Hepatic sinusoidal obstruction syndrome (SOS) is a serious complication in hematopoietic stem cell transplant (HSCT) recipients. To determine the impact of pretransplantation hyperferritinemia on the risk of SOS after HSC transplantation, we retrospectively studied 427 HSCT recipients (179 autologous and 248 allogeneic). Serum ferritin levels were measured before transplantation. Patients with and without a diagnosis of SOS were compared regarding demographics; underlying disease; transplant characteristics; receipt of imatinib, busulfan, total body irradiation, gemtuzumab, vancomycin, acyclovir, or methotrexate; and baseline serum ferritin. Univariate and multivariate (stepwise logistic regression) analyses were performed. SOS was diagnosed in 88 patients (21%) at a median of 10 days (range, 2-29 days) after transplantation. By multivariate analysis, allogeneic HSC transplantation (odds ratio [OR] = 8.25; 95% confidence interval [95% CI], 3.31-20.57), receipt of imatinib (OR = 2.60; 95% CI, 1.16-5.84), receipt of busulfan (OR = 2.18; 95% CI, 1.25-3.80), and ferritin serum level higher than 1000 ng/dL (OR = 1.78; 95% CI, 1.02-3.08) were risk factors for SOS. A ferritin serum level higher than 1000 ng/dL in the pretransplantation period is an independent risk factor for SOS. The results suggest the need for prospective studies addressing the use of iron chelation in the pretransplantation period.

Sinusoidal obstruction syndrome (SOS), previously referred to as veno-occlusive disease, is one of the most common life-threatening complications of hematopoietic stem cell (HSC) transplantation. It is characterized by the presence of at least 2 of the following features: hyperbilirubinemia, painful hepatomegaly, and weight gain. In patients undergoing allogeneic HSC transplantation, SOS is the third cause of transplant-related death, with a reported mortality rate of up to 50%.1-3  The frequency of SOS varies greatly, from 1% to 2% among pediatric HSC transplant (HSCT) recipients, to more than 50% in adult HSCT recipients with hematologic malignancies.1,2  Cytoreductive therapy is presumably the primary cause of SOS, but other factors may also play a role. Pretransplantation liver disease; viral hepatitis; cytomegalovirus (CMV) seropositivity; intensity of the conditioning regimen; receipt of busulfan, gemtuzumab, estrogen-progestin, methotrexate, everolimus, acyclovir, amphotericin, or vancomycin; and mismatched or unrelated allogeneic HSC transplantations have also been associated with an increase in the risk of SOS.3-9 

Recent studies have reported an association between pretransplantation iron overload and several outcomes in HSCT recipients, including an increased risk of infection10  and transplant-related mortality.11  In addition, 2 studies suggested that iron overload may be a risk factor for SOS.11,12 

We sought to determine the risk factors for SOS in a cohort of autologous and allogeneic HSCT recipients. We were particularly interested in examining the role of pretransplantation serum ferritin as a risk factor for SOS.

This is a retrospective cohort study conducted at Instituto Nacional de Câncer. Between January 2002 and March 2007, 429 HSCT recipients were included in the study. The study protocol was approved by the local ethics committee at the Instituto Nacional de Câncer. Data were retrospectively collected from the medical files and stored in a Microsoft Excel spreadsheet (Seattle, WA). All HSCT recipients were considered eligible if they were alive at day +3 after transplantation and had a ferritin serum level measured before transplantation.

The diagnosis of SOS was based on clinical criteria originally proposed by McDonald et al.13  Two of the following criteria had to be present within 20 days after transplantation, and not explained by other reasons: hyperbilirubinemia (bilirubin > 34.2 μM [2.0 mg/dL]), painful hepatomegaly, and unexplained weight gain (> 2% from baseline). Patients with a diagnosis of hepatic graft-versus-host disease (GVHD) were not considered to have SOS even if all diagnostic criteria for SOS were fulfilled. SOS severity was classified as mild if resolved before day +100 without treatment; moderate if resolved before day +100, but requiring specific treatment; and severe if not resolved before day +100. Two of the authors (S.C.M. and M.C.) prospectively followed all patients, and one author (S.C.M.) reviewed all medical charts to determine who developed SOS and to classify its severity.

Hepatic dysfunction was defined as any elevation in serum transaminases, hypoalbuminemia (< 35 g/L [3.5 g/dL]), or histologic evidence of hepatic fibrosis or cirrhosis, documented before the administration of the conditioning regimen. Serum levels of ferritin were obtained before the start of the conditioning regimen, and were measured using the IMMULITE 2000 immunoassay system (Siemens Healthcare Diagnostics). Ferritin serum levels were measured prospectively in all patients. Status of the underlying disease was defined as early (nonmalignant diseases, acute leukemia in first complete remission, or chronic myeloid leukemia in first chronic phase) or advancer (other diseases). Fanconi anemia (5 patients), Krabbe disease and Wiskott-Aldrich syndrome (2 patients each) and Chédiak-Higashi syndrome, severe combined immunodeficiency syndrome, and Kostmann syndrome (1 patient each) were not classified.

