Matched-related bone marrow transplantation (BMT) may cure >80% of low-risk children with severe thalassemia (ST). Very long-term follow-up studies have shown how the standard busulfan-cyclophosphamide (BuCy) regimen may be associated with normalization of health-related quality of life, no second malignancies in the absence of chronic graft-versus-host disease, and fertility preservation in many patients. However, because BuCy may be associated with high rejection rates, some centers incorporate thiotepa (Tt) in busulfan- or treosulfan-based regimens, a combination that may increase the risk of permanent infertility. This study retrospectively compares matched-related BMT outcomes in 2 groups of low-risk ST patients conditioned with either Tt or anti–thymocyte globulin (ATG) in addition to BuCy. A total of 81 consecutive first BMTs were performed in 5 collaborating startup BMT centers in the Indian subcontinent between January 2009 and January 2016; 30 patients were transplanted after conditioning with Tt-BuCy between January 2009 and July 2013, whereas between August 2013 and January 2016, 51 patients received ATG-BuCy. All patients were <15 years and had no hepatomegaly (liver ≤2 cm from costal margin). Actuarial overall survival in the Tt-BuCy and ATG-BuCy groups was 87% and 94% and thalassemia-free survival was 80% and 85% at a median follow-up of 37 and 17 months, respectively, with no significant differences by log-rank statistics. Substituting Tt with ATG in the standard BuCy context seems safe and effective and may decrease transplant-related mortality. Higher fertility rates are expected for patients who received ATG-BuCy.

Severe thalassemia (ST) syndromes are the most prevalent life-threatening noncommunicable disorder of childhood1  in Southeast Asia and a major financial burden.2-4  In spite of improved iron chelation therapy and safer transfusions,5  ST remains a severe disease, particularly in developing countries, where often state-of-the-art care is not affordable and/or accessible. ST is also a major indication for blood and marrow transplantation (BMT) in children in the Indian subcontinent.6-8  BMT is currently the only definitive curative option with success rates >80%9  and potential normalization of health-related quality of life (HRQoL)10  in low-risk children with a compatible related donor. The cost of BMT is equivalent to that of a few years of noncurative supportive care.3,11,12  The standard combination of busulfan and cyclophosphamide (BuCy) is associated with rejection rates ranging from 12% to 35%,13,14  which prompted the addition of other engraftment-enhancing agents such as thiotepa (Tt)15,16  or anti–thymocyte globulin (ATG)13,17-19  to busulfan- or treosulfan-containing regimens. Very long-term follow-up studies have shown that BuCy may be associated with normalization of HRQoL, no second malignancies in the absence of chronic graft-versus-host disease (cGVHD), and preservation of fertility in a significant number of patients transplanted prior to age 15 years.10  The combination of 2 myeloablative alkylating agents such as Tt and busulfan or treosulfan may result in a very high infertility rate,20,21  which is increasingly unacceptable in the new thalassemia era of effective chelation, where a substantial proportion of individuals with ST may bear children.22 

This study compares retrospectively 2 standard preparative regimens based on BuCy in addition to either Tt or ATG for matched-related low-risk BMTs for ST. Moreover, all BMTs were performed in homogeneous lower-risk patients directly in startup BMT centers in lower middle income countries (LMICs)23  where this disease is most prevalent.

Study design and procedures

This analysis retrospectively compared 2 widely used preparative drug combinations, Tt-BuCy and ATG-BuCy, in a homogeneous group of low-risk patients with ST directly in LMICs. All BMTs were carried out in 5 newly developed BMT units in the Indian subcontinent: The Children’s Hospital of the Pakistan Institute of Medical Sciences (CHPIMS) in Islamabad, Pakistan; the South East Asia Institute for Thalassemia in (SEAIT) in Jaipur, India; the Asiri Central Hospital (ACH) in Colombo, Sri Lanka; the Nawaloka Hospital (NH) in Colombo, Sri Lanka; and People Tree Hospital (PTH) in Bangalore, India. Institutional partnerships and collaboration methodology are summarized elsewere.24  The great majority of patients included in this report were fully supported by organizations mentioned in the Acknowledgments, and patients were purely selected on medical grounds rather than funding availability. Once enrolled in the program, BMT was free of cost as well as family support.

An information technology platform facilitating interaction among BMT professionals and promoting continuing quality improvement was employed by each participating center, and data were entered prospectively on a daily basis by local physicians and nurses (BMTPlus; Jagrity Innovations, Bangalore, India).25  Computer-generated (Microsoft Excel) individualized treatment plans developed according to good clinical practices and providing clear and simple operating instructions to point-of-care professionals were generated for each patient and double checked by BMT consultants. Written informed consent was obtained from parents or donors >18 years, including sharing data on the online database, the use of which was approved by institutional review boards at each participating center.

All patients and their caretakers were hosted in single rooms with split air conditioning but no centralized HEPA filtration with positive pressure gradients; strict hand washing and sanitation of all personnel and visitors was enforced, and BMT units were thoroughly cleaned daily by dedicated personnel.

Neutrophil recovery was defined as the first of three consecutive days with an absolute neutrophil count (ANC) >500/µL and platelet recovery as the first day of a stable platelet count >20 000/µL without platelet transfusions in the preceding week. Toxicity was graded according to Eastern Cooperative Oncology Group (ECOG) common toxicity criteria.26  Hypertension was categorized using standard criteria.27  Sinusoidal obstructive syndrome was defined and graded according to the Settle criteria,28  acute graft-versus-host disease (aGVHD) according to standard criteria,29  and cGVHD according to National Institutes of Health guidelines.30  Mixed chimerism was categorized in 3 levels according to percentage of donor cells (level 1, 90% to 99%; level 2, 75% to 89%; and level 3, 5% to 74%).31 

Patient characteristics and transplant procedure

Inclusion criteria were (a) a diagnosis of severe thalassemia defined as a thalassemia syndrome with the inability to spontaneously maintain hemoglobin levels ≥7 g/dL; (b) liver ≤2 cm below the costal margin on abdominal palpation; (c) availability of a suitable HLA genoidentical related donor; (d) clear understanding of BMT risks and benefits, including parental/care taker informed consent for transplantation; (e) patients <15 years, with no active severe infectious diseases (HIV, hepatitis B, tuberculosis, malaria) or other conditions affecting transplant outcome; (f) creatinine, bilirubin, and serum glutamic-pyruvic transaminase (SGPT) less than three times normal values (SGPT up to 10 times normal values was accepted in case of hepatitis C virus positivity), normal chest radiograph and echocardiogram (shortening fraction ≥35%), and normal age-appropriate performance scale; and (g) institutional commitment to sharing patient data according to privacy regulations.

