4 × 107 /kg (range 3.1 to 7.6) and the median post-thaw infused CD34 cell dose was 1.5 × 105 /kg (range 0.2 to 2.3). All patients achieved neutrophil recovery (ANC>500/mm3) by day 33 (median 22 days). Three patients who engrafted had 100% donor cells by day 100, which was sustained and five patients had autologous hematopoietic recovery. Six of eight patients had a platelet recovery to > 50,000/mm3 by day 100. Two patients developed grade II acute GVHD. Of these, one developed extensive chronic GVHD and died of respiratory failure 14 months post-transplant. With a median follow up of 1.8 years (range 1 – 2.6), 7 patients are alive with a 1-year survival of 100% and 3 of 8 are alive without graft failure or disease recurrence.

Conclusion

Based upon the high incidence of graft rejection after unrelated donor CBT, enrollment onto the cord blood arm of the SCURT trial was suspended. However, since this reduced intensity regimen has demonstrated a favorable safety profile, this trial remains open to enrollment for unrelated marrow donor transplants. Novel approaches aimed at improving engraftment will be needed before unrelated CBT can be widely adopted for transplanting patients with severe SCD.

Keywords: cord blood, unrelated donor, transplantation, children, sickle cell disease

INTRODUCTION

Several studies have shown that hematopoietic cell transplantation (HCT) for sickle cell disease using HLA-identical sibling donors has curative potential, including a NIH-supported multicenter prospective trial that demonstrated an event-free survival (EFS) probability of 85% in children following a myeloablative preparative regimen of busulfan, cyclophosphamide and anti-thymocyte globulin1. In addition, long-term follow-up studies from this and other patient series have demonstrated cessation of sickle cell disease-related events after successful HCT, even among those with mixed donor-host hematopoietic chimerism25. However, these observations have done little to expand the application of HCT for sickle cell disease because fewer than 20% of individuals have HLA-identical sibling donors and because there are ongoing concerns about the toxicity of this therapy. To address these challenges, a prospective multicenter, phase II clinical trial of HCT was initiated using a reduced intensity conditioning regimen and well matched unrelated donor cells. The over-arching goal of this ongoing trial is to assess the toxicity and efficacy of such an approach and to expand the availability of HCT so that more individuals with severe sickle cell disease might benefit from this treatment option.

The hypothesis for this reduced intensity regimen is the premise that host immune ablation can support successful donor cell engraftment without myeloablation. The trial was developed based on the results of a preliminary study of reduced intensity HCT in 16 children with non-malignant disorders who received alemtuzumab, fludarabine and melphalan before HLA-identical sibling or unrelated donor HCT 6,7. Graft rejection and graft-versus-host disease rates were low, and prompt engraftment occurred after transplantation from both related and unrelated donors. No late graft rejection was noted despite the early cessation of post-grafting immunosuppression. The regimen-related toxicity observed was early and related primarily to infections associated with this highly immunosuppressive conditioning regimen. This experience was distinctly different from the very high rate of graft rejection that was observed in several small series after non-myeloablative HCT for sickle cell disease8,9, and in another recent trial of non-myeloablative MSD transplantation for adults with SCD where post-grafting immunosuppression had to be extended indefinitely to prevent late graft rejection10. Thus, we reasoned that our reduced-intensity conditioning approach might be sufficiently immunoablative to suppress host-versus-graft rejection and promote engraftment of donor cells after unrelated donor HCT in sickle cell disease. With careful attention to infection prevention and pre-emptive treatment of viral reactivation, it was also our aim to reduce the short and long-term toxicity of transplantation.

The preliminary results of the ongoing phase II trial strongly suggest that this is a safe approach to unrelated donor HCT for sickle cell disease. However, we have observed a high rate of graft rejection after unrelated donor CBT, which has resulted in closure of the cord blood arm of the study and caused us to look more carefully at factors that may have contributed to graft rejection. Here, we present the preliminary results for the cord blood cohort of this multicenter trial which continues to enroll patients with 8/8 HLA-allele matched unrelated bone marrow donors.

STUDY DESIGN AND METHODS

Study Design

The SCURT trial is a Phase II multi-center trial designed to estimate the efficacy and toxicity of unrelated donor HCT using a reduced-intensity conditioning regimen in children with SCD and high risk features. It was initiated in 2008 and included 2 cohorts based on the donor hematopoietic stem cell source. The primary objective of the study was to determine event-free survival at 1 year after URD transplant using either bone marrow or cord blood as a stem cell source. The secondary objectives included the determination of the effects of HCT on clinical and laboratory manifestations of SCD and the incidence of HCT complications such as transplant-related toxicity, graft failure, acute and chronic GVHD. Stopping rules for the study included unacceptable rates of mortality, GVHD or graft rejection.