For the evaluation of risk factors for SOS, patients with and without SOS were compared regarding age, sex, underlying disease, type (malignant vs nonmalignant) and status (early vs advanced) of the underlying disease, type of HSC transplantation (allogeneic vs autologous/syngeneic), relatedness of donor (sibling vs unrelated), human leukocyte antigen (HLA) match (matched vs mismatched), history of hepatitis, cytomegalovirus (CMV) serology of the recipient, previous HSC transplantation, receipt of imatinib before HSC transplantation, receipt of gemtuzumab, vancomycin, or acyclovir until 7 days before the start of the conditioning regimen, receipt of busulfan or total body irradiation in the conditioning regimen, intensity of the conditioning regimen (myeloablative vs nonmyeloablative), stem cell source (bone marrow vs peripheral blood vs cord blood), receipt of methotrexate in the prophylaxis of graft-versus-host disease (GVHD), and the presence of hepatic dysfunction.

For univariate analyses, chi-square and Fisher exact tests were used to analyze the association between SOS and the categoric variables. The comparison of continuous variables was performed with the Mann-Whitney test. Variables with P value less than .05 by univariate analysis were entered in a forward stepwise logistic regression analysis. To make sure that potential interactions between variables could negatively influence the results of the multivariate analysis, because some variables could be related, we tested the potential interaction between these variables (such as underlying disease and type of HSC transplantation or acute leukemia and receipt of busulfan in the conditioning regimen). If there was an interaction, the interaction term was entered in the multivariate model. Survival curves were constructed using the Kaplan-Meier method and compared by the log-rank test. The selection of the best cutoff point for ferritin as a risk factor for the diagnosis of SOS was done by constructing receiver operating characteristic (ROC) curves, with estimation of sensitivity, specificity, and area under the curve. All analyses were performed using SPSS 13.0 for Windows (SPSS). P values less than .05 were considered statistically significant.

The characteristics of the 427 patients and transplants are summarized in Table 1. Two patients from the original cohort of 429 patients were excluded from the analysis: 1 died before day +3 due to sepsis, and the other had a diagnosis of SOS made on day +174, and was retrospectively considered a misdiagnosis.

Table 1

Characteristics of the 427 patients

No.
Male:female 255:172 
Median age, y (range) 31 (0-66) 
Underlying disease, no. (%)  
    Lymphoma, multiple myeloma 196 (46) 
    Acute leukemia, myelodysplasia, PNH 136 (32) 
    Myeloproliferative disease 56 (13) 
    Other 39 (9) 
Advanced underlying disease,* no. (%) 315/415 (76) 
Type of HSC transplantation, no. (%)  
    Autologous/syngeneic 181 (42) 
    Allogeneic, myeloablative 208 (49) 
    Allogeneic, nonmyeloablative 38 (9) 
Type of donor and HLA status, no. (%)  
    HLA-compatible sibling 187/246 (76) 
    HLA-compatible unrelated 28/246 (11) 
    HLA-mismatched unrelated 33/246 (13) 
Conditioning regimen, no. (%)  
    Busulfan based 119 (28) 
    TBI based 85 (20) 
    Other 223 (52) 
Stem cell source, no. (%)  
    Peripheral blood 225 (53) 
    Bone marrow 168 (39) 
    Cord blood 34 (8) 
No.
Male:female 255:172 
Median age, y (range) 31 (0-66) 
Underlying disease, no. (%)  
    Lymphoma, multiple myeloma 196 (46) 
    Acute leukemia, myelodysplasia, PNH 136 (32) 
    Myeloproliferative disease 56 (13) 
    Other 39 (9) 
Advanced underlying disease,* no. (%) 315/415 (76) 
Type of HSC transplantation, no. (%)  
    Autologous/syngeneic 181 (42) 
    Allogeneic, myeloablative 208 (49) 
    Allogeneic, nonmyeloablative 38 (9) 
Type of donor and HLA status, no. (%)  
    HLA-compatible sibling 187/246 (76) 
    HLA-compatible unrelated 28/246 (11) 
    HLA-mismatched unrelated 33/246 (13) 
Conditioning regimen, no. (%)  
    Busulfan based 119 (28) 
    TBI based 85 (20) 
    Other 223 (52) 
Stem cell source, no. (%)  
    Peripheral blood 225 (53) 
    Bone marrow 168 (39) 
    Cord blood 34 (8) 

PNH indicates paroxysmal nocturnal hemoglobinuria; HSC, hematopoietic stem cell; HLA, human leukocyte antigen; and TBI, total body irradiation.

*

Not applicable to hereditary disorders such as Fanconi anemia and Wiskott-Aldrich syndrome.

The most frequent conditioning regimens used were CBV (cyclophosphamide 7200 mg/m2, carmustine [BCNU] 450 mg/m2, etoposide 2400 mg/m2), given to 119 patients; Bu-Cy (busulfan 16 mg/kg by mouth, cyclophosphamide 120 mg/kg), given to 110 patients; and MEL200 (melphalan 200 mg/m2), given to 51 patients. Among the 246 allogeneic HSCT recipients, 218 (89%) received cyclosporine and methotrexate as graft-versus-host disease (GVHD) prophylaxis. Methotrexate was given at a dose of 10 mg/m2 on day +1, and 5 mg/m2 on days +3 and +6 for nonmyeloablative allogeneic HSC transplantation, and 15 mg/m2 on day +1, 10 mg/m2 on days +3 and +6 for myeloablative allogeneic HSC transplantation. Patients with aplastic anemia and recipients of unrelated donor transplants received and an additional dose of 10 mg/m2 on day +11.

