We describe outcomes after human leukocyte antigen-matched sibling bone marrow transplantation (BMT) for 179 patients with β-thalassemia major. The median age at transplantation was 7 years and the median follow-up was 6 years. The distribution of Pesaro risk class I, II, and III categories was 2%, 42%, and 36%, respectively. The day 30 cumulative incidence of neutrophil recovery and day 100 platelet recovery were 90% and 86%, respectively. Seventeen patients had graft failure, which was fatal in 11. Six of 9 patients with graft failure are alive after a second transplantation. The day 100 probability of acute graft-versus-host disease and 5-year probability of chronic graft-versus-host disease was 38% and 13%, respectively. The 5-year probabilities of overall- and disease-free survival were 91% and 88%, respectively, for patients with Pesaro risk class II, and 64% and 62%, respectively, for Pesaro risk class III. In multivariate analysis, mortality risks were higher in patients 7 years of age and older and those with hepatomegaly before BMT. The leading causes of death were interstitial pneumonitis (n = 7), hemorrhage (n = 8), and veno-occlusive disease (n = 6). Proceeding to BMT in children younger than 7 years before development of end-organ damage, particularly in the liver, should improve results after BMT for β-thalassemia major.

β-Thalassemia major, a genetic defect that causes reduced or absent β-globin synthesis, results in an imbalanced accumulation of α-globin chains and ineffective erythropoiesis with hemolysis.1  The hallmarks of treatment include regular red blood cell (RBC) transfusions with iron chelation therapy for transfusion-related iron overload and supportive care to treat the complications of iron overload.2,3  However, the only curative therapy is replacement of the defective gene by hematopoietic cell transplantation.4-6 

Over the past 2 decades, transplantation as a therapeutic option for thalassemia has undergone considerable investigation and refinement. The largest experience occurred in Pesaro, Italy where a standard pretransplantation risk assessment was developed. It divides patients into 3 risk classes based on liver size by physical examination, the presence or absence of fibrosis by liver biopsy, and adherence to regular iron chelation.4-6  In the initial reports, outcomes after transplantation were affected significantly by the pretransplantation risk status. More recently, modifications to the conditioning regimen have been used to reduce the risk of transplantation-related complications in high-risk recipients. As a result, outcomes after transplantation have become more similar across risk categories.7-11  The most recent results after human leukocyte antigen (HLA)-matched sibling bone marrow transplantation (BMT) for Pesaro class I or II and class III recipients show thalassemia-free survival probabilities of 87%, 85%, and 80%, respectively. Despite these improvements, pretransplantation transfusion exposures and organ damage from iron overload still appear to impact transplantation outcomes negatively. In the most recent update, the transplantation-related mortality in class I or II and class III patients was 3% and 10%, respectively, and 8% to 12% of pediatric recipients experienced graft rejection after BMT.6 

Most patient series outside Italy have observed comparatively worse outcomes after HLA-matched sibling BMT for thalassemia major.12-16  Although the reasons for this disparity are unknown, there have been differences in recipient ethnicity, in the ability to assign a pretransplantation risk status, in how patients were prepared for transplantation, and in graft-versus-host disease (GVHD) prophylaxis, which might account for the disparate outcomes. There is also the possibility of a center effect, which might have contributed to an apparent poorer outcome in the smaller patient series outside Italy. To investigate these possibilities, we conducted a survey of β-thalassemia major transplantations recently in locations other than Italy treated by HLA-matched BMT and reported to the Center for International Blood and Marrow Transplant Research.

Data source

Data were obtained from the Center for International Blood and Marrow Transplant Research, a voluntary group of more than 450 transplantation centers worldwide that report data on consecutive transplantations to a Statistical Center at the Medical College of Wisconsin. Patients are followed longitudinally, with yearly follow-up. Computerized checks for errors and on-site audits of participating centers ensure the quality of data. This study included patients (age < 20 years) who had received an HLA-matched sibling BMT for β-thalassemia major between 1995 and 2001 and received myeloablative conditioning (busulfan + cyclophosphamide). During this period, 233 patients met the selection criteria. Thirty-one transplantation centers were contacted for participation/disease-specific information for their patients. Nineteen centers in 12 countries participated (n = 179 patients); 12 centers declined to participate (n = 54 patients). Approximately half of the transplantations were performed in India, 28% in Iran, and 12% in China. The remaining 10% of transplantations occurred in Australia, Belgium, Canada, Denmark, Germany, Israel, South Africa, Spain, and the United States. Information on busulfan administration was available for 111 of 179 patients; all except 2 patients received oral busulfan. In 43% of the cases, the conditioning regimen included antithymocyte globulin (ATG), and 85% of patients received cyclosporine and methotrexate for GVHD prophylaxis. Among survivors, the median follow-up was 73 months (range, 6-127 months). This study was approved by the Institutional Review Board, Medical College of Wisconsin.