Patients

The protocol was approved by the Institutional Review Board at each of the 7 institutions that enrolled patients and by the IRB of the National Marrow Donor Program. Consent was obtained from legal guardians for all patients prior to enrollment; assents were obtained where appropriate. In addition, prior to enrollment, an independent external review committee approved enrollment by ensuring that the patient met disease related eligibility criteria. An important eligibility criteria design consideration was to enroll only patients with sickle cell disease manifestations associated with poor long term outcomes despite optimal supportive care. For the patients described in this report, eligibility included recipients 3–16 years of age with severe SCD with one or more of the following: 1) a clinically significant neurologic event (overt stroke) defined as any neurologic defect lasting > 24 hours and accompanied by an infarct on cerebral magnetic resonance imaging (MRI); OR, 2) a minimum of two episodes of acute chest syndrome (ACS) within the preceding 2-year period defined as new pulmonary alveolar consolidation involving at least one complete lung segment (associated with acute symptoms including fever, chest pain, tachypnea, wheezing, rales or cough that is not attributed to asthma or bronchiolitis) despite adequate supportive care measures; or, 3) a history of 3 or more severe pain events or vaso-occlusive crises (VOC) per year in the 2 years prior to enrollment. An additional criterion for patients with recurrent ACS or VOC was the requirement that they either fail to significantly benefit from or were unable or unwilling to continue supportive care therapy that included hydroxyurea.

Patients with HLA-matched sibling donors, HIV seropositivity, Lansky performance score less than 50, or uncontrolled bacterial, viral or fungal infections were ineligible. Multi-organ assessment performed prior to enrollment was required to demonstrate the following: serum creatinine < 1.5 upper limit of normal for age and GFR > 100 ml/min/1.73 m2; ALT and AST < 5 times upper limits of normal and direct serum bilirubin < 2 × upper limit of normal; left ventricular ejection fraction > 40% or LV shortening fraction > 26%; and DLCO > 40% of predicted (corrected for hemoglobin). For patients who had received regular blood transfusions for over a year and whose serum ferritin was > 1000 ng/ml, a liver biopsy had to demonstrate the absence of cirrhosis or bridging fibrosis. The Hb S percentage had to be ≤ 45% 7 days prior to initiation of alemtuzumab. Iron chelation and/or hydroxyurea had to be discontinued 48 hours prior to alemtuzumab.

Donor Selection

The donor/recipient HLA match for UCBT was accomplished by low/intermediate resolution molecular typing for class I HLA-A and -B alleles, and high resolution molecular typing for HLA-DRB1 alleles (original HLA match). For the purpose of this report, high resolution molecular typing for HLA-A, -B, -C, and -DRB1 alleles was retrospectively reviewed and reported as the final HLA match. The selected CBU was required to be ≥ 5/6 matched and provide a minimum of 3 × 107 total nucleated cells (pre-cryopreservation) per kilogram (kg) of recipient weight.

Preparative regimen

All patients received increasing doses of alemtuzumab intravenously daily for 3 days (10 mg, 15 mg and 20 mg) starting on day -21 after a 3 mg intravenous test dose. Patients were discharged after alemtuzumab administration and readmitted to start fludarabine 30 mg/m2 daily from day – 8 to day -4. Melphalan (140 mg/m2/day) was given on day -36. GVHD prophylaxis consisted of cyclosporine or tacrolimus and mycophenolate mofetil (MMF). Cyclosporine or tacrolimus was started on day -3 with regular monitoring to maintain therapeutic levels and continued to day 100, then tapered weekly and discontinued on day + 180 if recipients had no evidence of GVHD. MMF (1 gram IV q 8 hours for children ≥50 kg or 15 mg/kg IV q 8 hours for children < 50 kg) was started on day -3 and given through Day +45 or 7 days after engraftment, whichever was later. Monitoring of MMF levels was not mandated by the protocol. G-CSF 5 µg/kg/day was started on day +7 and continued until neutrophil engraftment.

Endpoint Definitions

The primary endpoint of the study was event-free survival at 1 year post-transplant. Primary or late graft rejection, disease recurrence, or death was considered an event for this endpoint. Primary graft rejection was defined as the presence of < 20% donor cells as assessed by bone marrow or peripheral blood chimerism assays on Day 42 or the infusion of a second stem cell product on or prior to Day 42. Late graft rejection was defined as the presence of < 20% donor derived hematopoietic cells in peripheral blood or bone marrow after Day 42 in a patient with prior evidence of > 20% donor cells or the infusion of a second stem cell product beyond Day 42. Survival was defined as the time from transplant to day of death or day of last follow up. Neutrophil recovery was defined as the first day of achieving an ANC of at least 500/mm3 for three consecutive days. Platelet recovery was defined as the first day of a minimum of three consecutive measurements on different days that the patient had achieved a platelet count > 50,000/mm3, and was platelet transfusion independent for a minimum of seven days. The time to neutrophil or platelet recovery was defined as the time from transplant to the first day of engraftment.

Acute GVHD assessments were made every 7 days to day 100 post-CBT, 6 months, 1 year and 2 years after CBT. The grading of acute GVHD followed GVHD consensus grading scheme and methods of Weisdorf et al11. Clinically significant infections were reported post-transplant by site of infection, organism and severity.. Re–admissions after the initial discharge following transplant were reported by date and primary and secondary reasons for admission. The Common Terminology Criteria for Adverse Events (CTCAE) version 3.0 was used to report expected grade 3–5 adverse events through 1 year post-transplant. Maximum CTCAE grades across all organ systems were reported monthly for 3 months and at 6 and 12 months post-transplant.