A total of 81 consecutive patients received first BMT from January 2009 to July 2016, 30 patients were transplanted after conditioning with Tt, busulfan (oral), and cyclophosphamide (Tt-BuCy) (Figure 1A) between January 2009 and July 2013, whereas between August 2013 and January 2016, 51 patients were conditioned with ATG, busulfan (oral), and cyclophosphamide (ATG-BuCy) (Figure 1B). Single-lumen central venous catheters, which were tunneled in 49 patients and nontunneled in 31 patients, were employed; 1 patient (who was hepatitis C positive) was managed with peripheral vein cannulas throughout BMT. Tt-BuCy consisted of oral busulfan 3.5 mg/kg day in 4 divided doses on days −10 to −7 (total dose 14 mg/kg), Tt 10 mg/kg per day in 2 divided doses on day −6 (total dose 10 mg/kg), and cyclophosphamide 50 mg/kg per day once daily on days −5 to −2 (total dose 200 mg/kg) followed by the infusion of freshly harvested HLA-compatible marrow on day 0. ATG-BuCy consisted of ATG (rabbit thymoglobulin) 4 mg/kg total dose on days −12 to −10 (at CHPIMS, rabbit-Fresenius was used at 16 mg/kg total dose, because thymoglobulin was not available), busulfan and cyclophosphamide were given as in Tt-BuCy, except busulfan administration was started 1 day later (days −9 to −6). The busulfan dose was not adjusted to blood levels. On day 0, both groups received exclusively bone marrow, which was unprimed in the Tt-BuCy group but granulocyte colony-stimulating factor (G-CSF) primed in 65% of the ATG-BuCy group by treating the donor with filgrastim 5 μg/kg per dose twice daily for either 3 (19 patients) or 5 days (14 patients) prior to harvest. After both regimens, GVHD prophylaxis consisted of cyclosporin A in 2 daily doses aiming at trough levels of 150 to 250 ng/mL for up to day +180, after which it was tapered by −5%/week and discontinued at 12 months post-BMT unless otherwise indicated. A “short” methotrexate course consisting of 10 mg/m2 IV on day +1 (24 hours after marrow infusion) and 8 mg/m2 IV +3, +6, and +11 (the latter dose only in the absence of severe mucositis) with folinic acid rescue at 24 hours after each methotrexate with 3 doses of 10 mg/m2 IV at 8-hour intervals. Methylprednisolone at 0.5 mg/kg per day IV from day −1 to +30, tapered by −1/3 every 5 days over 15 days and stopped on day +45, was also used. Erythrocyte depletion from marrow harvest was required only in cases of major ABO mismatches with isohemagglutinin titers >1:32. All patients received mebendazole 100 mg twice daily for 3 days before conditioning for antihelminthic empirical therapy regardless of stool examination result. No antibacterial prophylaxis was employed. Antifungal prophylaxis consisted of fluconazole 6 mg/kg once daily from day +4 until ANC >500/µL. Acyclovir 250-500 mg/m2 per dose 3 times daily from day +1 to +90 was used for herpes varicella-zoster and simplex prophylaxis. Peripheral blood weekly cytomegalovirus (CMV) DNA copy was monitored by qualitative polymerase chain reaction from day +30 to day +100 in uncomplicated cases; qualitative real-time polymerase chain reaction was employed if the former result was positive, and ganciclovir pre-emptive therapy was administered for DNA copies >1000/mL. For Pneumocystis jiroveci prophylaxis, cotrimoxazole at 5 mg/kg per dose twice daily on alternate days 3 times a week was administered from the day the total leukocyte count reached >1000/µL to day +100.

Figure 1.

Conditioning regimens. (A) Tt-BuCy. (B) ATG-BuCy. CSA, cyclosporin A; MTX, methotrexate; PDN, prednisone.

Figure 1.

Conditioning regimens. (A) Tt-BuCy. (B) ATG-BuCy. CSA, cyclosporin A; MTX, methotrexate; PDN, prednisone.

Close modal

Post-BMT, all blood products except for the allograft were irradiated at ≥25 Gy. Immunoglobulin prophylaxis was not employed. No autologous backup marrow was collected. Engraftment was monitored at least at 1, 2, 4, and 8 months by molecular (short tandem repeats) analysis, Y chromosome cytogenetics, or fluorescent in situ hybridization in case of sex mismatch. Table 1 summarizes patient distribution by center and treatment regimen, and Table 2 summarizes patient characteristics by treatment regimen. All patients had at least 6 months of follow-up post-BMT. None of these patients regularly received leukodepleted erythrocyte transfusions prior to BMT.

Table 1.

Patient distribution by center

CenterDate of first BMTTotal BMTsTt-Bu-CyATG-Bu-Cy
CHPIMS January 2009 41 21 19 
SEAIT February 2012 28 19 
ACH June 2014 
NHC June 2015 
PTH July 2015 
CenterDate of first BMTTotal BMTsTt-Bu-CyATG-Bu-Cy
CHPIMS January 2009 41 21 19 
SEAIT February 2012 28 19 
ACH June 2014 
NHC June 2015 
PTH July 2015 
Table 2.