Endpoints

Primary endpoints were acute GVHD (grade II-IV), chronic GVHD, and overall survival (OS). Other endpoints studied include hematopoietic recovery and disease-free survival (DFS). Neutrophil recovery was defined as first of 3 consecutive days with absolute neutrophil count more than or equal to 0.5 × 109/L and platelets, more than or equal to 20 × 109/L, unsupported for 7 days. Acute grade II-IV GVHD and chronic GVHD were graded using standard criteria.17,18  DFS was defined as survival without graft failure, a second transplantation, or death. For analyses of OS, failure was defined as death from any cause; surviving patients were censored at the date of last contact.

Statistical analyses

Univariate probabilities of OS and DFS were calculated by the Kaplan-Meier estimator; the log-rank test was used for univariate comparisons. Probabilities of hematopoietic recovery and GVHD were calculated using the cumulative incidence estimator. For hematopoietic recovery and acute or chronic GVHD, death without the event was the competing risk. Estimates of SE for the survival function were calculated by the Greenwood formula, and 95% confidence intervals (CI) were constructed using arcsine-transformed intervals. Potential patient-, disease-, and treatment-related prognostic factors for GVHD and OS were evaluated in multivariate analyses using Cox proportional hazards regression. Multivariate models were built using a stepwise forward selection with a significance level of .05. In each model, the assumption of proportional hazards was tested for each variable. The variables considered in the multivariate model included patient-related factors (age, sex, performance score, and pre-BMT cytomegalovirus serologic status); disease related factors (Pesaro risk class, liver size [ ≤ 2 cm or > 2 cm or unknown], presence or absence of hepatic fibrosis, serum ferritin [ < 2500 ng/mL, ≥ 2500 ng/mL, or unknown], alanine aminotransferase/aspartate aminotransferase [ < 2 times or ≥ 2 times the upper limit of normal or unknown], number of RBC transfusions [ < 20, 20-50, ≥ 50, or unknown]); and transplantation-related factors (donor-recipient gender match and year of transplantation [1995-1997 or 1998-2001]). All P values are 2-sided. All analyses were done with SAS, Version 9.0.

Patients

The demographic data are summarized in Table 1. The median age at BMT was 7 years, and the median follow-up among survivors was 6 years. The transplantation regimens for conditioning and GVHD prophylaxis were fairly uniform. All patients received busulfan and cyclophosphamide for transplantation conditioning; busulfan (16 mg/kg) with cyclophosphamide (200 mg/kg) was the most frequent combination used. Almost all patients (98%) received cyclosporine-containing GVHD prophylaxis. There were significant transfusion exposures before BMT; in 85% of patients, more than 20 RBC units were administered and 62% of patients were transfused more than 50 RBC units before transplantation. Iron chelation therapy was deemed inadequate in 94% of the transplant recipients; inadequate chelation was defined as not initiating chelation therapy within 18 months of the first transfusion and administered subcutaneously for 8 to 10 hours per day at least 5 days per week. Inadequate iron chelation therapy in almost all patients and the presence of hepatomegaly and hepatic fibrosis in approximately 50% of patients imply that most transplant recipients were in a high-risk category with respect to transplantation-related morbidity and mortality. Of the 87% of recipients who had a liver biopsy performed before transplantation, 59% had histologic evidence of portal fibrosis. Although serum bilirubin was normal in 84% of patients, hepatic inflammation was common. The serum aspartate aminotransferase or alanine aminotransferase was greater than 2 times the upper limit of normal in more than 30% of patients. Only 9% of the patients had a documented pretransplantation history of viral hepatitis. Data about hepatic synthetic function were not collected.