The overall incidence of SOS was 21% (88 of 427). The median time of the diagnosis of SOS was 10 days from day zero of transplantation (range, 2-29 days). The severity of SOS was graded as mild in 35%, moderate in 50%, and severe in 15% of cases. Liver biopsy was not performed in any patient. Histopathology confirmed the diagnosis of SOS in the 3 patients who died and an autopsy was performed. The death rate among the 88 patients with SOS was 65%. The overall survival at day +60 after transplantation was significantly lower in patients with SOS compared with patients who did not develop SOS (73% vs 93%, P < .001, log-rank test). The 60-day and 5-year cumulative incidence of treatment-related mortality were 10% and 27%, respectively.

Table 2 shows the univariate analysis of risk factors for SOS. Patients with SOS were more likely to have chronic myeloid leukemia or acute leukemia as underlying disease, and less likely to have lymphoma or multiple myeloma. In addition, there was a strong association between SOS and allogeneic HSC transplantation (odds ratio [OR] = 14.60; 95% confidence interval [95% CI], 6.20-34.30), bone marrow as the source of stem cells (OR = 4.58; 95% CI, 2.69-7.83), receipt of imatinib (OR = 4.77; 95% CI, 2.21-10.30), and receipt of busulfan (OR = 3.99; 95% CI, 2.44-6.53). In the analysis of potential interaction between variables, the following variables presented interaction: a diagnosis of lymphoma and autologous HSC transplantation, a diagnosis of chronic myeloid leukemia and allogeneic HSC transplantation, a diagnosis of acute leukemia and receipt of busulfan in the conditioning regimen, and receipt of imatinib and a diagnosis of chronic myeloid leukemia.

Table 2

Risk factors for sinusoidal obstruction syndrome by univariate analysis

SOS
POdds ratio (95% CI)
Yes n=88No n=339
Male:female 49:39 206:133 .38 0.81 (0.50-1.30) 
Median age, y (range) 34.5 (6-33) 30 (0-66) .62 NA 
Underlying disease, no. (%)     
    Lymphoma 7 (8) 135 (40) < .001 0.13 (0.06-0.30) 
    Acute leukemia/MDS 41 (46) 88 (26) < .001 2.49 (1.49-4.15) 
    Chronic myeloid leukemia 26 (29) 29 (8) < .001 4.48 (2.37-8.48) 
    Multiple myeloma 1 (1) 52 (15) < .001 0.06 (0.02-0.43) 
Neoplastic disease, no. (%) 79 (90) 316 (93) .27 0.64 (0.28-1.43) 
Advanced underlying disease, no. (%) 53 (60) 262 (77) .001 0.44 (0.27-0.73) 
Allogeneic HSC transplantation, no. (%) 82 (93) 164 (48) < .001 14.60 (6.20-34.30) 
Unrelated donor allogeneic HSC transplantation, no. (%) 15/82 (18) 46/164 (28) .15 1.31 (0.69-2.47) 
Matched-related donor allogeneic HSC transplantation, no. (%) 79/82 (96) 136/164 (83) .003 5.42 (1.60-18.40) 
History of hepatitis, no. (%) 4 (4) 3 (1) .04 5.33 (1.17-24.30) 
CMV-positive recipient, no. (%) 78 (89) 298 (88) .85 1.07 (0.51-2.24) 
Previous HSC transplantation, no. (%) 5 (6) 35 (10) .18 0.52 (0.20-1.38) 
Receipt of imatinib, no. (%) 15 (17) 14 (4) < .001 4.77 (2.21-10.30) 
Receipt of gemtuzumab, no. (%) 1 (1) .21 NA 
Receipt of vancomycin or acyclovir, no. (%) 2 (2) 7 (2) .999 1.10 (0.22-5.40) 
Busulfan-based conditioning regimen, no. (%) 46 (52) 73 (21) < .001 3.99 (2.44-6.53) 
TBI-based conditioning regimen, no. (%) 23 (26) 62 (18) .10 1.58 (0.91-2.74) 
Myeloablative HSC transplantation, no. (%) 81 (92) 308 (91) .73 1.16 (0.49-2.74) 
Bone marrow as the source of stem cells, no. (%) 60 (68) 108 (32) < .001 4.58 (2.69-7.83) 
Methotrexate in the prophylaxis for GVHD, no. (%) 76/82 (93) 142/164 (86) .13 1.96 (0.76-5.05) 
Hepatic dysfunction, no. (%) 26 (29) 94 (28) .73 1.09 (0.65-1.83) 
SOS
POdds ratio (95% CI)
Yes n=88No n=339
Male:female 49:39 206:133 .38 0.81 (0.50-1.30) 
Median age, y (range) 34.5 (6-33) 30 (0-66) .62 NA 
Underlying disease, no. (%)     
    Lymphoma 7 (8) 135 (40) < .001 0.13 (0.06-0.30) 
    Acute leukemia/MDS 41 (46) 88 (26) < .001 2.49 (1.49-4.15) 
    Chronic myeloid leukemia 26 (29) 29 (8) < .001 4.48 (2.37-8.48) 
    Multiple myeloma 1 (1) 52 (15) < .001 0.06 (0.02-0.43) 
Neoplastic disease, no. (%) 79 (90) 316 (93) .27 0.64 (0.28-1.43) 
Advanced underlying disease, no. (%) 53 (60) 262 (77) .001 0.44 (0.27-0.73) 
Allogeneic HSC transplantation, no. (%) 82 (93) 164 (48) < .001 14.60 (6.20-34.30) 
Unrelated donor allogeneic HSC transplantation, no. (%) 15/82 (18) 46/164 (28) .15 1.31 (0.69-2.47) 
Matched-related donor allogeneic HSC transplantation, no. (%) 79/82 (96) 136/164 (83) .003 5.42 (1.60-18.40) 
History of hepatitis, no. (%) 4 (4) 3 (1) .04 5.33 (1.17-24.30) 
CMV-positive recipient, no. (%) 78 (89) 298 (88) .85 1.07 (0.51-2.24) 
Previous HSC transplantation, no. (%) 5 (6) 35 (10) .18 0.52 (0.20-1.38) 
Receipt of imatinib, no. (%) 15 (17) 14 (4) < .001 4.77 (2.21-10.30) 
Receipt of gemtuzumab, no. (%) 1 (1) .21 NA 
Receipt of vancomycin or acyclovir, no. (%) 2 (2) 7 (2) .999 1.10 (0.22-5.40) 
Busulfan-based conditioning regimen, no. (%) 46 (52) 73 (21) < .001 3.99 (2.44-6.53) 
TBI-based conditioning regimen, no. (%) 23 (26) 62 (18) .10 1.58 (0.91-2.74) 
Myeloablative HSC transplantation, no. (%) 81 (92) 308 (91) .73 1.16 (0.49-2.74) 
Bone marrow as the source of stem cells, no. (%) 60 (68) 108 (32) < .001 4.58 (2.69-7.83) 
Methotrexate in the prophylaxis for GVHD, no. (%) 76/82 (93) 142/164 (86) .13 1.96 (0.76-5.05) 
Hepatic dysfunction, no. (%) 26 (29) 94 (28) .73 1.09 (0.65-1.83) 