A truncated sequential probability ratio test (SPRT) for a binomial outcome was used to monitor for each of the three key safety endpoints, namely overall mortality, graft rejection and GVHD. The stopping rules were triggered if there was significant evidence that the day 100 overall mortality rate was greater than 15% based on the truncated SPRT, or if the day 100 graft rejection rate exceeded 20% or if the day 100 grade III–IV GVHD rate exceeded 15%.

Anti-HLA Donor Specific Antibodies

A retrospective analysis for the presence of anti-HLA donor specific antibodies (DSA) was carried out in 6 recipients using stored pre-transplant sera. Donor mismatched HLA alleles were identified and recipient DSA response was screened using the LABScreen® Single Antigen HLA Class I/II Antibody Detection system that uses microbeads coated with purified Class I or Class II HLA antigens for the detection of Class I or Class II HLA antibodies in the patient’s plasma. Each bead in a LABScreen Single Antigen assay is specific for a single HLA antigen permitting assessment of specificity for mismatched antigens carried by the donor.

Statistical Analysis

Survival estimates were calculated using the Kaplan-Meier method. Neutrophil and platelet recoveries, acute GVHD and chronic GVHD, and graft rejection/disease relapse were analyzed using the cause-specific failure probability method (or cumulative incidence, CINC) 12. Due to the small sample size, only univariate analyses were performed.

RESULTS

Study Population

Eight patients were enrolled for UCBT at 7 transplant centers. Baseline characteristics are shown in Table 1. The median age was 13.8 years (range 7.4 to 16.2). The primary diagnosis was Hemoglobin SS disease (n=6), Hgb S/β0 Thalassemia (n=1) and Hgb S/β+ Thalassemia (n=1). The indication for CBT was stroke in 2 patients, recurrent ACS in 3 patients and recurrent VOC in 3 patients. The median pre-cryopreserved total nucleated cell (TNC) dose was 6.4 × 107 /kg (range 3.1 to 7.6), and the post thaw infused TNC and CD34+ cell dose per kg were 4.5 × 107 (range 2.1 to 6.3) and 1.5 × 105 (range 0.2 – 2.3) respectively.

Table 1.

Patient Demographics

Patient
ID
Gender Race Age at
transplant
(yrs)
Disease
Type
Lansky
Score
Indication for
Transplant
02376 F Black/AA 11.2 Hgb SS 80 VOC
02395 F Black/AA 16.3 Hgb SS 80 Stroke
02497 M Black/AA 14.3 Hgb SS 80 ACS
02541 F Black/AA 7.4 Hgb SS 100 ACS
02611 M Black/AA 13.9 Hgb Sβo thal 100 VOC
02613 F Black/AA 10.9 Hgb SS 100 ACS
02760 F White Hispanic 15.0 Hgb Sβ+ thal 100 VOC
02901 F Black/AA 13.6 Hgb SS 100 Stroke

Results of donor-recipient blood groups and HLA matching are shown in Table 2. Based on original donor-recipient HLA typing, all 8 patients were matched at 5 of 6 HLA antigens. However, at the allele level high resolution molecular HLA typing for HLA-A, –B, -C and –DRB1, all donor/recipient pairs were mismatched for one (n=2), two (n=4), or three (n=2) alleles. Of note, both patients who were allele matched at the C locus engrafted whereas 5/6 mismatched at one or both C loci rejected their grafts. The retrospective analysis for the presence of anti-HLA donor specific antibodies failed to detect DSA in any of the six pre-transplant serum samples tested.

Table 2.

Donor-Recipient Characteristics

Patient
ID
Original
HLA
Match
Donor -
Recipient
Allele level HLA
Match
Prior Chronic
Transfusion
Therapy
Recipient
Blood
Group
Donor
Blood
Group
Red-cell
Alloantibody
Anti-donor
specific
HLA
Antibodies
02376 5/6 7/8 Yes B +ve A +ve Yes No
02395 5/6 6/8 Yes A +ve A +ve No No
02497 5/6 5/8 Yes A +ve O −ve Yes not tested
02541 5/6 6/8 No O +ve O +ve Yes No
02611 5/6 6/8 No O +ve B +ve No No
02613 5/6 6/8 No A +ve B +ve No No
02760 5/6 7/8 No AB +ve O +ve No No
02901 5/6 5/8 Yes B +ve O +ve No not tested

Engraftment and Survival

All eight patients achieved the neutrophil recovery with an ANC > 500/mm3 at a median of 22 days (range 13–33) after CBT. Six of eight patients had platelet recovery to 50,000/mm3 by day 100. In five patients, blood count recovery was the result of graft rejection and autologous hematopoietic reconstitution, as demonstrated by 100% recipient DNA in chimerism studies performed on day 42 samples. No late graft rejections were observed and the remaining three patients had sustained donor engraftment through last follow-up. Due to primary graft rejection, the one-year event free survival was 37.5% (95% CI: 8.7 – 67.4%). All graft rejections were non-lethal and were not characterized by marrow aplasia. With a median follow-up of 1.8 years (1 to 2.6), one year survival was 100%. The solitary death on this study occurred on day 430 from complications of chronic GVHD.