Patient characteristics

Tt-BuCyATG-BuCyP
No. of patients 30 51  
Follow up, mo* 36.8 (17.3-80.5) 16.9 (7.4-30.7) <.001 
Age, y* 3.1 (0.9-6.4) 4.8 (1.4-14.5) .002 
Sex ratio, male/female 23/7 31/20 .222 
Height-for-age z-score* −1.085 (−2.90 to 0.93) −1.380 (−3.08 to 1.33) .328 
Body mass index-for-age z-score* −0.29 (−2.85 to 1.58) −0.85 (−3.88 to 1.32) .110 
Consanguinity 13 of 27 (3 NA) (48%) 25 of 46 (5 NA) (54%) .636 
No. of previous transfusions 30 (6-128) 42 (3-338) .026 
Donor age 7.1 (1.5-22.3) 6.8 (1.1-37.4) .907 
Matched-related donor type Sibling in 29 of 30 (1 first-degree cousin) Sibling in 50 of 51 (1 mother) 1.000 
No sibling sex mismatch 12 of 29 (41%) 21 of 50 (42%) 1.000 
Sister donor to brother 13 of 29 (45%) 18 of 50 (36%) .480 
Brother donor to sister 4 of 29 (14%) 11 of 50 (22%) .553 
Donor thalassemia minor 23 of 30 (77%) 37 of 51 (73%) .796 
Major ABO mismatch 7 of 30 (23%) 8 of 51 (16%) .394 
Ferritin at BMT, ng/mL* 2282 (591-8008) 1920 (376 to 6171) .354 
Hydroxyurea therapy prior to BMT 1 of 30 (3%) 21 of 51 (41%) <.001 
Liver size, cm from costal margin all ≤2 cm all ≤2 cm  
Spleen size, cm from costal margin 28 ≤ 2 cm, 1 = 3 cm, 1 NA 49 ≤ 2 cm, 1 = 3 cm, 1 = 4 cm  
Splenectomy 1 (3%)  
SGPT, IU/L (HCV-negative patients)* 49.5 (12-149) 32 (11 to 151) .008 
HCV positive 7 of 30 (23%) 5 of 51 (10%) .108 
SGPT, IU/L (HCV-positive patients)* 161 (23-537) 90 (35 to 118) .268 
Tt-BuCyATG-BuCyP
No. of patients 30 51  
Follow up, mo* 36.8 (17.3-80.5) 16.9 (7.4-30.7) <.001 
Age, y* 3.1 (0.9-6.4) 4.8 (1.4-14.5) .002 
Sex ratio, male/female 23/7 31/20 .222 
Height-for-age z-score* −1.085 (−2.90 to 0.93) −1.380 (−3.08 to 1.33) .328 
Body mass index-for-age z-score* −0.29 (−2.85 to 1.58) −0.85 (−3.88 to 1.32) .110 
Consanguinity 13 of 27 (3 NA) (48%) 25 of 46 (5 NA) (54%) .636 
No. of previous transfusions 30 (6-128) 42 (3-338) .026 
Donor age 7.1 (1.5-22.3) 6.8 (1.1-37.4) .907 
Matched-related donor type Sibling in 29 of 30 (1 first-degree cousin) Sibling in 50 of 51 (1 mother) 1.000 
No sibling sex mismatch 12 of 29 (41%) 21 of 50 (42%) 1.000 
Sister donor to brother 13 of 29 (45%) 18 of 50 (36%) .480 
Brother donor to sister 4 of 29 (14%) 11 of 50 (22%) .553 
Donor thalassemia minor 23 of 30 (77%) 37 of 51 (73%) .796 
Major ABO mismatch 7 of 30 (23%) 8 of 51 (16%) .394 
Ferritin at BMT, ng/mL* 2282 (591-8008) 1920 (376 to 6171) .354 
Hydroxyurea therapy prior to BMT 1 of 30 (3%) 21 of 51 (41%) <.001 
Liver size, cm from costal margin all ≤2 cm all ≤2 cm  
Spleen size, cm from costal margin 28 ≤ 2 cm, 1 = 3 cm, 1 NA 49 ≤ 2 cm, 1 = 3 cm, 1 = 4 cm  
Splenectomy 1 (3%)  
SGPT, IU/L (HCV-negative patients)* 49.5 (12-149) 32 (11 to 151) .008 
HCV positive 7 of 30 (23%) 5 of 51 (10%) .108 
SGPT, IU/L (HCV-positive patients)* 161 (23-537) 90 (35 to 118) .268 

Bold type indicates statistical significance.

HCV, hepatitis C virus; NA, not available.

*

Median (range).

Statistics

Data were collected and analyzed in December 2016. Patient characteristics were summarized through the use of medians and ranges. Fisher’s exact tests were used to compare proportions, and Mann-Whitney nonparametric tests were used to compare continuous distribution values. Kaplan-Meier survival curves were compared using the log-rank (Mantel-Cox) test. All P values are 2 tailed. Statistical analysis was performed using GraphPad Prism software version 5 (www.graphpad.com/prism/Prism.htm), except for contingency table Fisher exact test P values, which were generated at www.graphpad.com/quickcalcs/contingency1.cfm. Height-for-age and body mass index-for-age z-scores were calculated based on World Health Organization anthropometric data.32 

Patients in the Tt-BuCy group had a median age of 3.1 years, and no patient was >7 years of age. In the ATG-BuCy group, median age was 4.8 years, and 15 patients (29%) were >7 years and 3 (6%) were between 12 and 15 years of age. Follow-up was significantly longer in the Tt-BuCy group, with a median of 36.8 months (vs 16.9 months in the ATG-BuCy group). Pre-BMT ferritin values were slightly but not significantly higher in the Tt-BuCy than in the ATG-BuCy group (median 2282 ng/mL vs 1920 ng/mL); in fact, the latter group received more aggressive pre-BMT chelation and hydroxyurea (Hu) therapy (Table 2), and 10 patients had liver size between 2.5 to 5 cm, which decreased to ≤2 cm prior to BMT. SGPT median value was slightly higher in the hepatitis C–negative Tt-BuCy group but still within 3 times the normal value. There was no difference in cell dose between cohorts. Median day to neutrophil engraftment seemed moderately but significantly delayed (ie, 15 days in the Tt-BuCy vs 18 days in the ATG-BuCy group); however, since in the latter group the ANC nadir was reached a few days later, the difference in total number of neutropenic days was less evident (ie, 14 vs 16 days with ANC <500/µL and 11 and 12 days with ANC <200/µL, respectively). There was no statistically significant difference in platelet count recovery, number of platelet or erythrocyte transfusions, or day of last erythrocyte transfusion as well as day of discharge (Table 3).

Table 3.

Engraftment data

Tt-BuCyATG-BuCyP
No. of patients 30 51  
Cell dose, 108 nucleated cells/kg* 5.4 (3.1-16.8) 5.5 (2.6-18.9) .587 
G-CSF marrow priming (5 µg/kg × 3-5 d) 32 of 51 (63%) <.001 
Days to ANC >500/µL* 15 (12-28) 18 (13-32) <.001 
Total days of neutropenia <500/µL* 14 (10-27) 15 (8-28) .092 
Days to ANC >200/µL* 12 (10-28) 16 (11-29) <.001 
Total days of neutropenia <200/µL* 11 (8-27) 12 (6-24) .489 
Use of G-CSF post-BMT 2 (7%) 7 (14%) .473 
Days to platelet count >20 000/µL* 20 (13-68) 21 (13-38) .429 
Total red cell transfusions* 2 (0-9) 3 (0 to 9) .2392 
Last day of red cell transfusion* 11 (−14 to 152) 12 (−3 to 58) .643 
Total platelet transfusions* 4 (0-19) 4 (0-28) .945 
Discharge day* 39 (21-165) 46 (23-99) .235 
Tt-BuCyATG-BuCyP
No. of patients 30 51  
Cell dose, 108 nucleated cells/kg* 5.4 (3.1-16.8) 5.5 (2.6-18.9) .587 
G-CSF marrow priming (5 µg/kg × 3-5 d) 32 of 51 (63%) <.001 
Days to ANC >500/µL* 15 (12-28) 18 (13-32) <.001 
Total days of neutropenia <500/µL* 14 (10-27) 15 (8-28) .092 
Days to ANC >200/µL* 12 (10-28) 16 (11-29) <.001 
Total days of neutropenia <200/µL* 11 (8-27) 12 (6-24) .489 
Use of G-CSF post-BMT 2 (7%) 7 (14%) .473 
Days to platelet count >20 000/µL* 20 (13-68) 21 (13-38) .429 
Total red cell transfusions* 2 (0-9) 3 (0 to 9) .2392 
Last day of red cell transfusion* 11 (−14 to 152) 12 (−3 to 58) .643 
Total platelet transfusions* 4 (0-19) 4 (0-28) .945 
Discharge day* 39 (21-165) 46 (23-99) .235 

Bold type indicates statistical significance.

*

Median (range).