Thus, among the 142 (79%) patients who were eligible for risk-group assignment, the assignment to risk categories was 2% to class I, 42% to class II, and 36% to class III. It was not possible to assign a risk category in 20% of the study population. As there were so few patients who had class I features, the findings presented herein pertain to patients with class II and III risk-group features.

Hematopoietic recovery and graft failure

By day 30 after transplantation, the cumulative incidence of neutrophil recovery was 90% (95% CI, 85%-94%) and platelet recovery, 51% (95% CI, 43%-58%). The cumulative incidence of platelet recovery by day 100 improved to 86% (95% CI, 80%-90%). We also examined for the effect of Pesaro risk group and of ATG on neutrophil recovery and did not observe significant differences. The day 30 cumulative incidence of neutrophil recovery for patients in Pesaro class II and class III was 92% (95% CI, 85%-97%) and 91% (95% CI, 83%-96%), respectively (P = .44). Corresponding cumulative incidence of neutrophil recovery for patients who received ATG and did not receive ATG was 89% (95% CI, 82%-94%) and 91% (95% CI, 84%-94%), respectively (P = .13). Seventeen patients (10%) had graft rejection that was fatal in 11 cases. Five of these 17 patients were assigned to Pesaro risk class II, 8 patients risk class III, and in the remaining 4 patients we could not assign a risk category. Five patients with graft rejection received ATG. Six of 9 patients with graft failure are alive and free of thalassemia after a successful second transplantation. The source of hematopoietic cells for the second transplantation was bone marrow in 5 patients, and 4 patients received mobilized peripheral blood stem cells.

GVHD

The day 100 probability of acute GVHD (grade II-IV) was 38% (95% CI, 31%-45%) and the 5-year probability of chronic GVHD was 13% (95% CI, 9%-18%). In multivariate analysis, the administration of more than 50 RBC transfusions before transplantation was associated with a lower risk of acute GVHD (relative risk [RR] = 0.52, 95% CI, 0.31-0.86, P = .010). No risk factors were identified for the development of chronic GVHD. The use of ATG had no apparent association with the risk of GVHD in this analysis. Of those who received ATG before transplantation, 29 of 77 (38%) developed acute GVHD compared with 44 of 99 (44%) in patients who did not receive ATG. There was also no effect of ATG on the incidence of chronic GVHD, in which 10 of 77 (13%) who received ATG and 16 of 99 (16%) who did not receive ATG developed chronic GVHD.

Overall and disease-free survival

A total of 144 patients are alive at last follow-up. In multivariate analysis, the risk of mortality was higher in patients 7 years of age or older compared with patients younger than 7 years (RR = 2.72, 95% CI, 1.29-5.70; 69% vs 90%, respectively, P = .008) and in those with hepatomegaly defined as more than 2 cm below the coastal margin at BMT compared with those without hepatomegaly (RR = 5.17, 95% CI, 1.98-13.47; 66% vs 94%, respectively, P < .001). Age at transplantation and liver size are independent predictors of mortality after transplantation. The probabilities of OS and DFS are shown in Figures 1 and 2. In patients younger than 7 years and without hepatomegaly, the 5-year probabilities of OS and DFS were 98% (95% CI, 92%-100%) and 94% (95% CI, 85%-99%), respectively. The 5-year probabilities of OS and DFS in older patients (age ≥ 7 years) without hepatomegaly were 86% (95% CI, 72%-96%) and 83% (95% CI, 67%-94%), respectively.

Figure 1

Probability of OS by age and hepatomegaly.

Figure 1

Probability of OS by age and hepatomegaly.

Close modal
Figure 2

Probability of DFS by age and hepatomegaly.

Figure 2

Probability of DFS by age and hepatomegaly.

Close modal

Pesaro risk class had a significant association with survival in univariate analysis. The 5-year overall survival was 91% (95% CI, 83%–96%) and 64% (95% CI, 52%–75%) in patients with risk class II and class III, respectively (P = .0002; Figure 3). Pesaro risk class did not enter the final multivariate model because it is highly correlated with liver size; hepatomegaly entered the stepwise model-building process early and was retained in the final multivariate model. Given the relatively small numbers of patients in each risk group, it is difficult to examine the effect of liver size in patients assigned to risk group II. In subset analysis, mortality risks were higher in patients assigned to risk group II with hepatomegaly compared with those assigned risk group II without hepatomegaly; however, this difference was not statistically significant (RR = 3.63, 95% CI, 0.91-14.57, P = .07). Only 3 patients who were 4, 12, and 16 years of age were assigned to risk class I; one patient died and the 2 remaining patients were alive at last follow-up.