Acute and Chronic GVHD

The maximum GVHD grade was grade II (n=2). An additional patient developed a limited skin rash (grade I). There was one case of chronic extensive GVHD.

Regimen Related Toxicity

Serious, but reversible, transplant related neurological toxicities in the first 100 days developed in two patients. One patient, with hypertension, experienced a grade 4 intraventricular hemorrhage with seizures on day 7. A second patient developed posterior reversible leukoencephalopathy syndrome due to cyclosporine. Both patients experienced complete recovery. Two patients had grade 4 liver toxicity, which was transient. No patient developed idiopathic pneumonia syndrome or hepatic veno-occlusive disease. One patient developed EBV reactivation with post-transplant lymphoproliferative disease and a brain abscess in the context of chronic GVHD and eventually died on day 430 from respiratory failure secondary to pneumonia.

There were a total of 19 infections (bacterial and viral) reported in six of eight patients with no infections reported in 2 patients. These included 9 episodes of bacteremia (8 gram positive organisms, 1 Klebsiella), 3 patients with CMV viremia, 1 with EBV reactivation progressing to PTLD, 3 with upper respiratory tract infections (1 adenovirus, 1 parainfluenza-3 and 1 Pseudomonas), 1 with lower respiratory tract parainfluenza-3 infection, 1 adenovirus gastroenteritis and 1 C. difficile enterocolitis.

DISCUSSION

Although allogeneic hematopoietic cell transplantation (HCT) has curative potential for patients with severe SCD, there are two major barriers to making this therapy available to eligible patients: the absence of HLA identical sibling donors and concerns about the risk of mortality or treatment related toxicities13. In order to circumvent these barriers, we initiated a clinical trial of a reduced intensity preparative regimen before unrelated donor HCT for severe sickle cell disease, in particular relying upon the use of cord blood as a source of hematopoietic cells for allogeneic transplantation in patients with hemoglobinopathies14,15. With cord blood as a source of hematopoietic cells, it was our expectation that the risk of graft-vs-host disease would be lower compared to marrow-derived hematopoietic cells and that ≥ 5/6 matched CB units would be available for the vast majority of potential recipients16,17. In the first report of sibling donor CBT using a myeloablative regimen in 44 children with hemoglobinopathies, the EFS was 79% for thalassemia and 90% for SCD with no treatment related mortality; all the treatment failures were related to graft rejection 18. Thus, we reasoned that the risk of graft rejection/disease recurrence might pose a similar challenge after unrelated CBT and we applied stringent HLA matching and cell dose requirements in anticipation of this problem.

Despite these considerations, we observed a high incidence of graft rejection after CBT (5 of 8 recipients) that was accompanied by autologous recovery of sickle erythropoiesis by 6 weeks after transplantation. This experience is consistent with other reports of graft rejection following non-myeloablative or reduced intensity preparation before CBT and suggests that further intensification in the preparative regimen may be necessary to ensure donor engraftment. An early report of unrelated CBT in 3 children with SCD included a single case of graft rejection followed by recovery of autologous hematopoiesis19, and successful donor cell engraftment followed by GVHD in 2 of 3 children. In a subsequent retrospective review of a four-center experience with unrelated cord blood transplantation in children with SCD that included 4 additional recipients (n=7), 5 patients received a single cord blood unit matched at 4/6 HLA-antigens and 2 were matched 5/6 HLA-antigens. All 3 patients who received a reduced intensity regimen experienced graft rejection. Of the 4 who received myeloablative regimens, 3 engrafted and 2 developed grade III-IV acute GVHD20. A recent retrospective survey of UCBT in 51 patients with hemoglobinopathies conducted by the CIBMTR with Eurocord and NY Blood Center participation observed primary graft failure in 7/16 recipients despite the use of myeloablative conditioning therapy in 4 of the 7 patients. A better disease-free survival among recipients that received CBT with a TNC content ≥ 5 × 107/kg was observed21. The notion that a larger cellular content might promote durable engraftment after CBT for hemoglobinopathies was also suggested in a report by Jaing et al of successful durable donor engraftment in 4 of 5 children with thalassemia major who received dual unrelated CB units matched at 4/6 HLA-antigens22. Thus, CBT from unrelated donors appears to present unique challenges to a successful outcome among recipients with hemoglobinopathies.