Actuarial rejection rates (defined as <5% donor DNA in peripheral blood) did not seem to differ between the Tt-BuCy and ATG-BuCy groups (11% vs 9%, respectively, with a log-rank P value = .79). In the ATG-BuCy group, in which most patients received Hu prior to BMT (as opposed to the Tt-BuCy group), of the 4 patients who rejected the transplant, 2 received Hu prior to BMT and 2 did not. Transplant-related mortality was higher, albeit not significantly, in the Tt-BuCy group than in the ATG-BuCy group (13% vs 6%, respectively; P = .37) and overall survival lower (87% vs 94%; P = .31), thalassemia-free survival was similar with 80% vs 85% (P = .59). Moderate to severe cGVHD could affect HRQoL as much as ST itself, and thus its occurrence might be considered a treatment failure. Composite cGVHD-free and thalassemia-free survival was 73% in the Tt-BuCy and 81% in the ATG-BuCy group (P = .79). In the 40 children <4 years at BMT (ie, the age group in which the addition of Tt to BuCy has been advocated because of higher rejection risk),15  the rejection rate was 10% in the 21 patients in the Tt-BuCy group and 0% in the 19 ATG-BuCy patients (P = .17). None of these outcome differences approached statistical significance, and both groups had a comparable number of patients at risk 18 months post-BMT (Figure 2). Mixed chimerism was observed in 80% of patients in the Tt group and 87% of patients in the ATG-BuCy group; all patients in the former group had chimerism >90% (level 1), while in the ATG-BuCy group, 7 patients (16%) had level 2 or 3 chimerism (Table 3). As outlined in Table 4, immunosuppression duration in thalassemia-free patients was significantly longer in the Tt-BuCy group, partly because donor mixed chimerism of <90%, which was observed only in the ATG-BuCy group, often prompted early immunosuppression taper, so that in patients with level 2 or 3 chimerism, the median (range) day of immunosuppression discontinuation was 257 (97-562) as opposed to 374 (158-497) in those patients with chimerism persistently ≥90% (P = .01). Patients with >8 months of follow-up had chimerism at least at 8 months, a time after which mixed chimerism tends to be stable.33,34  For cost-containment reasons, most children with a stable hemoglobin >9 g/dL after that time point did not have further chimerism testing, because it was not deemed clinically relevant. Only 1 patient in the Tt-BuCy group received a second transplant and is thalassemia-free at 46.5 months from the second BMT (described elsewere35 ); no child in the ATG-BuCy group has received a second BMT at the time of this analysis. Donors were all harvested under general anesthesia with no adverse events. All recipients tolerated the conditioning regimen well. One patient had an anaphylactic reaction to intravenous cyclosporine that did not recur after switching to the oral formulation. BMT-related complications are summarizes in Table 4. A total of 4 patients (13%) in the Tt-BuCy group and 3 patients (6%) in the ATG-BuCy group died. Mucositis ECOG score >2 (ie, severe enough to cause inability to eat) was more frequent in the former group (23% vs 2%), while the frequency of hemorrhagic cystitis ECOG score >2 (ie, gross hematuria with clots) was similar. Acute GVHD >2 occurred in 3% of patients in the Tt-BuCy group and 8% of patients in the ATG-BuCy group, with 2 GVHD-related deaths in the latter group. The incidence of moderate to severe cGVHD seemed to be higher, albeit not significantly, in the Tt-BuCy group, with 3 cases (10%) of bronchiolitis obliterans (BO), while in the ATG-BuCy, it occurred in 2 cases (4%) (1 BO and 1 nephrotic syndrome). In the latter group, there was 1 case of immune thrombocytopenia at day +75, which resolved. Two patients (both in the ATG-BuCy group) experienced intracranial hemorrhage, one with visual cortex involvement and decreased sight (which has been improving) and the other in the context of sepsis and disseminated intravascular coagulation. Two patients died of cGVHD (both BO) in the Tt-BuCy group, while both patients in the ATG-BuCy are alive and had normal performance scores at the last follow-up. All recipients were CMV positive at BMT, and in spite of comparable proportions of CMV-negative donors, CMV reactivation was seen significantly more often in the Tt-BuCy group (ie, 43% vs 13% in the ATG-BuCy), all patients received ganciclovir, and none had overt CMV disease. One patient in each group had a diagnosis of tuberculosis post-BMT (on day +190 [pulmonary] in the Tt-BuCy group and on day +406 [vertebral body] in the ATG-BuCy group); both patients have been treated and are doing well. One patient in the Tt-BuCy group died of encephalitis, the etiological cause of which could not be ascertained.

Figure 2.

Survival curves. (A) Overall survival; (B) thalassemia-free survival; (C) transplant-related mortality; (D) rejection; (E) composite GVHD and thalassemia-free survival; and (F) rejection in patients <4 years of age.

Figure 2.

Survival curves. (A) Overall survival; (B) thalassemia-free survival; (C) transplant-related mortality; (D) rejection; (E) composite GVHD and thalassemia-free survival; and (F) rejection in patients <4 years of age.

Close modal
Table 4.

Complications

Tt-BuCyATG-BuCyP
No. of patients 30 51  
Transplant-related deaths 4 (13%) (2 BO, 1 encephalitis, 1 TMA) 3 (6%) (2 aGVHD, 1 sepsis) .414 
Severe mucositis* 7 (23%) 1 (2%) .003 
Severe hemorrhagic cystitis* 3 (10%) 7 (14%) .737 
Hypertension stage 2 13 (43%) 26 (51%) .646 
aGVHD > grade 2 1 (3%) (grade 3) 4 (8%) (3 grade 3, 1 grade 4) .648 
cGVHD > mild 3 (10%) (all BO) 2 (4%) (1 BO, 1 NS) .350 
Immune cytopenias 1 (2%) (ITP) 1.000 
Sinusoidal obstructive syndrome 2 (7%) 1 (2%) .551 
Thrombotic microangiopathy 3 (10%) 1 (2%) .136 
CMV-positive recipient 100% 100% 1.000 
CMV-negative donor 5 (17%) 7 (14%) .753 
CMV reactivation 13 (43%) 7 (14%) .006 
Central venous line infection 5 (17%) 17 (33%) .126 
Clinical sepsis 2 (7%) 7 (13%) .473 
Fungal infection (proven or probable) 1.000 
Empirical use of antifungals 3 (10%) 5 (10%) 1.000 
Tuberculosis 1 (3%) 1 (2%) 1.000 
Malaria 1 (3%) .370 
Other 2 intracranial hemorrhage, 1 encephalitis 1 cutaneous leishmaniasis, 1 endocarditis, 1 Salmonella bacteremia, 1 HAV infection  
Immunosuppression withdrawal 409 (158-562) 320 (101-419) .013 
Mixed chimerism (5%-99%) with transfusion independence 20 of 25 evaluable cases (80%) 39 of 44 evaluable cases (87%) .478 
Level 1 (90%-99% donor) 20 32 .506 
Level 2 (75%-89% donor) .549 
Level 3 (5%-74% donor) .289 
Tt-BuCyATG-BuCyP
No. of patients 30 51  
Transplant-related deaths 4 (13%) (2 BO, 1 encephalitis, 1 TMA) 3 (6%) (2 aGVHD, 1 sepsis) .414 
Severe mucositis* 7 (23%) 1 (2%) .003 
Severe hemorrhagic cystitis* 3 (10%) 7 (14%) .737 
Hypertension stage 2 13 (43%) 26 (51%) .646 
aGVHD > grade 2 1 (3%) (grade 3) 4 (8%) (3 grade 3, 1 grade 4) .648 
cGVHD > mild 3 (10%) (all BO) 2 (4%) (1 BO, 1 NS) .350 
Immune cytopenias 1 (2%) (ITP) 1.000 
Sinusoidal obstructive syndrome 2 (7%) 1 (2%) .551 
Thrombotic microangiopathy 3 (10%) 1 (2%) .136 
CMV-positive recipient 100% 100% 1.000 
CMV-negative donor 5 (17%) 7 (14%) .753 
CMV reactivation 13 (43%) 7 (14%) .006 
Central venous line infection 5 (17%) 17 (33%) .126 
Clinical sepsis 2 (7%) 7 (13%) .473 
Fungal infection (proven or probable) 1.000 
Empirical use of antifungals 3 (10%) 5 (10%) 1.000 
Tuberculosis 1 (3%) 1 (2%) 1.000 
Malaria 1 (3%) .370 
Other 2 intracranial hemorrhage, 1 encephalitis 1 cutaneous leishmaniasis, 1 endocarditis, 1 Salmonella bacteremia, 1 HAV infection  
Immunosuppression withdrawal 409 (158-562) 320 (101-419) .013 
Mixed chimerism (5%-99%) with transfusion independence 20 of 25 evaluable cases (80%) 39 of 44 evaluable cases (87%) .478 
Level 1 (90%-99% donor) 20 32 .506 
Level 2 (75%-89% donor) .549 
Level 3 (5%-74% donor) .289 