SOS indicates sinusoidal obstruction syndrome; 95% CI, 95% confidence interval; NA, not applicable; MDS, myelodysplasia; HSC, hematopoietic stem cell; CMV, cytomegalovirus; TBI, total body irradiation; GVHD, graft-versus-host disease; and TPN, total parenteral nutrition.

A total of 305 patients (71%) had serum levels of ferritin above the normal range, with a median value of 622 ng/mL (range, 5.81-7785 ng/mL). The median ferritin was higher in patients who developed SOS (917.5 vs 531, P = .002). In the assessment of risk factors for SOS, ferritin was analyzed as a categoric variable with different cutoff points. Although different cutoff values were significantly associated with the development of SOS, a value of 1000 ng/mL showed the best sensitivity (48%) and specificity (71%), according to the ROC curve analysis (Table 3). Patients with ferritin level higher than 1000 ng/mL were more likely to develop SOS, with an OR of 2.49 (95% CI, 1.54-4.02). High ferritin levels (> 1000 ng/mL) were also strongly associated with lower 5-year overall survival (42% vs 73% for patients with ferritin ≤ 1000 ng/mL, P < .001, log-rank test).

Table 3

Univariate analysis of different cutoff values of serum ferritin and the risk for sinusoidal obstruction syndrome

Ferritin serum level, ng/mLSOS, no. (%)
POdds ratio (95% CI)ROC curve analysis
Yes n=88No n=339AUCP*
623 (median) 54 (61) 159 (47) .02 1.80 (1.08-2.99) 0.572 .04 
1000 43 (49) 94 (28) < .001 2.49 (1.50-4.14) 0.606 .002 
1500 30 (34) 56 (16) < .001 2.61 (1.49-4.57) 0.588 .01 
2000 21 (24) 41 (12) .005 2.28 (1.21-4.27) 0.559 .09 
3000 11 (12) 14 (4) .003 3.32 (1.34-8.13) 0.542 .23 
4000 7 (8) 7 (2) .01 4.10 (1.25-13.44) 0.529 .39 
Ferritin serum level, ng/mLSOS, no. (%)
POdds ratio (95% CI)ROC curve analysis
Yes n=88No n=339AUCP*
623 (median) 54 (61) 159 (47) .02 1.80 (1.08-2.99) 0.572 .04 
1000 43 (49) 94 (28) < .001 2.49 (1.50-4.14) 0.606 .002 
1500 30 (34) 56 (16) < .001 2.61 (1.49-4.57) 0.588 .01 
2000 21 (24) 41 (12) .005 2.28 (1.21-4.27) 0.559 .09 
3000 11 (12) 14 (4) .003 3.32 (1.34-8.13) 0.542 .23 
4000 7 (8) 7 (2) .01 4.10 (1.25-13.44) 0.529 .39 

SOS indicates sinusoidal obstruction syndrome; 95% CI, 95% confidence interval; ROC, receiver operating characteristic; and AUC, area under the curve.

*

Probability that the area is different from .5.