Another factor that might contribute to the high incidence of graft rejection that we and others have observed after CBT for sickle cell disease is donor-recipient HLA incompatibility. In a cohort of 269 patients with hematologic malignancies undergoing myeloablative BMT from mismatched related donors, there was a linear correlation between a higher rate of graft rejection and increasing donor HLA incompatibility with prior alloimmunization (as assessed by a positive crossmatch for anti-donor lymphocytotoxic antibody) 23. A non-engraftment/graft failure rate that approached 20% was observed in the largest trial of unrelated CBT (COBLT) in children with hematologic malignancies following a myeloablative regimen 24. This was primarily attributed to cell dose and donor-recipient mismatching as a lower cell dose and a greater degree of HLA mismatching were associated with higher non-engraftment rates2528. In patients with hemoglobinopathies, these risk factors are probably amplified by the effect of multiple transfusion exposures that might sensitize the recipient to donor alloantigens. In addition, the cytokine milieu of sickle cell disease, which is one of inflammation and immune activation at the baseline, might also promote a host-versus-graft reaction and thereby interfere with engraftment even after myeloablative preparation29. While it is possible this risk might be overcome by employing more immunosuppressive preparative regimens by the addition of either antithymocyte globulin or low dose total body irradiation as reported in multiply transfused patients with aplastic anemia3032, we found that even the use of a highly immunosuppressive conditioning regimen was not sufficient to overcome these factors in patients with severe sickle cell disease.

While donor recipient ABO incompatibility can cause complications such as delayed red blood cell engraftment, pure red cell aplasia or hemolytic anemia, it is not associated with graft rejection in patients with hematologic malignancies33. However, RBC alloimmunization recently was reported as an independent predictor of HLA alloimmunization34. The RBC alloimmunization frequency approaches 25% of SCD patients in the absence of extended phenotype matching of RBC transfusions and thus might have contributed to graft rejection in this series. A retrospective analysis of archived pre-transplant sera from unrelated donor HCT recipients showed that the presence of donor-directed, HLA-specific alloantibodies was significantly associated with graft failure35. A similar recent analysis of sera from 386 myeloablative UCBT recipients showed that the presence of donor-specific antibodies correlated with significantly lower neutrophil recovery as compared to those who lacked alloantibodies36. The presence of DSA has also been shown to predict outcomes in double umbilical CBT with higher graft failure and 100 day mortality in those with preformed DSA37. We were unable to demonstrate the presence of HLA-specific DSA in any of the six patients in whom stored pre-transplant serum samples were available for testing.

Van Rood et al analyzed the impact of administering a CB unit that had a non-inherited maternal HLA antigen (NIMA) that was shared with a mismatched HLA antigen in the cord blood donor among patients with hematologic malignancies treated by UCBT. They showed that having a donor with a NIMA shared HLA-antigen resulted in lower transplant related mortality and speculated that this was related to improved neutrophil recovery especially in those who received a low TNC cell dose38. Thus, in the future, it might be important to perform screening for HLA directed allo-antibodies and use novel donor selection criteria to improve the results of CBT for sickle cell disease.

We speculate that a number of modifications should be explored to improve the rate of engraftment after UCBT for severe SCD. The use of a myeloablative regimen might improve outcomes by overcoming host barriers to engraftment, but this effect can also cause more short and long-term toxicity including marrow aplasia in the setting of a graft rejection. Alternatively, we have observed very little toxicity with the regimen currently employed in the SCURT trial, and thus have considered modifications that might not significantly alter the toxicity profile while increasing the immunosuppressive intensity of the regimen. The addition of hydroxyurea well before HCT to eradicate thalassemic clones and/or the addition of thiotepa to the preparative regimen resulted in lower rejection rates after HLA-ID sibling BMT in advanced thalassemia39,40 and is currently being studied in a reduced intensity study of unrelated CBT for thalassemia. A higher TNC dose might also improve engraftment21. This might also be accomplished by the use of dual cord blood unit transplantation, however this approach has not been validated in the context of a prospective clinical trial22,41,42. Whenever possible, the selection of CB units that are negative for antigens against which there are pre-existing recipient antibodies should be pursued. At the present time, the role of NIMA matching in impacting engraftment in UCBT is unclear and requires additional investigation. In summary, because the less stringent need for HLA matching in CBT might still expand HCT access to many SCD patients who might not otherwise be able to pursue HCT, strategies to overcome engraftment barriers in CBT should be pursued and tested in clinical trials.

Table 3.

Transplant Characteristics

Patient
ID
Pre-
Thaw
TNC
Dose
(×107/k
g)
Post
Thaw
TNC Dose
(×107/kg)
Post
Thaw
CD34+
Dose
(×105/k
g)
CMV
status
Intra-transplant
Complications (Day 90)
Day to
ANC >
500
Day
to Plt
> 50K
Donor chimerism
Day +42
Acute
GVHD
Chronic
GVHD
Outcome
02376 7.4 6.1 1.7 Pos Grade II GVHD +19 +31 100% donor Gr II day +28 None Alive Day +704
02395 3.1 2.1 0.2 Neg Seizure with intraventricular hemorrhage; C. difficile infection +29 NR 100% host (6% donor Day +98; 0% donor day +170) None None Graft Rejection;
02497 3.7 2.5 0.5 Pos CMV viremia S. viridans infection +25 +36 100% host None None Graft Rejection
02541 6.9 5.5 1.7 Neg None +33 NR 100% host None None Graft Rejection; disease recurrence day 181
02611 6.0 4.7 1.5 Pos CMV viremia Streptococcus infection +22 +40 100% host None None Graft Rejection
02613 7.6 6.3 2.3 Pos CMV viremia Grade II GVHD +14 +47 100% donor Gr II day +14 Extensi ve cGVHD day +393 Died on day +430
02760 3.4 2.5 1.0 Neg Gemella infection +13 +42 100% donor None None Alive Day + 365
02901 6.7 4.3 Not done Pos Fever day 11, presumed engraftment syndrome +29 +45 100% host None None Graft Rejection disease recurrence day 127

ACKNOWLEDGEMENTS

We also gratefully acknowledge the contributions of Nancy Poland and Jeffrey Chell, MD who provided support to the conduct of this trial and to Nancy DiFronzo, PhD, who provided support to the conduct of this trial and helpful suggestions to the preparation of the manuscript. We would like to thank the patients and parents who consented to participation.