Bold type indicates statistical significance.

HAV, hepatitis A virus; ICH, intracranial hemorrhage; ITP, immune thrombocytopenic purpura; NS, nephritic syndrome.

*

Greater than ECOG grade 2.

This study is the first multinational study in startup BMT services in LMICs comparing homogeneous patient groups and BMT regimens for ST. All centers had never performed BMT before, had an almost exclusive focus on matched-related BMT for thalassemia, and extensively employed information technology tools for daily interaction with experienced BMT professionals. In this selected but relatively common group of low-risk patients, outcomes were comparable to those reported in high-income countries.9  Our retrospective comparison suggests that the addition of a second myeloablative agent may not be warranted and that the ATG-BuCy combination can be quite tolerable and associated with low rejection rates.

We used the term severe thalassemia instead of thalassemia major to encompass higher-risk thalassemia intermedia patients and also bypass a possible confusion related to the Western working definition of thalassemia major (ie, requiring >8 erythrocyte transfusions per year).36  In fact, pretransfusion hemoglobin is often much lower in lower income countries than in the West, and thus transfusion frequency may not necessarily reflect disease severity. We also did not apply the Pesaro classification relying on liver biopsy,37  because the combination of liver size and age have been shown to be reliable predictors of transplant-associated risk in thalassemia.38,39  All children in this report had a liver ≤2 cm from costal margin, so they might be considered Pesaro class I or II. The age cutoff we adopted was extended from 8 to 15 years in the ATG-BuCy group. A high level of ferritin is known to be correlated with increased transplant-related mortality.40  The majority of our patients were aggressively chelated prior to BMT, and median serum ferritin values were <2500 ng/mL, a possible relevant cutoff in terms of transplant-related morbidity and mortality.39,41,42  Many patients, particularly in the ATG-BuCy group, were treated with Hu in order to minimize both liver and spleen size as well as extramedullary hematopoiesis.43,44 

There is a general perception that BMT centers need complex engineering standards requiring undue investments, even more so in developing countries, where the risk of infection might be greater. In fact, there is no evidence that the latter is true.45,46  On the contrary, local physicians might be in the best position to identify and treat locally prevalent diseases (eg, the patients with tuberculosis, malaria, and leishmaniasis in our series are doing well). There is increasing evidence that allogeneic transplant patients may be safely cared for in regular hospital rooms46  or even as outpatients,47,48  and many widely held practices calling for “sterile” BMT units are being reconsidered. In fact, international guidelines do not mandate admission in HEPA-filtered or positive pressure gradient rooms, at least for low-risk BMTs.49  The human development index does not seem to correlate with non–relapse mortality after BMT for acute leukemia,50  suggesting that the quality of remission assessment, an issue that does not pertain to ST, might be more relevant than where the BMT procedure itself is carried out. The infectious complications we observed did not seem to be substantially different from those observed internationally.19,51 

The chronic nature of ST, which allows to perform BMT in patients when conditions are optimal, combined with their lack of prior exposure to chemotherapy, low mucositis, and GVHD rates, as well as relative short duration of profound neutropenia, most likely played a role in the very low rate of fungal infections we observed, and the same may not apply to higher-risk diseases such as leukemias or severe aplastic anemias.

The 2 initial centers of this project, CHPIMS and SEAIT, were both staffed by professionals with little or no BMT experience who relied on on-site, whole-team–focused training by experienced BMT physicians followed by daily interaction via video conferencing and web-based medical records.24  The centers in Sri Lanka (ACH and NH) were run by adult hematologists with some BMT experience in collaboration with pediatricians, as well as on-site and online consultation with pediatric transplant physicians. This is the first report on a large series of thalassemia patients undergoing BMT exclusively in startup centers and directly in LMICs and might provide a relevant precedent to address the dire shortage of such services in emerging countries,52  where hemoglobinopathies are the most frequent life-threatening noncommunicable disease among children1  and a major indication for BMT.6,7,53 

Adding Tt54  or ATG13,17-19  to the BuCy backbone has substantially reduced rejection rates. In the age group <4 years of age, where Tt seems particularly important to reduce rejection,15  we observed no significant rejection differences between the ATG and Tt groups. A combination of treosulfan, Tt, and fludarabine has also been advocated as the preferable BMT conditioning for patients with thalassemia, particularly those with higher transplantation risk55-57 ; however, this recommendation is not based on prospective studies, and the combined use of 2 myeloablative alkylating agents may be associated with high infertility rates20  as well as substantial increases in costs. A formal cost analysis could not be performed, but the average all-inclusive cost per BMT was ∼12 000 US dollars and that of family support was ∼2000 US dollars. We also believe that in the current era of effective chelation, it might no longer be justified to take higher-risk patients directly to transplant without aggressive chelation and possibly Hu prior to BMT. In our series, 10 patients (all in the ATG-BuCy group) had an initial liver size >2 cm from costal margin and could thus have been considered higher-risk patients, but they were downstaged into a lower risk category by aggressive supportive care prior to BMT.

In long-term follow-up, BuCy has been shown to be associated with birth rates comparable to the control population in former ST patients.10  However, our current report does not have sufficient follow-up to directly assess the impact of substituting Tt with ATG on pubertal development or fertility.

In our experience with Tt-BuCy, severe GVHD (grade III-IV) was quite rare (in the 3% range). For this reason, we opted for early and low-dose ATG administration (days −12 to −10) with little or no ATG carryover at marrow infusion, thus favoring engraftment and immune reconstitution as opposed to the standard day −5 to −1 ATG timing geared also toward in vivo donor T-cell depletion and GVHD prevention. Moreover, all our patients received bone marrow rather than mobilized leukapheresis, the latter having been associated with an increased risk of extensive cGVHD,58-60  a major factor affecting HRQoL and risk of secondary malignancies post-BMT.10  Most patients in the ATG-BuCy group received G-CSF–primed marrow, which may have further contributed to decreased GVHD occurrence.61  None had autologous backup marrow stored prior to transplant, since its indication is questionable.62  No formal immune reconstitution studies were performed, but the decreased frequency of CMV reactivation in the ATG-BuCy group suggests that the ATG approach employed in this study might be less immunosuppressive than Tt-BuCy. This might also be reflected by an increased rate of stable mixed chimerism in the ATG-BuCy group, although rejection rates were comparable. Persistent mixed chimerism with transfusion independence is a well-known occurrence after BMT for thalassemia33  and might actually be an advantage given the reduced risk of GVHD associated with mixed chimerism.63 

The major limitation of this report is its retrospective nature, which makes it difficult to tease out whether the apparent, albeit not statistically significant, improvement in thalassemia-free and overall survival observed with ATG-BuCy over Tt-BuCy is related to differences in duration of follow-up and other confounding factors. However, the number of patients at risk at 18 months (a relevant time point after which rejection is rarely observed and mixed chimerism is stable in most patients31 ) was comparable. Also, median age was significantly higher in the ATG-BuCy group, with 8 patients being older than 7 years as opposed to none in the Tt-BuCy group, which might have skewed outcomes in favor of the latter,39  while pre-BMT therapy with Hu and increased use of G-CSF priming of marrow might have favored the ATG-BuCy group, as well as a time-period effect as the startup centers gained more experience.

In conclusion, the ATG-BuCy combination seems to be as effective as, and possibly safer than, Tt-BuCy. Within structured and intensive cooperation programs with experienced BMT professionals focusing on a single disease and a low-risk transplant procedure, it is possible to obtain high success rates directly in LMICs, even without highly trained physicians and nurses.

The authors thank the patients, their parents, and all the nurses and physicians at collaborating institutions.

This work was partially supported by Cure2Children Foundation (Florence, Italy), Sankalp India Foundation (Bangalore, India), Heartfile (Islamabad, Pakistan), Fondazione Monte dei Paschi di Siena (Siena, Italy), Cassa di Risparmio di Firenze (Florence, Italy), and The Italian and Pakistani Governments (Cooperazione Italiana allo Sviluppo through the Pakistan-Italy Dept for Development Swap Agreement). The authors are also grateful to DKMS (Tübingen, Germany) for providing free HLA typing.

Contribution: L.F. designed and performed the research, collected the data, performed the statistical analyses, interpreted the data, and wrote the manuscript; R.K.A., A.S., and R. Dhanya collected the data and performed the statistical analyses; L.F, C.U., S.K., P.M., R.S., N.Y., S.A., I.F., S.K.G., T.Z., S.R., L.G., R. Dissanayake, S.W., W.R., and R.S. participated in patient management; R.K.A., A.S., R.S., R. Dhanya, L.P., and A.K. coordinated patient registration, data management, and HLA typing; and all authors critically reviewed and approved the manuscript.

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

Correspondence: Lawrence Faulkner, Medical Coordinator Cure2Children Foundation, via Marconi 30, 50131 Florence, Italy; e-mail: lawrence.faulkner@cure2children.org.