As shown in Table 4, by multivariate analysis, the following variables were associated with SOS: allogeneic HSC transplantation (OR = 8.25; 95% CI, 3.31-20.57; P < .001), receipt of imatinib before HSC transplantation (OR = 2.60; 95% CI, 1.16-5.84; P = .02), receipt of busulfan in the conditioning regimen (OR = 2.18; 95% CI, 1.25-3.80; P = .006), and serum ferritin level higher than 1000 ng/mL (OR = 1.78; 95% CI, 1.02-3.08; P = .04).

Table 4

Factors associated with sinusoidal obstruction syndrome by multivariate analysis

VariableOdds ratio95% CIP
Allogeneic HSC transplantation 8.25 3.31-20.57 < .001 
Receipt of imatinib before HSC transplantation 2.60 1.16-5.84 .02 
Receipt of busulfan in the conditioning regimen 2.18 1.25-3.80 .006 
Serum ferritin level higher than 1000 ng/mL 1.78 1.02-3.08 .04 
VariableOdds ratio95% CIP
Allogeneic HSC transplantation 8.25 3.31-20.57 < .001 
Receipt of imatinib before HSC transplantation 2.60 1.16-5.84 .02 
Receipt of busulfan in the conditioning regimen 2.18 1.25-3.80 .006 
Serum ferritin level higher than 1000 ng/mL 1.78 1.02-3.08 .04 

95% CI indicates 95% confidence interval; and HSC, hematopoietic stem cell.

Because most of the cases of SOS occurred in allogeneic HSCT recipients (82 of the 88 cases), we ran an analysis of risk factors in these patients. Multivariate predictors of SOS were receipt of imatinib before HSC transplantation (OR = 2.16; 95% CI, 1.22-3.82; P = .01), receipt of busulfan in the conditioning regimen (OR = 2.22; 95% CI, 1.26-3.89; P = .005), and serum ferritin level higher than 1000 ng/mL (OR = 2.16; 95% CI, 1.22-3.82; P = .008).

Our results show that high ferritin serum level is an independent pretransplantation risk factor for SOS. Other risk factors identified were allogeneic HSC transplantation, and receipt of imatinib before transplantation and busulfan in the conditioning regimen.

Studies evaluating the impact of iron overload before HSC transplantation on different outcomes have been increasingly reported. Miceli et al observed that iron overload, assessed by bone marrow biopsy, was an independent risk factor for severe infection among patients with multiple myeloma receiving autologous HSCT conditioned with melphalan 200 mg/m2.10  In another study, high serum ferritin level was associated with a higher incidence ofrelapse and relapse-associated mortality among 315 patients with lymphoma who received autologous HSCT.14  Kataoka et al reported that high pretransplantation serum ferritin was associated with higher 5-year mortality among allogeneic HSCT recipients. The increase in death was attributed to organ failure (cardiac, renal, hepatic, and multiorgan) and infection.15  An association between high pretransplantation ferritin serum levels and higher mortality was also reported in a study of 190 allogeneic HSCT recipients. In this study, patients with iron overload were more likely to die by day +100, both from GVHD and bloodstream infections.16  In another study with a larger number of HSCT recipients, an elevated pretransplantation serum ferritin level was associated with lower overall and disease-free survival, especially in patients with acute leukemia and myelodysplasia.17  We also observed an association between iron overload and higher 5-year mortality. It is not clear if iron overload per se contributes to the poor survival or if it is merely a surrogate marker of poor prognosis. Other studies are needed to explore these possibilities.

The association between high iron stores and SOS was reported by Morado et al in a study in 180 autologous HSCT recipients (mostly breast cancer and lymphoma). Diagnostic criteria for SOS were similar to ours. The incidence of SOS was 12.2%. By multivariate analysis, serum ferritin level more than 300 ng/dL remained significant as a risk factor for SOS.12  A trend toward an association between high ferritin serum levels and the occurrence of SOS was reported in another study.17 

In the present study, we confirm the association between high ferritin serum levels before transplantation and the occurrence of SOS, in a large cohort of both autologous and allogeneic HSCT recipients. Our patient's population was representative of a typical HSC transplantation center, and the period of study was relatively recent (between 2002 and 2007).

An association between iron overload and liver injury has been reported in different clinical scenarios. For example, in a study of 76 survivors of allogeneic HSC transplantation for at least 1 year, 88% had high ferritin serum levels. Impaired liver function was commonly observed among these patients, and improved with iron chelation.18  Iron-induced hepatotoxicity is multifactorial, and involves oxidative stress and modulation of gene expression of Kupffer cells.19  Cellular injury is induced by iron-generated oxyradicals and peroxidation of lipid membranes.20  However, the mechanism by which iron overload is associated with SOS remains poorly understood.

In our study, other risk factors for SOS were allogeneic HSC transplantation, and the exposure to busulfan and imatinib. The association between allogeneic HSC transplantation and SOS was reported in other studies.21,22  Oral busulfan in association with cyclophosphamide in the conditioning regimen is associated with liver toxicity and SOS.22-24  By contrast, regimens containing busulfan in combination with other drugs, such as melphalan and thiotepa, seem not to be associated with liver toxicity.24-26  In the present study, the overwhelming majority of patients received oral busulfan followed by cyclophosphamide as the conditioning regimen.