This BMT CTN trial is supported in part by grant #U01HL069294 from the National Heart, Lung, and Blood Institute and the National Cancer Institute, and by the National Marrow Donor Program, NIH’s National Center of Minority Health and Health Disparities and the NHLBI’s Sickle Cell Disease Clinical Research Network

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Contribution: NRK and SS: participated in the study design, data analysis and interpretation, and wrote the paper; MW, SLC, JEB, ME, JEL, SP, JP, MP, KS participated in the study design and revised the manuscript; B.R.L. participated in study design, data analysis and interpretation, performed statistical analysis, and revised the manuscript. SS and BF performed the DSA analysis; VA, JB, SC, MG, TM, SP, and JS enrolled patients and provided clinical care.

Bibliography

  • 1.Walters M, Patience M, Leisenring W, Rogers Z, Aquino V, Buchanan G, et al. Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anemia. Biol Blood Marrow Transplant. 2001;7(12):665–673. doi: 10.1053/bbmt.2001.v7.pm11787529. [DOI] [PubMed] [Google Scholar]
  • 2.Bernaudin F, Socie G, Kuentz M, Chevret S, Duval M, Bertrand Y, et al. Long-term results of myeloablative stem-cell transplantation to cure sickle cell disease. Blood. 2007;110(7):2749–2756. doi: 10.1182/blood-2007-03-079665. [DOI] [PubMed] [Google Scholar]
  • 3.Vermylen C, Cornu G, Ferster A, Brichard B, Ninane J, Ferrant A, et al. Haematopoietic stem cell transplantation for sickle cell anemia: the first 50 patients transplanted in Belgium. Bone Marrow Transplant. 1998;22(1):1–6. doi: 10.1038/sj.bmt.1701291. [DOI] [PubMed] [Google Scholar]
  • 4.Walters M, Patience M, Leisenring W, Eckman J, Scott J, Mentzer W, et al. Bone marrow transplantation for sickle cell disease. N Engl J Med. 1996;335(6):369–376. doi: 10.1056/NEJM199608083350601. [DOI] [PubMed] [Google Scholar]
  • 5.Eggleston B, Patience M, Edwards S, Adamkiewicz T, Buchanan G, Davies S, et al. Effect of myeloablative bone marrow transplantation on growth in children with sickle cell anaemia: results of the multicenter study of haematopoietic cell transplantation for sickle cell anaemia. Br J Haematol. 2007;136(4):673–676. doi: 10.1111/j.1365-2141.2006.06486.x. [DOI] [PubMed] [Google Scholar]
  • 6.Shenoy S, Grossman W, DiPersio J, Yu L, Wilson D, Barnes Y, et al. A novel reduced-intensity stem cell transplant regimen for nonmalignant disorders. Bone Marrow Transplant. 2005;35(4):345–352. doi: 10.1038/sj.bmt.1704795. [DOI] [PubMed] [Google Scholar]
  • 7.Rao A, Kamani N, Filopovich A, Lee SM, Davies SM, Dalal J, Shenoy S. Successful bone marrow transplantation for IPEX syndrome after reduced intensity conditioning. Blood. 2007;109(1):383–385. doi: 10.1182/blood-2006-05-025072. [DOI] [PubMed] [Google Scholar]
  • 8.Iannone R, Casella J, Fuchs E, Chen A, Jones R, Woolfrey A, et al. Results of minimally toxic nonmyeloablative transplantation in patients with sickle cell anemia and beta-thalassemia. Biol Blood Marrow Transplant. 2003;9(8):519–528. doi: 10.1016/s1083-8791(03)00192-7. [DOI] [PubMed] [Google Scholar]
  • 9.Horan J, Liesveld J, Fenton P, Blumberg N, Walters M. Hematopoietic stem cell transplantation for multiply transfused patients with sickle cell disease and thalassemia after low-dose total body irradiation, fludarabine, and rabbit anti-thymocyte globulin. Bone Marrow Transplant. 2005;35(2):171–177. doi: 10.1038/sj.bmt.1704745. [DOI] [PubMed] [Google Scholar]
  • 10.Hsieh M, Kang E, Fitzhugh C, Link M, Bolan C, Kurtlander R, et al. Allogeneic hematopoietic stem-cell transplantation for sickle cell disease. N Engl J Med. 2009;361(24):2309–2317. doi: 10.1056/NEJMoa0904971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Weisdorf D, Hurd D, Carter S, Howe C, Jensen L, Wagner J. Prospective grading of graft-versus-host disease after unrelated donor marrow transplantation: a grading algorithm versus blinded expert panel review. Biol Blood Marrow Transplant. 2003;9(8):512–518. doi: 10.1016/s1083-8791(03)00162-9. [DOI] [PubMed] [Google Scholar]