1.
Weatherall
DJ
.
The inherited diseases of hemoglobin are an emerging global health burden
.
Blood
.
2010
;
115
(
22
):
4331
-
4336
.
2.
de Silva
S
,
Fisher
CA
,
Premawardhena
A
, et al
;
Sri Lanka Thalassaemia Study Group
.
Thalassaemia in Sri Lanka: implications for the future health burden of Asian populations
.
Lancet
.
2000
;
355
(
9206
):
786
-
791
.
3.
Ho
W-L
,
Lin
K-H
,
Wang
J-D
, et al
.
Financial burden of national health insurance for treating patients with transfusion-dependent thalassemia in Taiwan
.
Bone Marrow Transplant
.
2006
;
37
(
6
):
569
-
574
.
4.
Riewpaiboon
A
,
Nuchprayoon
I
,
Torcharus
K
, et al
. Economic burden of beta-thalassemia/Hb E and beta-thalassemia major in Thai children. BMC Res Notes.
2010
;3:29.
5.
Rachmilewitz
EA
,
Giardina
PJ
.
How I treat thalassemia
.
Blood
.
2011
;
118
(
13
):
3479
-
3488
.
6.
Chandy
M
.
Stem cell transplantation in India
.
Bone Marrow Transplant
.
2008
;
42
(
Suppl 1
):
S81
-
S84
.
7.
Shamsi
T
,
Hashmi
K
,
Adil
S
, et al
.
The stem cell transplant program in Pakistan--the first decade
.
Bone Marrow Transplant
.
2008
;
42
(
suppl 1
):
S114
-
S117
.
8.
Aljurf
MD
,
Zaidi
SZ
,
El Solh
H
, et al
.
Special issues related to hematopoietic SCT in the Eastern Mediterranean region and the first regional activity report
.
Bone Marrow Transplant
.
2009
;
43
(
1
):
1
-
12
.
9.
Angelucci
E
,
Matthes-Martin
S
,
Baronciani
D
, et al
;
EBMT Inborn Error and EBMT Paediatric Working Parties
.
Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: indications and management recommendations from an international expert panel
.
Haematologica
.
2014
;
99
(
5
):
811
-
820
.
10.
La Nasa
G
,
Caocci
G
,
Efficace
F
, et al
.
Long-term health-related quality of life evaluated more than 20 years after hematopoietic stem cell transplantation for thalassemia
.
Blood
.
2013
;
122
(
13
):
2262
-
2270
.
11.
Leelahavarong
P
,
Chaikledkaew
U
,
Hongeng
S
,
Kasemsup
V
,
Lubell
Y
,
Teerawattananon
Y
.
A cost-utility and budget impact analysis of allogeneic hematopoietic stem cell transplantation for severe thalassemic patients in Thailand
.
BMC Health Serv Res
.
2010
;
10
:
209
-
221
.
12.
Scalone
L
,
Mantovani
LG
,
Krol
M
, et al
.
Costs, quality of life, treatment satisfaction and compliance in patients with beta-thalassemia major undergoing iron chelation therapy: the ITHACA study
.
Curr Med Res Opin
.
2008
;
24
(
7
):
1905
-
1917
.
13.
Galambrun
C
,
Pondarré
C
,
Bertrand
Y
, et al
;
French Rare Disease Center for Thalassemia; French Society of Bone Marrow Transplantation
.
French multicenter 22-year experience in stem cell transplantation for beta-thalassemia major: lessons and future directions
.
Biol Blood Marrow Transplant
.
2013
;
19
(
1
):
62
-
68
.
14.
Lucarelli
G
,
Giardini
C
,
Baronciani
D
.
Bone marrow transplantation in thalassemia
.
Semin Hematol
.
1995
;
32
(
4
):
297
-
303
.
15.
Isgrò
A
,
Gaziev
J
,
Sodani
P
,
Lucarelli
G
.
Progress in hematopoietic stem cell transplantation as allogeneic cellular gene therapy in thalassemia
.
Ann N Y Acad Sci
.
2010
;
1202
:
149
-
154
.
16.
Bernardo
ME
,
Piras
E
,
Vacca
A
, et al
.
Allogeneic hematopoietic stem cell transplantation in thalassemia major: results of a reduced-toxicity conditioning regimen based on the use of treosulfan
.
Blood
.
2012
;
120
(
2
):
473
-
476
.
17.
Li
CK
,
Shing
MMK
,
Chik
KW
, et al
.
Haematopoietic stem cell transplantation for thalassaemia major in Hong Kong: prognostic factors and outcome
.
Bone Marrow Transplant
.
2002
;
29
(
2
):
101
-
105
.
18.
Lawson
SE
,
Roberts
IAG
,
Amrolia
P
,
Dokal
I
,
Szydlo
R
,
Darbyshire
PJ
.
Bone marrow transplantation for beta-thalassaemia major: the UK experience in two paediatric centres
.
Br J Haematol
.
2003
;
120
(
2
):
289
-
295
.
19.
Goussetis
E
,
Peristeri
I
,
Kitra
V
, et al
.
HLA-matched sibling stem cell transplantation in children with β-thalassemia with anti-thymocyte globulin as part of the preparative regimen: the Greek experience
.
Bone Marrow Transplant
.
2012
;
47
(
8
):
1061
-
1066
.
20.
Anserini
P
,
Chiodi
S
,
Spinelli
S
, et al
.
Semen analysis following allogeneic bone marrow transplantation. Additional data for evidence-based counselling
.
Bone Marrow Transplant
.
2002
;
30
(
7
):
447
-
451
.
21.
Vatanen
A
,
Wilhelmsson
M
,
Borgström
B
, et al
.
Ovarian function after allogeneic hematopoietic stem cell transplantation in childhood and adolescence
.
Eur J Endocrinol
.
2013
;
170
(
2
):
211
-
218
.
22.
Zafari
M
,
Kosaryan
M
.
Marriage and child bearing in patients with transfusion-dependent thalassemia major
.
J Obstet Gynaecol Res
.
2014
;
40
(
8
):
1978
-
1982
.
23.
World Bank
. Lower middle income data. http://data.worldbank.org/income-level/lower-middle-income. Accessed 3 October 2016.
24.
Faulkner
LB
,
Uderzo
C
,
Masera
G
.
International cooperation for the cure and prevention of severe hemoglobinopathies
.
J Pediatr Hematol Oncol
.
2013
;
35
(
6
):
419
-
423
.
25.
Agarwal
RK
,
Sedai
A
,
Dhimal
S
, et al
. A prospective international cooperative information technology platform built using open-source tools for improving the access to and safety of bone marrow transplantation in low- and middle-income countries. J Am Med Inform Assoc.
2014
;21(6):1125-1128.
26.
Eastern Cooperative Oncology Group
. Common toxicity criteria. http://www.ecog.org/general/common_tox.htmlm. Accessed 1 October 2016.
27.
National Institutes of Health
. The fourth report on the diagnosis, evaluation, and treatment of high blood pressure in children and adolescents. NIH publication no. 05-5267;
2005
. https://www.nhlbi.nih.gov/files/docs/resources/heart/hbp_ped.pdf. Accessed 2 May 2017.
28.
McDonald
GB
,
Hinds
MS
,
Fisher
LD
, et al
.
Veno-occlusive disease of the liver and multiorgan failure after bone marrow transplantation: a cohort study of 355 patients
.
Ann Intern Med
.
1993
;
118
(
4
):
255
-
267
.
29.
Przepiorka
D
,
Weisdorf
D
,
Martin
P
, et al
.
1994 Consensus Conference on Acute GVHD Grading
.
Bone Marrow Transplant
.
1995
;
15
(
6
):
825
-
828
.
30.
Filipovich
AH
,
Weisdorf
D
,
Pavletic
S
, et al
.
National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report
.
Biol Blood Marrow Transplant
.
2005
;
11
(
12
):
945
-
956
.
31.
Andreani
M
,
Testi
M
,
Battarra
M
, et al
.
Relationship between mixed chimerism and rejection after bone marrow transplantation in thalassaemia
.
Blood Transfus
.
2008
;
6
(
3
):
143
-
149
.
32.
World Health Organization
. The WHO Child Growth Standards. http://www.who.int/childgrowth/en/. Accessed 20 October 2016.
33.
Andreani
M
,
Nesci
S
,
Lucarelli
G
, et al
.
Long-term survival of ex-thalassemic patients with persistent mixed chimerism after bone marrow transplantation
.
Bone Marrow Transplant
.
2000
;
25
(
4
):
401
-
404
.
34.
Li
CK
,
Chik
KW
,
Tsang
KS
,
Pong
H
,
Shing
MM
,
Yuen
PM
.
Mixed chimerism after bone marrow transplantation for thalassemia major
.
Haematologica
.
2002
;
87
(
7
):
781
-
782
.
35.
Yaqub
N
,
Khalid
S
,
Fatima
I
, et al
.
Second BMT for thalassemia major using CY post transplant
.
Bone Marrow Transplant
.
2014
;
49
(
6
):
845
-
846
.
36.
Cunningham
MJ
,
Macklin
EA
,
Neufeld
EJ
,
Cohen
AR
;
Thalassemia Clinical Research Network
.
Complications of beta-thalassemia major in North America
.
Blood
.
2004
;
104
(
1
):
34
-
39
.
37.
Lucarelli
G
,
Galimberti
M
,
Polchi
P
, et al
.
Bone marrow transplantation in patients with thalassemia
.
N Engl J Med
.
1990
;
322
(
7
):
417
-
421
.
38.
Mathews
V
,
Lakshmi
KM
,
Viswabandya
A
,
George
B
,
Chandy
M
,
Srivastava
A
.
Risk stratification without a liver biopsy of patients with β thalassemia major undergoing a matched related allogeneic bone marrow transplant [abstract]
.
Blood
.
2009
;
114
(
22
). Abstract
659
.
39.
Sabloff
M
,
Chandy
M
,
Wang
Z
, et al
.
HLA-matched sibling bone marrow transplantation for β-thalassemia major
.
Blood
.
2011
;
117
(
5
):
1745
-
1750
.
40.
Lee
JW
,
Kang
HJ
,
Kim
EK
,
Kim
H
,
Shin
HY
,
Ahn
HS
.
Effect of iron overload and iron-chelating therapy on allogeneic hematopoietic SCT in children
.
Bone Marrow Transplant
.
2009
;
44
(
12
):
793
-
797
.
41.
Armand
P
,
Kim
HT
,
Cutler
CS
, et al
.
Prognostic impact of elevated pretransplantation serum ferritin in patients undergoing myeloablative stem cell transplantation
.
Blood
.
2007
;
109
(
10
):
4586
-
4588
.
42.
Wang
Z
,
Jia
M
,
Zhao
H
, et al
.
Prognostic impact of pretransplantation hyperferritinemia in adults undergoing allogeneic hematopoietic SCT: a meta-analysis
.
Bone Marrow Transplant
.
2014
;
49
(
10
):
1339
-
1340
.
43.
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
;
104
(
4
):
1201
-
1203
.
44.
El-Beshlawy
A
,
El-Ghamrawy
M
,
EL-Ela
MA
, et al
.
Response to hydroxycarbamide in pediatric β-thalassemia intermedia: 8 years’ follow-up in Egypt
.
Ann Hematol
.
2014
;
93
(
12
):
2045
-
2050
.
45.
George
B
,
Mathews
V
,
Viswabandya
A
,
Srivastava
A
,
Chandy
M
.
Infections in children undergoing allogeneic bone marrow transplantation in India
.
Pediatr Transplant
.
2006
;
10
(
1
):
48
-
54
.
46.
Kumar
R
,
Naithani
R
,
Mishra
P
, et al
.
Allogeneic hematopoietic SCT performed in non-HEPA filter rooms: initial experience from a single center in India
.
Bone Marrow Transplant
.
2009
;
43
(
2
):
115
-
119
.
47.
Svahn
B-M
,
Remberger
M
,
Myrbäck
K-E
, et al
.
Home care during the pancytopenic phase after allogeneic hematopoietic stem cell transplantation is advantageous compared with hospital care
.
Blood
.
2002
;
100
(
13
):
4317
-
4324
.
48.
Solomon
SR
,
Matthews
RH
,
Barreras
AM
, et al
.
Outpatient myeloablative allo-SCT: a comprehensive approach yields decreased hospital utilization and low TRM
.
Bone Marrow Transplant
.
2010
;
45
(
3
):
468
-
475
.
49.
FACT-JACIE. International Standards for Hematopoietic Cellular Therapy Product Collection, Processing and Administration.
6th ed. http://www.jacie.org/standards/6th-edition-2015. Accessed 2 May 2017.
50.
Giebel
S
,
Labopin
M
,
Ehninger
G
, et al
;
Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation
.
Association of Human Development Index with rates and outcomes of hematopoietic stem cell transplantation for patients with acute leukemia
.
Blood
.
2010
;
116
(
1
):
122
-
128
.
51.
Nihtinen
A
,
Anttila
V-J
,
Richardson
M
, et al
.
Invasive Aspergillus infections in allo-SCT recipients: environmental sampling, nasal and oral colonization and galactomannan testing
.
Bone Marrow Transplant
.
2010
;
45
(
2
):
333
-
338
.
52.
Gratwohl
A
,
Baldomero
H
,
Aljurf
M
, et al
;
Worldwide Network of Blood and Marrow Transplantation
.
Hematopoietic stem cell transplantation: a global perspective
.
JAMA
.
2010
;
303
(
16
):
1617
-
1624
.
53.
Aljurf
MD
,
Zaidi
SZ
,
El Solh
H
, et al
.
Special issues related to hematopoietic SCT in the Eastern Mediterranean Region and the first regional activity report
.
Bone Marrow Transplant
.
2009
;
43
(
1
):
1
-
12
.
54.
Marziali
M
,
Isgrò
A
,
Gaziev
J
,
Lucarelli
G
.
Hematopoietic stem cell transplantation in thalassemia and sickle cell disease. Unicenter experience in a multi-ethnic population
.
Mediterr J Hematol Infect Dis
.
2009
;
1
(
1
):
e2009027
.
55.
Bernardo
ME
,
Zecca
M
,
Piras
E
, et al
.
Treosulfan-based conditioning regimen for allogeneic haematopoietic stem cell transplantation in patients with thalassaemia major
.
Br J Haematol
.
2008
;
143
(
4
):
548
-
551
.
56.
Choudhary
D
,
Sharma
SK
,
Gupta
N
, et al
.
Treosulfan-thiotepa-fludarabine-based conditioning regimen for allogeneic transplantation in patients with thalassemia major: a single-center experience from north India
.
Biol Blood Marrow Transplant
.
2013
;
19
(
3
):
492
-
495
.
57.
Mathews
V
,
Savani
BN
.
Conditioning regimens in allo-SCT for thalassemia major
.
Bone Marrow Transplant
.
2014
;
49
(
5
):
607
-
610
.
58.
Eapen
M
,
Horowitz
MM
,
Klein
JP
, et al
.
Higher mortality after allogeneic peripheral-blood transplantation compared with bone marrow in children and adolescents: the Histocompatibility and Alternate Stem Cell Source Working Committee of the International Bone Marrow Transplant Registry
.
J Clin Oncol
.
2004
;
22
(
24
):
4872
-
4880
.
59.
Kumar
R
,
Kimura
F
,
Ahn
KW
, et al
.
Comparing outcomes with bone marrow or peripheral blood stem cells as graft source for matched sibling transplants in severe aplastic anemia across different economic regions
.
Biol Blood Marrow Transplant
.
2016
;
22
(
5
):
932
-
940
.
60.
Zaidman
I
,
Rowe
JM
,
Khalil
A
,
Ben-Arush
M
,
Elhasid
R
.
Allogeneic stem cell transplantation in congenital hemoglobinopathies using a tailored busulfan-based conditioning regimen: single-center experience
.
Biol Blood Marrow Transplant
.
2016
;
22
(
6
):
1043
-
1048
.
61.
Deotare
U
,
Al-Dawsari
G
,
Couban
S
,
Lipton
JH
.
G-CSF-primed bone marrow as a source of stem cells for allografting: revisiting the concept
.
Bone Marrow Transplant
.
2015
;
50
(
9
):
1150
-
1156
.
62.
Stelljes
M
,
van Biezen
A
,
Slavin
S
, et al
;
complications subcommittee of the Chronic Leukemia Working Party
.
The harvest and use of autologous back-up grafts for graft failure or severe GVHD after allogeneic hematopoietic stem cell transplantation: a survey of the European Group for Blood and Marrow Transplantation
.
Bone Marrow Transplant
.
2008
;
42
(
11
):
739
-
742
.
63.
Huss
R
,
Deeg
HJ
,
Gooley
T
, et al
.
Effect of mixed chimerism on graft-versus-host disease, disease recurrence and survival after HLA-identical marrow transplantation for aplastic anemia or chronic myelogenous leukemia
.
Bone Marrow Transplant
.
1996
;
18
(
4
):
767
-
776
.