Busulfan is eliminated through glutathione conjugation, and therefore its metabolism consumes glutathione. The depletion of glutathione in the liver seems to play a major role in the pathogenesis of SOS, because many drugs that potentially cause injury to the endothelium (an early and key event in the development of SOS) are metabolized through the glutathione pathway.27 

Pharmacogenetic profiles seem also to play a role in chemotherapy-induced liver injury and SOS. In a study in 84 adult allogeneic HSCT recipients who received busulfan and cyclophosphamide as part of the conditioning regimen, genetic polymorphisms of the methylene-tetrahydrofolate reductase (MTHFR) were evaluated by a polymerase chain reaction (PCR) technique. Multivariate analysis identified one polymorphism (MTHFR-1298A>C) as an independent predictor of hyperbilirubinemia and SOS.28 

We observed that the use of imatinib before transplantation was associated with an increased risk of SOS. To our knowledge this is the first report of this association. Imatinib mesylate is the first tyrosine kinase inhibitor introduced in clinical practice. It is effective in the treatment of chronic myeloid leukemia, Philadelphia-positive acute lymphoid leukemia, and gastrointestinal stromal cell tumors. Imatinib is metabolized in the liver, using cytochrome P-450 (CYP) 3A4 enzyme.29  CYP3A4 inducers, such as phenitoin, reduce serum concentration of imatinib, whereas CYP3A4 inhibitors (such as ketoconazole and some food products) increase significantly imatinib serum levels. Grades III-IV hepatic toxicity include elevations in transaminases in 1% to 3%, and hyperbilirubinemia in 0.4% to 3.5%.29  In addition, reports of liver toxicity of imatinib have been published.30-35  Liver toxicity usually occurs after more than 3 months of treatment, and drug interruption reverses the toxicity in the large majority of cases. The mechanisms of liver toxicity are unknown. Histology shows a hepatocellular pattern of injury, with mild cholestasis.36  It is possible that liver toxicity of imatinib may predispose to toxic damage from the conditioning regimen and the subsequent development of SOS. However, further studies are needed, because the relationship between imatinib use and SOS was based on a small group of patients.

We used serum ferritin as a measure of iron stores. Other measures to assess iron stores include x-ray fluorescence of iron, magnetic resonance imaging, computed tomography, biomagnetic susceptometry,37  and iron staining in biopsy sections of the bone marrow and the liver.38,39  Serum ferritin may be increased in conditions other than iron overload, including chronic inflammation and infection.40  Nevertheless, serum ferritin is considered a valuable method for hepatic iron evaluation.41  Furthermore, values higher than 1000 ng/mL (the cutoff value that we used) are rarely reported in these conditions. Therefore, we think that our pretransplantation estimation of iron overload is reliable. Even if we consider the potential limitations of serum ferritin in the estimation of iron stores, a pretransplantation value higher than 1000 ng/mL (irrespective of iron stores) is still a valid risk factor for SOS.

Our study is limited by its retrospective nature. Therefore, other potentially important variables, such as duration of imatinib therapy before transplantation and exposure to other drugs that may increase liver toxicity, were not evaluated. Another limitation ofretrospective studies, the absence of important data, was not a problem in our study, because no patient was excluded due to missing data.

The results of the present study and of other recent reports argue in favor of the development of future studies to investigate the usefulness of prophylactic and therapeutic measures aimed at reducing the negative effects of iron overload on the outcome of HSCT recipients. Pretransplantation iron chelation in patients with ferritin levels higher than 1000 ng/mL would be a reasonable measure to be explored. Another strategy to reduce the incidence of SOS would be to change the way busulfan is administered. The use of intravenous (instead of oral) busulfan may decrease the risk of SOS by avoiding a hepatic first-pass extraction effect that results in high concentration of busulfan in the portal-hepatic venous system.42  Finally, clinicians should be aware of the potential of chronic use of imatinib before transplantation to increase the risk of SOS. However, as mentioned, further studies are needed to confirm our findings.

In summary, our study identified high ferritin serum level, allogeneic HSC transplantation, and receipt of imatinib pretransplantation and busulfan in the conditioning regimen as independent risk factors for SOS.

An Inside Blood analysis of this article appears at the front of this issue.

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.

We thank Karla Gonçalves Camacho for helping in the organization of data and patients' charts.

This study was supported by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brazil, grants 301025/2008-8 (M.N.), and 301455/2006-6 (A.M.).

Contribution: S.C.M. designed and performed the research, analyzed the data, and wrote the paper; A.M. designed the research and reviewed the paper; A.M.d.A. analyzed the data and wrote the paper; M.C. collected and reviewed the data; L.F.B. reviewed the paper; and M.N. analyzed the data and wrote the paper.

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

Correspondence: Marcio Nucci, University Hospital, Federal University of Rio de Janeiro, Rua Professor Rodolpho Paulo Rocco 255, Sala 4A 12, Rio de Janeiro 21941-913, Brazil; e-mail: mnucci@hucff.ufrj.br.