  • 12.Gooley T, Leisenring W, Crowley J, Storer B. Estimation of failure probabilities in the presence of competing risks: new representations of old estimators. Stat Med. 1999;18(6):695–706. doi: 10.1002/(sici)1097-0258(19990330)18:6<695::aid-sim60>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 13.Walters M, Patience M, Leisenring W, Eckman J, Buchanan G, Rogers Z, et al. Barriers to bone marrow transplantation for sickle cell anemia. Biol Blood Marrow Transplant. 1996;2(2):100–104. [PubMed] [Google Scholar]
  • 14.Kelly P, Kurtzberg J, Vichinsky E, Lubin B. Umbilical cord blood stem cells: application for the treatment of patients with hemoglobinopathies. J Pediatr. 1997;130(5):695–703. doi: 10.1016/s0022-3476(97)80009-1. [DOI] [PubMed] [Google Scholar]
  • 15.Laughlin M. UCB allogeneic transplantation for hemoglobinopathies. Blood. 2003;101(6):2077. [Google Scholar]
  • 16.Adamkiewicz T, Boyer M, Bray R, Haight A, Yeager A. Identification of unrelated cord blood units for hematopoietic stem cell transplantation in children with sickle cell disease. J Pediatr Hematol Oncol. 2006;28(1):29–32. [PubMed] [Google Scholar]
  • 17.Krishnamurti L, Abel S, Maiers M, Flesch S. Availability of unrelated donors for hematopoietic stem cell transplantation for hemoglobinopathies. Bone Marrow Transplant. 2003;31(7):547–550. doi: 10.1038/sj.bmt.1703887. [DOI] [PubMed] [Google Scholar]
  • 18.Locatelli F, Rocha V, Reed W, Bernaudin F, Ertem M, Grafakos S, et al. Related umbilical cord blood transplantation in patients with thalassemia and sickle cell disease. Blood. 2003;101(6):2137–2143. doi: 10.1182/blood-2002-07-2090. [DOI] [PubMed] [Google Scholar]
  • 19.Adamkiewicz T, Mehta P, Boyer M, Kedar A, Olson T, Olson E, et al. Transplantation of unrelated placental blood cells in children with high-risk sickle cell disease. Bone Marrow Transplant. 2004;34(5):405–411. doi: 10.1038/sj.bmt.1704606. [DOI] [PubMed] [Google Scholar]
  • 20.Adamkiewicz T, Szabolcs P, Haight A, Baker K, Staba S, Kedar A, et al. Unrelated cord blood transplantation in children with sickle cell disease: review of four-center experience. Pediatr Transplant. 2007;11(6):641–644. doi: 10.1111/j.1399-3046.2007.00725.x. [DOI] [PubMed] [Google Scholar]
  • 21.Ruggeri A, Eapen M, Scaravadou A, Cairo M, Bhatia M, Kurtzberg J, et al. Umbilical cord blood transplantation for children with thalassemia and sickle cell disease. Biol Blood Marrow Transplant. 2011;17(9):1375–1382. doi: 10.1016/j.bbmt.2011.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jaing T, Sun C, Lee W, Wen Y, Yang C, Hung I. Second transplant with two unrelated cord blood units for early graft failure after cord blood transplantation for thalassemia. Pediatr Transplant. 2009;13(6):766–768. doi: 10.1111/j.1399-3046.2008.01021.x. [DOI] [PubMed] [Google Scholar]
  • 23.Anasetti C, Amos D, Beatty P, Appelbaum F, Bensinger W, Buckner C, et al. Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma. N Engl J Med. 1989;320(4):197–204. doi: 10.1056/NEJM198901263200401. [DOI] [PubMed] [Google Scholar]
  • 24.Kurtzberg J, Prasad V, Carter S, Wagner J, Baxter-Lowe L, Wall D, et al. Results of the Cord Blood Transplantation Study (COBLT): clinical outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with hematologic malignancies. Blood. 2008;112(10):4318–4327. doi: 10.1182/blood-2007-06-098020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gluckman E, Rocha V, Arcese W, Michel G, Sanz G, Chan K, et al. Factors associated with outcomes of unrelated cord blood transplant: guidelines for donor choice. Exp Hematol. 2004;32(4):397–407. doi: 10.1016/j.exphem.2004.01.002. [DOI] [PubMed] [Google Scholar]
  • 26.Kamani N, Spellman S, Hurley C, Barker J, Smith F, Oudshoorn M, et al. State of the art review: HLA matching and outcome of unrelated donor umbilical cord blood transplants. Biol Blood Marrow Transplant. 2008;14(1):1–6. doi: 10.1016/j.bbmt.2007.11.003. [DOI] [PubMed] [Google Scholar]
  • 27.Rocha V, Gluckman E group E-NraEBaMT. Improving outcomes of cord blood transplantation: HLA matching, cell dose and other graft- and transplantation-related factors. Br J Haematol. 2009;147(2):262–274. doi: 10.1111/j.1365-2141.2009.07883.x. [DOI] [PubMed] [Google Scholar]