Figure 3

Probability of OS by Pesaro risk class.

Figure 3

Probability of OS by Pesaro risk class.

Close modal

Thirty-five patients died, and their primary causes of death are shown in Tables 2 and 3. Most deaths (n = 25) occurred within the first 3 months after BMT. Veno-occlusive disease of the liver (VOD) occurred frequently after transplantation, particularly in patients with high-risk disease. Fifty-eight patients (32%) developed VOD; of these, 2 patients had risk class I, 22 patients had risk class II, and 33 patients had risk class III features. Approximately one-third of patients with risk class II and 50% of patients with risk class III features developed VOD. VOD occurred more frequently in patients with a serum ferritin more than or equal to 2500 μg/L compared with those with a serum ferritin less than 2500 μg/L (52% and 32%, respectively). Of the 58 patients who developed VOD, 21 patients are dead (n = 6 reported VOD as the primary cause of death and n = 3 reported VOD as a contributing cause of death).

In this report, we have attempted to capture most of the most recent transplantation experience outside Italy that included children with β-thalassemia major who were treated with HLA-matched sibling BMT. Thus, this represents the largest contemporary series yet reported and generates new information about assigning transplantation risk, which should assist in optimizing outcomes and selecting suitable candidates for transplantation. These results must be interpreted while also considering recent advances in supportive therapy, particularly in iron chelation therapy, which have reduced the risks of morbidity and mortality in those who do not proceed to transplantation.2,16,19-21  Our results confirm that HLA-matched sibling BMT for β-thalassemia major is a suitable therapeutic option to consider, as we also observed DFS rates that exceeded 90% in children with good-risk features (Figure 1).4,9  However, as noted in the initial Pesaro experience, children with high-risk features fared much worse, with an event-free survival rate that approached 50%.4,22,23  These observations underscore the importance of carefully selecting good-risk patients for transplantation, but also if treating high-risk patients, the importance of using modifications to the transplantation regimen that might improve the safety of the procedure.6,7,11,24 

We were able to identify 2 readily accessible prognostic indicators in a cohort of high-risk thalassemia recipients, namely, liver size and patient age, which other groups also have identified.19,20  The observed effects of age and liver size were independent of each other, although liver size is highly correlated with Pesaro risk group. By classifying this high-risk cohort by these criteria (ie, age and liver size), it was possible to refine and broaden the class II and III Pesaro risk groupings into distinct patterns of survival and event-free survival, which are represented in Figures 1 and 2. The 5-year survival probabilities, which ranged from 53% to 96%, are broader than the initial Pesaro experience.4,9,20  Although these criteria do not supplant the utility of a liver biopsy, which provides important information about liver histology and can also direct post-transplantation interventions to reduce iron overload, there may be situations where a liver biopsy and the ability to assess liver histology and iron concentration are not readily available. Using the risk features identified in the current analysis, it may be possible to assign a conditioning therapy that is better suited to those who are most in need of such modifications and in clinical settings where assigning a Pesaro risk classification is not possible.7,12,22  We speculate that, for the diverse cohort in the current analysis, it may be appropriate to dispense with the Pesaro classification in high-risk patients and rely on this facile and novel risk assignment.

It appears that much of the toxicity of transplantation observed in this series might have been related to impaired hepatic function and a propensity for developing interstitial pneumonitis and VOD, particularly in the older, high-risk patients. Previously, a high rate of regimen-related toxicity and graft rejection was noted in class II and III patients after standard busulfan and cyclophosphamide preparation, particularly in young adult patients with thalassemia, for whom the transplantation-related mortality was 35%.25  An early study of busulfan pharmacokinetics as a predictor of clinical outcome in thalassemia major failed to identify a relationship between the busulfan level and toxicity, although this analysis included transplantation regimens that differed in their cyclophosphamide dosing and in postgrafting immunosuppression.26  Other studies in children with hematologic malignancies have demonstrated the utility of busulfan pharmacokinetics in predicting the risk of toxicity and have generally endorsed the adoption of targeted busulfan dosing to avoid this toxicity.27-29  Moreover, a more recent clinical study defined the important contribution by cyclophosphamide and its metabolites to the toxicity of the busulfan and cyclophosphamide regimen, suggesting that the administration of busulfan before cyclophosphamide alters cyclophosphamide pharmacokinetics and thus might also exacerbate the risk of VOD in patients with underlying hepatic injury.30,31  To mitigate these risks, investigative teams in Italy have reduced the dosing of cyclophosphamide in high-risk patients.7  Alternatively, the replacement of cyclophosphamide with fludarabine might also mitigate the risk of significant transplantation-related toxicity.32  Intravenous busulfan in lieu of oral busulfan also appears to improve the safety profile of busulfan as does the practice of therapeutic drug monitoring.11,33-35  The observations made in this series thus highlight the importance of modifying the standard busulfan and cyclophosphamide regimen in very high-risk patients to avoid unacceptably high transplantation-related mortality.

In other patient series, graft rejection after HLA-matched sibling BMT is generally accompanied by thalassemia recurrence and recovery of autologous hematopoietic function.5,36,37  However, in this series graft failure, which occurred in nearly 10% of cases, was fatal in 11 cases and required rescue by second transplantation to restore normal hematopoiesis. The reasons for this are unclear. The use of ATG did not appear to be useful in reducing the risk of graft failure. In the future, it is possible that the development of alternative regimens that provide more intensive immune suppression before transplantation without myeloablation might reduce the elevated risk of graft failure we observed in high-risk patients with thalassemia.24,38 

When defining the role of transplantation for β-thalassemia major, which is a chronic, nonmalignant condition, one must also consider the natural history of the disorder and the impact of improved supportive care measures on patient outcomes. In selected regions of the world, the outcomes are outstanding when there is ready access to expanded supportive care services, with very few persons dying of thalassemia or of complications related to its treatment through the third and fourth decades of life.12,20,21  In other areas of the world, however, supportive care measures are often difficult to access, which as predicted, has had a negative impact on survival.39,40  We speculate that many of the transplant recipients included in this series lacked ready access to vital supportive care measures, such as regular iron chelation therapy, and there may have been delays in proceeding to transplantation in cases where resources were limited. For these reasons, the results shown here reflect a variety of healthcare issues that have the potential to affect outcomes in children with β-thalassemia who reside outside North America and Europe. Considering the impact of these mitigating factors, we think that the transplantation outcomes reported should motivate families and their physicians to strongly consider the possibility of HLA-matched sibling BMT earlier in the course of their disease, particularly when there are limited resources for chronic thalassemia-centered medical care.

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

The Center for International Blood and Marrow Transplant Research is supported by the National Cancer Institute, the National Heart, Lung and Blood Institute, and the National Institute of Allergy and Infectious Diseases (Public Health Service Grant/Cooperative Agreement U24-CA76518); National Cancer Institute, the National Heart, Lung and Blood Institute (Grant/Cooperative Agreement 5U01HL069294); Health Resources and Services Administration (contract HHSH234200637015C); the Office of Naval Research (grants N00014–06-1–0704 and N00014–08-1–0058); and grants from the following: AABB; Allos, Inc; Amgen, Inc; anonymous donation to the Medical College of Wisconsin; Astellas Pharma US, Inc; Be the Match Foundation; Biogen IDEC; BioMarin Pharmaceutical, Inc; Biovitrum AB; BloodCenter of Wisconsin; Blue Cross and Blue Shield Association; Bone Marrow Foundation; Buchanan Family Foundation; CaridianBCT; Celgene Corporation; CellGenix, GmbH; ClinImmune Labs; CTI Clinical Trial and Consulting Services; Eisai, Inc; Genentech, Inc; Genzyme Corporation; Histogenetics, Inc; HKS Medical Information Systems; Hospira, Inc; Kirin Brewery Co, Ltd; Merck & Company; Medical College of Wisconsin; Millennium Pharmaceuticals, Inc; Miller Pharmacal Group; Milliman USA, Inc; Miltenyi Biotec, Inc; National Marrow Donor Program; Nature Publishing Group; Novartis Oncology; Oncology Nursing Society; Osiris Therapeutics, Inc; Otsuka America Pharmaceutical, Inc; Pall Life Sciences; Pfizer Inc; Schering Corporation; Sigma-Tau Pharmaceuticals; Soligenix, Inc; StemCyte, Inc; StemSoft Software, Inc; Sysmex America, Inc; THERAKOS, Inc; Vidacare Corporation; ViraCor Laboratories; ViroPharma, Inc; and Wellpoint, Inc.

The views expressed in this article do not reflect the official policy or position of the National Institute of Health, the Department of the Navy, the Department of Defense, or any other agency of the U.S. Government.

National Institutes of Health

Contribution: M.S., B.R.L., C.N.B., M.E., J.H., M.C.W., G.A.H., R.P.G., and S.M.I. participated in conception and design of the study; M.C., S.H., A.G., C.-K.L., and M.J.C. contributed patients; Z.W. and B.R.L. assembled the data and performed the statistical analysis; M.S. and M.C.W. had primary responsibility for manuscript preparation; and all authors participated in interpretation of data, manuscript preparation, and approval of the final manuscript.

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

Correspondence: Mark C. Walters, Children's Hospital & Research Center, Oakland, Pediatric Hematology/Oncology, 747 52nd St, Oakland, CA, 94609; e-mail: mwalters@mail.cho.org.

1
Rund
 
D
Rachmilewitz
 
E
Beta-thalassemia.
N Engl J Med
2005
, vol. 
353
 
11
(pg. 
1135
-
1146
)
2
Borgna-Pignatti
 
C
Rugolotto
 
S
De Stefano
 
P
, et al. 
Survival and complications in patients with thalassemia major treated with transfusion and deferoxamine.
Haematologica
2004
, vol. 
89
 
10
(pg. 
1187
-
1193
)
3
Morris
 
CR
Singer
 
ST
Walters
 
MC
Clinical hemoglobinopathies: iron, lungs and new blood.
Curr Opin Hematol
2006
, vol. 
13
 
6
(pg. 
407
-
418
)
4
Lucarelli
 
G
Galimberti
 
M
Polchi
 
P
, et al. 
Bone marrow transplantation in patients with thalassemia.
N Engl J Med
1990
, vol. 
322
 
7
(pg. 
417
-
421
)
5
Angelucci
 
E
Baronciani
 
D
Allogeneic stem cell transplantation for thalassemia major.
Haematologica
2008
, vol. 
93
 
12
(pg. 
1780
-
1784
)
6
Gaziev
 
J
Sodani
 
P
Lucarelli
 
G
Hematopoietic stem cell transplantation in thalassemia.
Bone Marrow Transplant
2008
, vol. 
42
 
suppl 1
pg. 
S41
 
7
Sodani
 
P
Gaziev
 
D
Polchi
 
P
, et al. 
New approach for bone marrow transplantation in patients with class 3 thalassemia aged younger than 17 years.
Blood
2004
, vol. 
104
 
4
(pg. 
1201
-
1203
)
8
Lucarelli
 
G
Galimberti
 
M
Giardini
 
C
, et al. 
Bone marrow transplantation in thalassemia: the experience of Pesaro.
Ann N Y Acad Sci
1998
, vol. 
850
 (pg. 
270
-
275
)
9
Lucarelli
 
G
Galimberti
 
M
Polchi
 
P
, et al. 
Marrow transplantation in patients with thalassemia responsive to iron chelation therapy.
N Engl J Med
1993
, vol. 
329
 
12
(pg. 
840
-
844
)
10
Giardini
 
C
Galimberti
 
M
Lucarelli
 
G
Polchi
 
P
Baronciani
 
D
Angelucci
 
E
Bone marrow transplantation in class 2 thalassemia patients.
Bone Marrow Transplant
1993
, vol. 
12
 
suppl 1
(pg. 
59
-
62
)
11
Chandy
 
M
Balasubramanian
 
P
Ramachandran
 
SV
, et al. 
Randomized trial of two different conditioning regimens for bone marrow transplantation in thalassemia: the role of busulfan pharmacokinetics in determining outcome.
Bone Marrow Transplant
2005
, vol. 
36
 
10
(pg. 
839
-
845
)
12
Ullah
 
K
Khan
 
B
Raza
 
S
, et al. 
Bone marrow transplant cure for beta-thalassaemia major: initial experience from a developing country.
Ann Hematol
2008
, vol. 
87
 
8
(pg. 
655
-
661
)
13
Boulad
 
F
Giardina
 
P
Gillio
 
A
, et al. 
Bone marrow transplantation for homozygous beta-thalassemia: the Memorial Sloan-Kettering Cancer Center experience.
Ann N Y Acad Sci
1998
, vol. 
850
 (pg. 
498
-
502
)
14
Walters
 
MC
Sullivan
 
KM
O'Reilly
 
RJ
, et al. 
Bone marrow transplantation for thalassemia: the USA experience.
Am J Pediatr Hematol Oncol
1994
, vol. 
16
 
1
(pg. 
11
-
17
)
15
Chern
 
JP
Su
 
S
Lin
 
KH
, et al. 
Survival, mortality, and complications in patients with beta-thalassemia major in northern Taiwan.
Pediatr Blood Cancer
2007
, vol. 
48
 
5
(pg. 
550
-
554
)
16
Cappellini
 
MD
Cohen
 
A
Piga
 
A
, et al. 
A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with beta-thalassemia.
Blood
2006
, vol. 
107
 
9
(pg. 
3455
-
3462
)
17
Rowlings
 
PA
Przepiorka
 
D
Klein
 
JP
, et al. 
IBMTR Severity Index for grading acute graft-versus-host disease: retrospective comparison with Glucksberg grade.
Br J Haematol
1997
, vol. 
97
 
4
(pg. 
855
-
864
)
18
Flowers
 
ME
Kansu
 
E
Sullivan
 
KM
Pathophysiology and treatment of graft-versus-host disease.
Hematol Oncol Clin North Am
1999
, vol. 
13
 
5
(pg. 
1091
-
1112
)
19
Borgna-Pignatti
 
C
Cappellini
 
MD
De Stefano
 
P
, et al. 
Cardiac morbidity and mortality in deferoxamine- or deferiprone-treated patients with thalassemia major.
Blood
2006
, vol. 
107
 
9
(pg. 
3733
-
3737
)
20
Maggio
 
A
Vitrano
 
A
Capra
 
M
, et al. 
Improving survival with deferiprone treatment in patients with thalassemia major: a prospective multicenter randomised clinical trial under the auspices of the Italian Society for Thalassemia and Hemoglobinopathies.
Blood Cells Mol Dis
2009
, vol. 
42
 
3
(pg. 
247
-
251
)
21
Borgna-Pignatti
 
C
Cappellini
 
MD
De Stefano
 
P
, et al. 
Survival and complications in thalassemia.
Ann N Y Acad Sci
2005
, vol. 
1054
 (pg. 
40
-
47
)
22
Mathews
 
V
George
 
B
Deotare
 
U
, et al. 
A new stratification strategy that identifies a subset of class III patients with an adverse prognosis among children with beta thalassemia major undergoing a matched related allogeneic stem cell transplantation.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
8
(pg. 
889
-
894
)
23
Lucarelli
 
G
Clift
 
RA
Galimberti
 
M
, et al. 
Marrow transplantation for patients with thalassemia: results in class 3 patients.
Blood
1996
, vol. 
87
 
5
(pg. 
2082
-
2088
)
24
Hongeng
 
S
Pakakasama
 
S
Chuansumrit
 
A
, et al. 
Reduced intensity stem cell transplantation for treatment of class 3 Lucarelli severe thalassemia patients.
Am J Hematol
2007
, vol. 
82
 
12
(pg. 
1095
-
1098
)
25
Lucarelli
 
G
Clift
 
RA
Galimberti
 
M
, et al. 
Bone marrow transplantation in adult thalassemic patients.
Blood
1999
, vol. 
93
 
4
(pg. 
1164
-
1167
)
26
Pawlowska
 
AB
Blazar
 
BR
Angelucci
 
E
Baronciani
 
D
Shu
 
XO
Bostrom
 
B
Relationship of plasma pharmacokinetics of high-dose oral busulfan to the outcome of allogeneic bone marrow transplantation in children with thalassemia.
Bone Marrow Transplant
1997
, vol. 
20
 
11
(pg. 
915
-
920
)
27
Bornhauser
 
M
Storer
 
B
Slattery
 
JT
, et al. 
Conditioning with fludarabine and targeted busulfan for transplantation of allogeneic hematopoietic stem cells.
Blood
2003
, vol. 
102
 
3
(pg. 
820
-
826
)
28
Chiesa
 
R
Cappelli
 
B
Crocchiolo
 
R
, et al. 
Unpredictability of intravenous busulfan pharmacokinetics in children undergoing hematopoietic stem cell transplantation for advanced beta thalassemia: limited toxicity with a dose-adjustment policy.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
5
(pg. 
622
-
628
)
29
Grochow
 
LB
Jones
 
RJ
Brundrett
 
RB
, et al. 
Pharmacokinetics of busulfan: correlation with veno-occlusive disease in patients undergoing bone marrow transplantation.
Cancer Chemother Pharmacol
1989
, vol. 
25
 
1
(pg. 
55
-
61
)
30
McCune
 
JS
Batchelder
 
A
Deeg
 
HJ
, et al. 
Cyclophosphamide following targeted oral busulfan as conditioning for hematopoietic cell transplantation: pharmacokinetics, liver toxicity, and mortality.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
7
(pg. 
853
-
862
)
31
McDonald
 
GB
Slattery
 
JT
Bouvier
 
ME
, et al. 
Cyclophosphamide metabolism, liver toxicity, and mortality following hematopoietic stem cell transplantation.
Blood
2003
, vol. 
101
 
5
(pg. 
2043
-
2048
)
32
Russell
 
JA
Tran
 
HT
Quinlan
 
D
, et al. 
Once-daily intravenous busulfan given with fludarabine as conditioning for allogeneic stem cell transplantation: study of pharmacokinetics and early clinical outcomes.
Biol Blood Marrow Transplant
2002
, vol. 
8
 
9
(pg. 
468
-
476
)
33
Gaziev
 
J
Nguyen
 
L
Puozzo
 
C
, et al. 
Novel pharmacokinetic behavior of intravenous busulfan in children with thalassemia undergoing hematopoietic stem cell transplantation: a prospective evaluation of pharmacokinetic and pharmacodynamic profile with therapeutic drug monitoring.
Blood
2010
, vol. 
115
 
22
(pg. 
4597
-
4604
)
34
Cappelli
 
B
Chiesa
 
R
Evangelio
 
C
, et al. 
Absence of VOD in paediatric thalassaemic HSCT recipients using defibrotide prophylaxis and intravenous busulphan.
Br J Haematol
2009
, vol. 
147
 
4
(pg. 
554
-
560
)
35
de Lima
 
M
Couriel
 
D
Thall
 
PF
, et al. 
Once-daily intravenous busulfan and fludarabine: clinical and pharmacokinetic results of a myeloablative, reduced-toxicity conditioning regimen for allogeneic stem cell transplantation in AML and MDS.
Blood
2004
, vol. 
104
 
3
(pg. 
857
-
864
)
36
Gaziev
 
J
Sodani
 
P
Lucarelli
 
G
, et al. 
Second hematopoietic SCT in patients with thalassemia recurrence following rejection of the first graft.
Bone Marrow Transplant
2008
, vol. 
42
 
6
(pg. 
397
-
404
)
37
Stepensky
 
P
Or
 
R
Shapira
 
MY
Revel-Vilk
 
S
Stein
 
J
Resnick
 
IB
Second bone marrow transplantation for patients with thalassemia: risks and benefits.
Haematologica
2009
, vol. 
94
 
9
(pg. 
1329
-
1330
)
38
Shenoy
 
S
Grossman
 
WJ
DiPersio
 
J
, et al. 
A novel reduced-intensity stem cell transplant regimen for nonmalignant disorders.
Bone Marrow Transplant
2005
, vol. 
35
 
4
(pg. 
345
-
352
)
39
Karimi
 
M
Jamalian
 
N
Sociocultural challenges of thalassemia birth in Islamic and developing countries.
J Pediatr Hematol Oncol
2008
, vol. 
30
 
4
pg. 
335
 
40
Weatherall
 
DJ
Keynote address: the challenge of thalassemia for the developing countries.
Ann N Y Acad Sci
2005
, vol. 
1054
 (pg. 
11
-
17
)
Sign in via your Institution