1
Barker
 
CC
Anderson
 
RA
Sauve
 
RS
Butzner
 
JD
GI complications in pediatric patients post-BMT.
Bone Marrow Transplant
2005
, vol. 
36
 (pg. 
51
-
58
)
2
Hasegawa
 
S
Horibe
 
K
Kawabe
 
T
, et al. 
Veno-occlusive disease of the liver after allogeneic bone marrow transplantation in children with hematologic malignancies: incidence, onset time and risk factors.
Bone Marrow Transplant
1998
, vol. 
22
 (pg. 
1191
-
1197
)
3
Kumar
 
S
DeLeve
 
LD
Kamath
 
PS
Tefferi
 
A
Hepatic veno-occlusive disease (sinusoidal obstruction syndrome) after hematopoietic stem cell transplantation.
Mayo Clin Proc
2003
, vol. 
78
 (pg. 
589
-
598
)
4
Carreras
 
E
Veno-occlusive disease of the liver after hemopoietic cell transplantation.
Eur J Haematol
2000
, vol. 
64
 (pg. 
281
-
291
)
5
Cesaro
 
S
Pillon
 
M
Talenti
 
E
, et al. 
A prospective survey on incidence, risk factors and therapy of hepatic veno-occlusive disease in children after hematopoietic stem cell transplantation.
Haematologica
2005
, vol. 
90
 (pg. 
1396
-
1404
)
6
Kalayoglu-Besisik
 
S
Yenerel
 
MN
Caliskan
 
Y
, et al. 
Time-related changes in the incidence, severity, and clinical outcome of hepatic veno-occlusive disease in hematopoietic stem cell transplantation patients during the past 10 years.
Transplant Proc
2005
, vol. 
37
 (pg. 
2285
-
2289
)
7
Ljungman
 
P
Hassan
 
M
Bekassy
 
AN
Ringden
 
O
Oberg
 
G
High busulfan concentrations are associated with increased transplant-related mortality in allogeneic bone marrow transplant patients.
Bone Marrow Transplant
1997
, vol. 
20
 (pg. 
909
-
913
)
8
McKoy
 
JM
Angelotta
 
C
Bennett
 
CL
, et al. 
Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project.
Leuk Res
2007
, vol. 
31
 (pg. 
599
-
604
)
9
Platzbecker
 
U
von Bonin
 
M
Goekkurt
 
E
, et al. 
Graft-versus-host disease prophylaxis with everolimus and tacrolimus is associated with a high incidence of sinusoidal obstruction syndrome and microangiopathy: results of the EVTAC trial.
Biol Blood Marrow Transplant
2009
, vol. 
15
 (pg. 
101
-
108
)
10
Miceli
 
MH
Dong
 
L
Grazziutti
 
ML
, et al. 
Iron overload is a major risk factor for severe infection after autologous stem cell transplantation: a study of 367 myeloma patients.
Bone Marrow Transplant
2006
, vol. 
37
 (pg. 
857
-
864
)
11
Altès
 
A
Remacha
 
AF
Sureda
 
A
, et al. 
Iron overload might increase transplant-related mortality in haematopoietic stem cell transplantation.
Bone Marrow Transplant
2002
, vol. 
29
 (pg. 
987
-
989
)
12
Morado
 
M
Ojeda
 
E
Garcia-Bustos
 
J
, et al. 
BMT: serum ferritin as risk factor for veno-occlusive disease of the liver: prospective cohort study.
Hematology
2000
, vol. 
4
 (pg. 
505
-
512
)
13
McDonald
 
GB
Sharma
 
P
Matthews
 
DE
Shulman
 
HM
Thomas
 
ED
Venocclusive disease of the liver after bone marrow transplantation: diagnosis, incidence, and predisposing factors.
Hepatology
1984
, vol. 
4
 (pg. 
116
-
122
)
14
Mahindra
 
A
Bolwell
 
B
Sobecks
 
R
, et al. 
Elevated ferritin is associated with relapse after autologous hematopoietic stem cell transplantation for lymphoma.
Biol Blood Marrow Transplant
2008
, vol. 
14
 (pg. 
1239
-
1244
)
15
Kataoka
 
K
Nannya
 
Y
Hangaishi
 
A
, et al. 
Influence of pretransplantation serum ferritin on nonrelapse mortality after myeloablative and nonmyeloablative allogeneic hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 (pg. 
195
-
204
)
16
Pullarkat
 
V
Blanchard
 
S
Tegtmeier
 
B
, et al. 
Iron overload adversely affects outcome of allogeneic hematopoietic cell transplantation.
Bone Marrow Transplant
2008
, vol. 
42
 (pg. 
799
-
805
)
17
Armand
 
P
Kim
 
HT
Cutler
 
CS
, et al. 
Prognostic impact of elevated pretransplantation serum ferritin in patients undergoing myeloablative stem cell transplantation.
Blood
2007
, vol. 
109
 (pg. 
4586
-
4588
)
18
McKay
 
PJ
Murphy
 
JA
Cameron
 
S
, et al. 
Iron overload and liver dysfunction after allogeneic or autologous bone marrow transplantation.
Bone Marrow Transplant
1996
, vol. 
17
 (pg. 
63
-
66
)
19
Videla
 
LA
Fernandez
 
V
Tapia
 
G
Varela
 
P
Oxidative stress-mediated hepatotoxicity of iron and copper: role of Kupffer cells.
Biometals
2003
, vol. 
16
 (pg. 
103
-
111
)
20
Ramm
 
GA
Ruddell
 
RG
Hepatotoxicity of iron overload: mechanisms of iron-induced hepatic fibrogenesis.
Semin Liver Dis
2005
, vol. 
25
 (pg. 
433
-
449
)
21
European Group for Blood and Marrow Transplantation Chronic Leukemia Working Party
Carreras
 
E
Bertz
 
H
Arcese
 
W
, et al. 
Incidence and outcome of hepatic veno-occlusive disease after blood or marrow transplantation: a prospective cohort study of the European Group for Blood and Marrow Transplantation.
Blood
1998
, vol. 
92
 (pg. 
3599
-
3604
)
22
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
, vol. 
118
 (pg. 
255
-
267
)
23
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
 (pg. 
853
-
862
)
24
Dix
 
SP
Wingard
 
JR
Mullins
 
RE
, et al. 
Association of busulfan area under the curve with veno-occlusive disease following BMT.
Bone Marrow Transplant
1996
, vol. 
17
 (pg. 
225
-
230
)
25
Grochow
 
LB
Busulfan disposition: the role of therapeutic monitoring in bone marrow transplantation induction regimens.
Semin Oncol
1993
, vol. 
20
 (pg. 
18
-
25
)
26
Slattery
 
JT
Sanders
 
JE
Buckner
 
CD
, et al. 
Graft-rejection and toxicity following bone marrow transplantation in relation to busulfan pharmacokinetics.
Bone Marrow Transplant
1995
, vol. 
16
 (pg. 
31
-
42
)
27
DeLeve
 
LD
Shulman
 
HM
McDonald
 
GB
Toxic injury to hepatic sinusoids: sinusoidal obstruction syndrome (veno-occlusive disease).
Semin Liver Dis
2002
, vol. 
22
 (pg. 
27
-
42
)
28
Goekkurt
 
E
Stoehlmacher
 
J
Stueber
 
C
, et al. 
Pharmacogenetic analysis of liver toxicity after busulfan/cyclophosphamide-based allogeneic hematopoietic stem cell transplantation.
Anticancer Res
2007
, vol. 
27
 (pg. 
4377
-
4380
)
29
Cohen
 
MH
Williams
 
G
Johnson
 
JR
, et al. 
Approval summary for imatinib mesylate capsules in the treatment of chronic myelogenous leukemia.
Clin Cancer Res
2002
, vol. 
8
 (pg. 
935
-
942
)
30
Ridruejo
 
E
Cacchione
 
R
Villamil
 
AG
, et al. 
Imatinib-induced fatal acute liver failure.
World J Gastroenterol
2007
, vol. 
13
 (pg. 
6608
-
111
)
31
Kong
 
JH
Yoo
 
SH
Lee
 
KE
, et al. 
Early imatinib-mesylate-induced hepatotoxicity in chronic myelogenous leukaemia.
Acta Haematol
2007
, vol. 
118
 (pg. 
205
-
208
)
32
Ramamoorthy
 
SK
Pandita
 
R
Prakash
 
A
Ramaswamy
 
NV
Al
 
BS
Safety of imatinib in chronic myeloid leukemia in blastic crisis presenting as cholestatic jaundice.
Acta Haematol
2007
, vol. 
118
 (pg. 
141
-
145
)
33
Ferrero
 
D
Pogliani
 
EM
Rege-Cambrin
 
G
, et al. 
Corticosteroids can reverse severe imatinib-induced hepatotoxicity.
Haematologica
2006
, vol. 
91
 
6
suppl
pg. 
ECR27
 
34
Pariente
 
A
Etcharry
 
F
Cales
 
V
, et al. 
Imatinib mesylate-induced acute hepatitis in a patient treated for gastrointestinal stromal tumour.
Eur J Gastroenterol Hepatol
2006
, vol. 
18
 (pg. 
785
-
787
)
35
Ohyashiki
 
K
Kuriyama
 
Y
Nakajima
 
A
, et al. 
Imatinib mesylate-induced hepato-toxicity in chronic myeloid leukemia demonstrated focal necrosis resembling acute viral hepatitis.
Leukemia
2002
, vol. 
16
 (pg. 
2160
-
2161
)
36
Cross
 
TJ
Bagot
 
C
Portmann
 
B
Wendon
 
J
Gillett
 
D
Imatinib mesylate as a cause of acute liver failure.
Am J Hematol
2006
, vol. 
81
 (pg. 
189
-
192
)
37
Brittenham
 
GM
Badman
 
DG
Noninvasive measurement of iron: report of an NIDDK workshop.
Blood
2003
, vol. 
101
 (pg. 
15
-
19
)
38
Brittenham
 
GM
Danish
 
EH
Harris
 
JW
Assessment of bone marrow and body iron stores: old techniques and new technologies.
Semin Hematol
1981
, vol. 
18
 (pg. 
194
-
221
)
39
Pippard
 
MJ
Measurement of iron status.
Prog Clin Biol Res
1989
, vol. 
309
 (pg. 
85
-
92
)
40
Jensen
 
PD
Evaluation of iron overload.
Br J Haematol
2004
, vol. 
124
 (pg. 
697
-
711
)
41
Brissot
 
P
Bourel
 
M
Herry
 
D
, et al. 
Assessment of liver iron content in 271 patients: a reevaluation of direct and indirect methods.
Gastroenterology
1981
, vol. 
80
 (pg. 
557
-
565
)
42
Ciurea
 
SO
Andersson
 
BS
Busulfan in hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 (pg. 
523
-
536
)
Sign in via your Institution