  • 28.Barker J, Scaradavou A, Stevens C. Combined effect of total nucleated cell dose and HLA match on transplantation outcome in 1061 cord blood recipients with hematologic malignancies. Blood. 2010;115(9):1843–1849. doi: 10.1182/blood-2009-07-231068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fleischhauer K, Locatelli F, Zecca M, Orofino M, Giardini C, De Stefano P, et al. Graft rejection after unrelated donor hematopoietic stem cell transplantation for thalassemia is associated with nonpermissive HLA-DPB1 disparity in host-versus-graft direction. Blood. 2006;107(7):2984–2992. doi: 10.1182/blood-2005-08-3374. [DOI] [PubMed] [Google Scholar]
  • 30.Gale R, Ho W, Feig S, Champlin R, Tesler A, Arenson E, et al. Prevention of graft rejection following bone marrow transplantation. Blood. 1981;57(1):9–12. [PubMed] [Google Scholar]
  • 31.Camitta B, Storb R, Thomas E. Aplastic anemia (first of two parts): pathogenesis, diagnosis, treatment, and prognosis. N Engl J Med. 1982;306(11):645–652. doi: 10.1056/NEJM198203183061105. [DOI] [PubMed] [Google Scholar]
  • 32.Camitta B, Storb R, Thomas E. Aplastic anemia (second of two parts): pathogenesis, diagnosis, treatment, and prognosis. N Engl J Med. 1982;306(12):712–718. doi: 10.1056/NEJM198203253061204. [DOI] [PubMed] [Google Scholar]
  • 33.Kanda J, Ichinohe T, Matsuo K, Benjamin R, Klumpp T, Rozman P, et al. Impact of ABO mismatching on the outcomes of allogeneic related and unrelated blood and marrow stem cell transplantations for hematologic malignancies: IPD-based meta-analysis of cohort studies. Transfusion. 2009;49(4):624–635. doi: 10.1111/j.1537-2995.2008.02043.x. [DOI] [PubMed] [Google Scholar]
  • 34.McPherson M, Anderson A, Castillejo M, Hillyer C, Bray R, Gebel H, et al. HLA alloimmunization is associated with RBC antibodies in multiply transfused patients with sickle cell disease. Pediatr Blood Cancer. 2010;54(4):552–558. doi: 10.1002/pbc.22327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Spellman S, Bray R, Rosen-Bronson S, Haagenson M, Klein J, Flesch S, et al. The detection of donor-directed, HLA-specific alloantibodies in recipients of unrelated hematopoietic cell transplantation is predictive of graft failure. Blood. 2010;115(13):2704–2708. doi: 10.1182/blood-2009-09-244525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Takanashi M, Atsuta Y, Fujiwara K, Kodo H, Kai S, Sato H, et al. The impact of anti-HLA antibodies on unrelated cord blood transplantations. Blood. 2010;116(15):2839–2846. doi: 10.1182/blood-2009-10-249219. [DOI] [PubMed] [Google Scholar]
  • 37.Cutler C, Kim HT, Sun L, Sese D, Glotzbecker B, Armand P, et al. Donor-specific anti-HLA antibodies predict outcome in double umbilical cord blood transplantation. Blood. 2011 Sep 22; doi: 10.1182/blood-2011-05-355263. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.van Rood J, Stevens C, Smits J, Carrier C, Carpenter C, Scaradavou A. Reexposure of cord blood to noninherited maternal HLA antigens improves transplant outcome in hematological malignancies. Proc Natl Acad Sci USA. 2009;106(47):19952–19957. doi: 10.1073/pnas.0910310106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sodani P, Gaziev D, Polchi P, Erer B, Giardini C, Angelucci E, et al. New approach for bone marrow transplantation in patients with class 3 thalassemia aged younger than 17 years. Blood. 2004;104(4):1201–1203. doi: 10.1182/blood-2003-08-2800. [DOI] [PubMed] [Google Scholar]
  • 40.Hongeng S, Pakakasama S, Chuansumrit A, Sirachainan N, Sura T, Ungkanont A, et al. Reduced intensity stem cell transplantation for treatment of class 3 Lucarelli severe thalassemia patients. Am J Hematol. 2007;82(12):1095–1098. doi: 10.1002/ajh.21002. [DOI] [PubMed] [Google Scholar]
  • 41.Barker J, Weisdorf D, DeFor T, Blazar B, McGlave P, Miller J, et al. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005;105(3):1343–1347. doi: 10.1182/blood-2004-07-2717. [DOI] [PubMed] [Google Scholar]
  • 42.Brunstein C, Barker J, Weisdorf D, DeFor T, Miller J, Blazar B, et al. Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease. Blood. 2007;110(8):3064–3070. doi: 10.1182/blood-2007-04-067215. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES