Skip to main content
Erschienen in: Journal of Cancer Research and Clinical Oncology 1/2024

Open Access 01.01.2025 | Research

Analysis of laboratory parameters before the occurrence of hepatic sinusoidal obstruction syndrome in children, adolescents, and young adults after hematopoietic stem cell transplantation

verfasst von: Lorena Johann, Bernd Gruhn

Erschienen in: Journal of Cancer Research and Clinical Oncology | Ausgabe 1/2024

Abstract

Purpose

Hepatic sinusoidal obstruction syndrome (SOS) is a serious complication following hematopoietic stem cell transplantation (HSCT) in which early diagnosis improves patient outcome. The aim of our study was to detect laboratory parameters following HSCT that can predict the occurrence of SOS.

Methods

This retrospective study included 182 children, adolescents, and young adults who underwent allogeneic or autologous HSCT for the first time (median age 7.2 years). The diagnosis of SOS was based on the pediatric criteria of European Society for Blood and Marrow Transplantation (EBMT). We investigated 15 laboratory parameters after HSCT before the onset of SOS.

Results

The overall incidence of SOS was 14.8%. SOS developed in 24 of 126 allogeneic (19.1%) and in 3 of 56 autologous (5.4%) HSCT patients at a median time of 13 days after HSCT. We observed a low SOS mortality rate of 11.1% within 100 days after HSCT. International normalized ratio (INR) ≥ 1.3, activated partial thromboplastin time (aPTT) ≥ 40 s, reptilase time ≥ 18.3 s, factor VIII ≤ 80%, antithrombin III ≤ 75%, protein C ≤ 48%, D-dimer ≥ 315 µg/L, bilirubin ≥ 9 µmol/L, and ferritin ≥ 3100 µg/L showed significant associations with the onset of SOS in the univariate analyses. In the multivariate analysis, INR ≥ 1.3 [odds ratio (OR) = 8.104, p = 0.006], aPTT ≥ 40 s (OR = 10.174, p = 0.001), protein C ≤ 48% (OR = 5.215, p = 0.014), and ferritin ≥ 3100 µg/L (OR = 7.472, p = 0.004) could be confirmed as independent risk factors after HSCT before SOS. If three of the four significant cut-off values were present, the probability of developing SOS was more than 70%. The probability of SOS was 96%, if all four laboratory parameters were changed according to the cut-off values. The values of factor XIII, von Willebrand factor (vWF), von Willebrand factor activity (vWF activity), protein S, fibrinogen, and alanine aminotransferase (ALT) were not relevant for the occurrence of SOS.

Conclusion

In summary, the laboratory parameters INR, aPTT, protein C, and ferritin were very useful to predict the occurrence of SOS. In addition, this is the first report on a significant association between SOS and high values of INR and aPTT after HSCT before SOS.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Hepatic sinusoidal obstruction syndrome (SOS), also known as veno-occlusive disease, is a serious complication after hematopoietic stem cell transplantation (HSCT), especially in children. The incidence of SOS following HSCT depends on diagnostic criteria, type of HSCT and patient age. Therefore the incidence varies in several publications. The median incidence of SOS is 14% and ranges from 0 to 60% in the published reports (Coppell et al. 2010; Xia et al. 2021). Following allogeneic HSCT the frequency of SOS is higher (12.9%) than after autologous HSCT (8.7%) (Coppell et al. 2010). A recent study by Coutsouvelis et al. (2023) reported an incidence of 11.5% in pediatric patients and 4.1% in adults. Further studies confirmed higher incidences in children (20%) than in adults (10%) (Barker et al. 2003; Cesaro et al. 2005; Corbacioglu et al. 2012, 2018).
Most patients with SOS suffer from hepatomegaly, ascites, and weight gain (Corbacioglu et al. 2018). There are several clinical diagnostic criteria used for SOS. Diagnostic standards are based on the Baltimore criteria (Jones et al. 1987) or the Seattle criteria (McDonald et al. 1984). In addition, new pediatric diagnostic criteria were published on behalf of the European Society for Blood and Marrow Transplantation (EBMT), stating that hyperbilirubinemia is a non-mandatory criterion and there is no time limit for the diagnosis of SOS (Corbacioglu et al. 2018). Furthermore, the pediatric EBMT criteria can be used for grading the severity of SOS, which consists of mild, moderate, severe, and very severe SOS (Corbacioglu et al. 2018).
Yoon et al. (2019) concluded an increased mortality of 36.7% within 100 days after HSCT in very severe SOS. Furthermore, Coppell et al. (2010) reported a mortality rate more than 80% in patients with the most severe SOS. Defibrotide is an effective therapy for SOS, which can improve survival (Mohty et al. 2022; Richardson et al. 2016). Mohty et al. (2022) reported better outcomes in patients with severe versus very severe SOS, suggesting early diagnosis and treatment is important before patients develop very severe SOS. In addition, defibrotide prophylaxis can reduce the incidence of SOS and is well tolerated by patients (Corbacioglu et al. 2012).
Different factors, such as conditioning therapy, endotoxins, cytokines, and certain drugs lead to endothelial imbalance. This results in a loss of sinusoidal endothelial cell fenestration and destruction of endothelial barrier. Erythrocytes penetrate into the space of Disse and damaged sinusoids induce vascular obstruction downstream (Carreras and Diaz-Ricart 2011; DeLeve et al. 2002). The reduced blood flow to the liver leads to liver damage (DeLeve et al. 2002). In addition, fibrinogen and factor VIII within the subendothelial zones can also lead to thrombosis of the vessels (Shulmann et al. 1987). Moreover, an exposure of tissue factors and initiation of the coagulation cascade, which is followed by reduced synthesis of coagulation factors, causes hemostatic imbalance (DeLeve et al. 2002; Nürnberger et al. 1997). Due to the pathogenesis, it is obvious that laboratory parameters, such as coagulation, fibrinolytic, and liver parameters are changed. An increase of D-dimer, bilirubin, and alanine aminotransferase (ALT) as well as a decrease of antithrombin III and protein C were observed in patients with SOS (Jevtić et al. 2011; Lee et al. 1998; Sartori et al. 2012).
The purpose of our retrospective study was to investigate laboratory parameters that can predict the development of SOS. Therefore, we analyzed coagulation, fibrinolytic, and liver parameters following HSCT before SOS.

Methods

Patients

Our study population consisted of 182 children, adolescents, and young adults who underwent HSCT at the Department of Pediatrics, Jena University Hospital, Germany between 2007 and 2022. The patients received allogeneic or autologous HSCT for the first time. We excluded patients with defibrotide prophylaxis and more than one HSCT, respectively. The conditioning therapy was always myeloablative and dependent on the type of HSCT and underlying disease. In addition, the transplanted patients underwent an infection prophylaxis and were isolated in rooms with special laminar airflow filtration systems.

SOS

The pediatric EBMT criteria were used to diagnose SOS (Corbacioglu et al. 2018).
Accordingly, there is no time limit of onset and SOS is diagnosed if two or more of the following criteria are present: transfusion-refractory thrombocytopenia, unexplained weight gain > 5%, or weight gain on three consecutive days despite diuretics, ascites, hepatomegaly, bilirubin ≥ 34 µmol/L within 72 h, or an increase in bilirubin on three consecutive days. The classification of the severity of SOS was also based on the pediatric severity EBMT criteria (Corbacioglu et al. 2018). The patients with SOS received 25 mg/kg/day defibrotide in four single doses for 30 days.

Risk factors

In our study, we considered the following 15 laboratory parameters: international normalized ratio (INR), activated partial thromboplastin time (aPTT), reptilase time, factor VIII, factor XIII, von Willebrand factor (vWF), von Willebrand factor activity (vWF activity), fibrinogen, antithrombin III, protein C, protein S, D-dimer, ALT, bilirubin, and ferritin. Laboratory parameters were measured weekly following HSCT. In the SOS patient group, the last laboratory values within one week before the onset of SOS were analyzed. We investigated the parameters for four weeks after transplantation in the non-SOS patient group and calculated the median.

Statistical analysis

A retrospective analysis of potential risk factors was performed. We always considered results with p values less than 0.05 as statistical significant. Differences in the laboratory values between the SOS and the non-SOS patient group were analyzed using the Mann–Whitney U test. We performed univariate and multivariate analyses to find significant associations between the values of laboratory parameters and the occurrence of SOS. The results were presented with corresponding p value (p), odds ratio (OR), and 95% confidence interval (CI). Cut-off values were selected for the continuous parameters based on existing reference values and receiver operating characteristic (ROC) curves analyses. We divided the values according to the corresponding cut-off values and performed the univariate analyses by Fisher's exact test for nominal parameters. The significant variables were entered as independent variables into multivariate analysis, which carried out by binary logistic regression and backward elimination. Using the significant independent variables, we calculated the probability of developing SOS. All calculations were performed using the software IBM SPSS Statistics 27.

Results

Patient characteristics

This retrospective analysis included 182 patients (109 males and 73 females) with a median age of 7.2 years (ranged from 2 months to 26 years). Our study population consisted of 126 patients who underwent allogeneic HSCT (69.2%) and 56 patients who received autologous HSCT (30.8%). The most frequent underlying diseases were solid tumors (34.1%), genetic diseases (19.2%), acute lymphoblastic leukemia (18.1%), and acute myeloid leukemia (13.7%). Peripheral blood (50.5%) or bone marrow (49.5%) was used as stem cell source. Table 1 summarizes the characteristics of the patients.
Table 1
Characteristics of patients
Characteristics
No. (%)
Patients
182 (100)
Sex
 Male
109 (59.9)
 Female
73 (40.1)
Median age, years (range)
7.2 (0.2–26.2)
Type of HSCT
 Allogeneic HSCT
126 (69.2)
 Autologous HSCT
56 (30.8)
Stem cell source
 Peripheral blood
92 (50.5)
 Bone marrow
90 (49.5)
Patient’s diagnoses
 Solid tumor
62 (34.1)
 Genetic diseases
35 (19.2)
 Acute lymphoblastic leukemia
33 (18.1)
 Acute myeloid leukemia
25 (13.7)
 Myelodysplastic syndrome
16 (8.8)
 Lymphoma
9 (5.0)
 Chronic myeloid leukemia
2 (1.1)
Conditioning regimen (based on)
 Chemotherapy
157 (86.3)
 Total body irradiation
25 (13.7)
GVHD prophylaxis—allogeneic HSCT patients
 Cyclosporine A/methotrexate
55 (43.6)
 Cyclosporine A
35 (27.8)
 Others
36 (28.6)
SOS diagnosis
 Yes
27 (14.8)
 No
155 (85.2)
SOS severity
 Mild
2 (7.4)
 Moderate
7 (25.9)
 Severe
8 (29.6)
 Very severe
10 (37.1)
HSCT hematopoietic stem cell transplantation, GVHD graft-versus-host disease, No. number, SOS sinusoidal obstruction syndrome
A total of 27 patients (14.8%) developed SOS. In the allogeneic patient group, SOS was diagnosed in 24 of 126 patients, which corresponds to a frequency of 19.1%. The SOS frequency was lower in the autologous patient group, 3 of 56 patients developed SOS (5.4%). The time period between HSCT and the onset of SOS ranged from 1 to 28 days (median 13 days). Two patients (7.4%) showed a mild SOS and 7 patients (25.9%) a moderate SOS. Furthermore, a severe SOS occurred in 8 patients (29.6%). The remaining 10 patients (37.1%) developed a very severe SOS. Only 3 out of 27 patients, who suffered from very severe SOS, died within 100 days after HSCT (11.1%). Six out of 155 patients without SOS died in this time period (3.9%).

Analysis of risk factors

Table 2 shows the medians and interquartile ranges of the investigated laboratory parameters as well as the results of the Mann–Whitney U test. The coagulation parameters INR, aPTT, and reptilase time demonstrated significantly higher values in the SOS group than in the non-SOS group. In Mann–Whitney U test, we detected that antithrombin III, protein C, and factor VIII were significantly lower in the SOS group than in the non-SOS group. Patients with SOS also had significantly higher levels of D-dimer, bilirubin, and ferritin. We observed no significant differences of factor XIII, vWF, vWF activity, fibrinogen, protein S, and ALT between the SOS group and the non-SOS group by using Mann–Whitney U test.
Table 2
Laboratory parameters with median, interquartile range, and p value of the Mann–Whitney U test
Laboratory parameters
SOS
No SOS
Mann–Whitney U test: p
INR
 Median
1.20
1.15
0.006
 IQR
0.22
0.15
 
aPTT in sec
 Median
42.00
34.50
 < 0.001
 IQR
6.00
5.50
 
Reptilase time in sec
 Median
19.40
18.00
0.012
 IQR
5.15
2.55
 
Factor VIII in %
 Median
71.10
110.85
 < 0.001
 IQR
65.43
33.70
 
Factor XIII in %
 Median
76.00
76.00
0.638
 IQR
35.00
34.00
 
vWF in %
 Median
156.10
180.15
0.367
 IQR
91.25
61.41
 
vWF activity in %
 Median
127.50
175.48
0.079
 IQR
115.15
98.33
 
Fibrinogen in g/L
 Median
3.70
3.45
0.946
 IQR
1.60
1.25
 
Antithrombin III in %
 Median
75.00
87.00
0.001
 IQR
25.00
21.00
 
Protein C in %
 Median
43.50
64.00
 < 0.001
 IQR
26.90
25.50
 
Protein S in %
 Median
44.00
52.00
0.071
 IQR
35.00
21.00
 
D-dimer in µg/L
 Median
506.00
312.00
0.006
 IQR
817.00
328.00
 
ALT in µmol/L*s
 Median
0.60
0.57
0.624
 IQR
0.47
0.47
 
Bilirubin in µmol/L
 Median
11.00
7.00
 < 0.001
 IQR
14.00
4.00
 
Ferritin in µg/L
 Median
4227.15
2079.85
0.002
 IQR
5705.68
2768.80
 
p-values of less than 0.05 indicated statistical significance (in bold)
ALT alanine aminotransferase, aPTT activated partial thromboplastin time, INR international normalized ratio, IQR interquartile range, SOS sinusoidal obstruction syndrome, vWF von Willebrand factor, vWF activity von Willebrand factor activity
We selected cut-off values, which were defined by reference values and ROC curve analyses. Figure 1 demonstrates the ROC curves of INR, aPTT, protein C, and ferritin with the cut-off values. The results of the univariate anlyses are presented in Table 3 with the corresponding p value, odds ratio, and 95% CI. For the values of factor XIII, vWF, VWF activity, fibrinogen, protein S, and ALT we could not find any significant results. If the coagulation parameters exceeded a certain value, such as INR ≥ 1.3, aPTT ≥ 40 s, and reptilase time ≥ 18.3 s, the SOS was diagnosed significantly more often. In addition, patients with factor VIII ≤ 80%, antithrombin III ≤ 75%, and protein C ≤ 48% showed a significantly higher incidence of SOS. We detected a significant association between the onset of SOS and the serum levels of D-dimer ≥ 315 µg/L, bilirubin ≥ 9 µmol/L, and ferritin ≥ 3100 µg/L after HSCT.
Table 3
Univariate analyses of laboratory parameters
Laboratory parameters
Total No.
SOS No.
Univariate analyses
OR
95% CI
p
INR
 ≥ 1.3
27
12 (44.4%)
7.429
2.907–18.982
 < 0.001
 < 1.3
144
14 (9.7%)
   
aPTT
 ≥ 40 s
44
21 (47.7%)
19.022
6.925–52.247
 < 0.001
 < 40 s
131
6 (4.6%)
   
Reptilase time
 ≥ 18.3 s
64
15 (23.4%)
4.041
1.376–11.869
0.014
 < 18.3 s
71
5 (7.0%)
   
Factor VIII
 ≤ 80%
18
12 (66.7%)
17.400
5.355–56.537
 < 0.001
 > 80%
97
10 (10.3%)
   
Factor XIII
 ≥ 75%
90
15 (16.7%)
1.511
0.621–3.677
0.386
 < 75%
77
9 (11.7%)
   
vWF
 ≤ 170%
66
14 (21.2%)
1.713
0.720–4.079
0.272
 > 170%
81
11 (13.6%)
   
vWF activity
 ≤ 160%
58
14 (24.1%)
2.256
0.943–5.395
0.075
 > 160%
89
11 (12.4%)
   
Fibrinogen
 ≥ 3.5 g/L
92
16 (17.4%)
1.512
0.659–3.467
0.405
 < 3.5 g/L
90
11 (12.2%)
   
Antithrombin III
 ≤ 75%
46
15 (32.6%)
4.960
2.109–11.664
 < 0.001
 > 75%
135
12 (8.9%)
   
Protein C
 ≤ 48%
40
18 (45.0%)
12.784
4.953–32.994
 < 0.001
 > 48%
133
8 (6.0%)
   
Protein S
 ≤ 50%
84
17 (20.2%)
2.284
0.925–5.638
0.083
 > 50%
80
8 (10.0%)
   
D-dimer
 ≥ 315 µg/L
90
21 (23.3%)
3.601
1.371–9.461
0.010
 < 315 µg/L
77
6 (7.8%)
   
ALT
 ≥ 1 µmol/L*s
30
5 (16.7%)
1.182
0.409–3.415
0.780
 < 1 µmol/L*s
152
22 (14.5%)
   
Bilirubin
 ≥ 9 µmol/L
70
19 (27.1%)
4.843
1.986–11.810
 < 0.001
 < 9 µmol/L
112
8 (7.1%)
   
Ferritin
 ≥ 3100 µg/L
54
17 (31.5%)
4.799
1.965–11.721
0.001
 < 3100 µg/L
103
9 (8.7%)
   
p-values of less than 0.05 indicated statistical significance (in bold)
ALT alanine aminotransferase, aPTT activated partial thromboplastin time, CI confidence interval, INR international normalized ratio, No. number, OR odds ratio, SOS sinusoidal obstruction syndrome, vWF von Willebrand factor, vWF activity von Willebrand factor activity
We investigated nine laboratory parameters in the multivariate analysis, which were significant in the univariate analyses. After binary logistic regression and backward elimination, we identified four laboratory parameters as significant independent variables. The significant correlation between INR ≥ 1.3 after HSCT and SOS could be confirmed (OR = 8.104, 95% CI = 1.847–35.554, p = 0.006). In addition, aPTT ≥ 40 s before SOS was significantly associated with the onset of SOS (OR = 10.174, 95% CI = 2.722–38.020, p = 0.001). Protein C ≤ 48% (OR = 5.215, 95% CI = 1.390–19.562, p = 0.014) and ferritin ≥ 3100 µg/L (OR = 7.472, 95% CI = 1.912–29.202, p = 0.004) could be affirmed as significant predictive variables. Table 4 displays the significant results of the multivariate analysis.
Table 4
Multivariate analysis of laboratory parameters
Laboratory parameters
Multivariate analysis
OR
95% CI
p
INR ≥ 1.3
8.104
1.847–35.554
0.006
aPTT ≥ 40 s
10.174
2.722–38.020
0.001
Protein C ≤ 48%
5.215
1.390–19.562
0.014
Ferritin ≥ 3100 µg/L
7.472
1.912–29.202
0.004
p-values of less than 0.05 indicated statistical significance (in bold)
aPTT activated partial thromboplastin time, CI confidence interval, INR international normalized ratio, OR odds ratio
The four significant laboratory parameters can be used to predict the probability of SOS, which depends on the regression coefficients of the parameters. We calculated the probability of SOS for each combination of the existing cut-off values using logistic regression. If three of the four significant cut-off values were present, the probability of developing SOS was more than 70%. Additionally, if all four laboratory parameters were changed according to the cut-off values, the probability of SOS was 96%.

Discussion

In this retrospective analysis, 27 out of 182 patients (14.8%) developed SOS after HSCT. Following allogeneic HSCT, the incidence was higher (19.1%) than in the autologous patient group (5.4%). Therefore, our study is in line with other publications, which report comparable incidence rates of 14–20% (Xia et al. 2021; Coppell et al. 2010). Additionally, the published lower incidence rate following autologous HSCT (8.7%) in comparison to allogeneic HSCT (12.9%) by Coppell et al. (2010), agrees with our findings. In our study, SOS was diagnosed within 1 to 28 days after HSCT (median 13 days). This is comparable to a study by Coutsouvelis et al. (2023), in which a median of 14 days after HSCT was reported. We observed a low overall mortality of 11.1% in SOS patients within 100 days following HSCT. Coutsouvelis et al. (2023) reported a similar mortality rate of 9.4%. An early treatment with defibrotide is associated with a better outcome and could be the reason for the low mortality rate in our study (Corbacioglu et al. 2004).
We found significantly higher INR values in the SOS group than in the non-SOS patient group after HSCT before diagnosis. Moreover, our study demonstrated in the univariate and the multivariate analysis (OR = 8.104, p = 0.006), that INR ≥ 1.3 after HSCT was significantly associated with the occurrence of SOS. A study by Roeker et al. (2019) showed that the INR values in SOS patient group were significantly higher than in non-SOS group on the day of diagnosis. In addition, a high INR ≥ 1.3 before HSCT was considered as a risk factor for the occurrence of SOS (Kloehn et al. 2022). To our knowledge, we report the association between SOS and high INR after HSCT before SOS for the first time. The INR is a blood coagulation screening test, which is particularly dependent on the coagulation factors II, V, VII, and X. Increased INR values are an indication of a bleeding tendency (Bonar et al. 2017; Favaloro and Adcock 2008). Reasons for the increased INR values before the SOS onset could be a reduced synthesis of the coagulation factors due to liver damage or a vitamin K deficiency. Activated coagulation in SOS could also lead to depletion of coagulation factors (Favaloro and Adcock 2008). In addition, an increased bleeding tendency could influence the pathogenesis of SOS, consequently hemorrhage into the Disse space is reported (Carreras and Diaz-Ricart 2011).
Coagulation parameters, such as aPTT and reptilase time were significantly prolonged in the SOS group compared to the non-SOS group. In the univariate analysis, an association between the occurrence of SOS and prolonged aPTT ≥ 40 s or reptilase time ≥ 18.3 s after HSCT could be demonstrated. Reptilase time showed no significant results in the multivariate analysis. In the multivariate analysis, we confirmed aPTT ≥ 40 s as a predictive factor for the development of SOS (OR = 10.174, p = 0.001). To our knowledge, there are no studies in the literature that reported a significant association between aPTT and SOS after HSCT before SOS. However, aPTT is also prolonged after the diagnosis of SOS (Jevtić et al. 2010; Sartori et al. 2005). The aPTT is a screening test to assess intrinsic blood coagulation. A prolonged aPTT is often associated with a deficiency of coagulation factors such as factor VIII, IX, XI or XII (Kershaw 2017). Therefore, the prolonged aPTT could be caused by a decrease of factor VIII, which was reduced in our study.
The median factor VIII activity was significantly lower in the SOS group than in the non-SOS group. We detected factor VIII activity ≤ 80% as a significant variable in the univariate analysis. This, however, could not be confirmed in the multivariate analysis, which agrees with Lee et al. (1998) who reported no significant differences of factor VIII activity in the groups. Nevertheless, factor VIII appears to be an important parameter in the pathogenesis of SOS. Some studies reported that factor VIII and fibrinogen are reasons for the occlusion of the sinusoidal vessels (DeLeve et al. 2002; Shulmann et al. 1987). The reduced factor VIII activity could be the result of liver damage in the context of SOS.
Antithrombin III and protein C are important anticoagulants and showed significant lower values before diagnosis in patients with SOS. In the multivariate analysis, we observed that only protein C ≤ 48% was significantly associated with the development of SOS (OR = 5.215, p = 0.014) and identified protein C as an independent risk factor. Iguchi et al. (2010) reported similar results in a prospective study. The deficiency of protein C can be associated with thrombotic processes, which have an influence on the pathogenesis of SOS. Protein C is synthesized in the liver and therefore it could be decreased by the liver damage (Iguchi et al. 2010; Lee et al. 1998). Thrombotic processes are also associated with increased D-dimer levels. Our study showed that patients after HSCT before SOS had significantly higher D-dimer levels than in the non-SOS patient group. Other studies were able to show similar results (Jevtic et al. 2011; Sartori et al. 2012). We observed a significant association between elevated D-dimer levels ≥ 315 µg/L and SOS in univariate analysis, but this could not be confirmed in multivariate analysis.
In the univariate analysis, bilirubin ≥ 9 µmol/L was significant associated with the occurrence of SOS. Sartori et al. (2012) also found a significantly elevated bilirubin level before diagnosis of SOS. Bilirubin is one of the pediatric EBMT criteria for diagnosing SOS. However, the presence of hyperbilirubinemia is a non-mandatory criterion (Corbacioglu et al. 2018). In a study by Corbacioglu et al. (2020), 29% of pediatric patients did not have a bilirubin level > 34 µmol/L despite SOS. This fits with the non-significant result of bilirubin ≥ 9 µmol/L in the multivariate analysis. High bilirubin levels are often a late finding in patients with SOS (Corbacioglu et al. 2018).
We could find significant increased serum levels of ferritin in the SOS patient group after HSCT before SOS. The multivariate analysis confirmed ferritin ≥ 3100 µg/L after HSCT before SOS as an independent risk factor (OR = 7.472, p = 0.004). High ferritin plasma concentrations after HSCT before the onset of SOS are also reported by Döring et al. (2016). The increased ferritin could be caused by iron overload, which leads to various liver diseases such as SOS (Evens et al. 2004). Additionally, ferritin is an acute-phase protein and is increased in various inflammations in the body and thus can demonstrated inflammatory processes (Armand et al. 2012). High ferritin levels before HSCT are considered as a risk factor for developing SOS (Kloehn et al. 2022; Lee et al. 2010). Therefore, increased ferritin levels are important for the occurrence of SOS before as well as after HSCT.
Factor XIII, vWF, vWF activity, fibrinogen, protein S, and ALT did not show significant p values in Mann–Whitney U test and univariate analyses. However, there are various reports on the association of these parameters with SOS in the published literature (Jevtic et al. 2011; Lee et al. 1998; Park et al. 1997; Sartori et al. 2012).
The laboratory parameters INR, aPTT, protein C, and ferritin are independent risk factors for onset of SOS and the probability of SOS can be calculated with the combination of these parameters. Patients can present different combinations of the certain cut-off values. For each combination, there is a different probability of the SOS development. If at least three of the cut-off values were present, the probability of developing SOS was more than 70%. The probability of SOS was 96%, if all four laboratory parameters are changed according to the cut-off values. To our knowledge, this is the first report in which the probability of SOS was calculated with laboratory parameters.
In conclusion, our study confirmed INR ≥ 1.3, aPTT ≥ 40 s, protein C ≤ 48%, and ferritin ≥ 3100 µg/L as predictive factors for the development of SOS and independent risk factors. To our knowledge, the significant association between the onset of SOS with increased INR (≥ 1.3) and aPTT (≥ 40 s) after HSCT is here demonstrated for the first time.
The regular measurement of INR, aPTT, protein C, and ferritin is a useful tool to predict the occurence of SOS after HSCT before SOS. These four parameters can enable earlier diagnosis and treatment, leading to an improved outcome. Other studies are necessary to validate our results, so these results can be used in clinical practice.

Declarations

Conflict of interest

There is no potential conflict of interest for the authors.

Ethical approval

All procedures complied with ethical standards. The study was accepted by the ethics committee of the University Hospital Jena (2023–2890). Informed consent was obtained from all responsible persons.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
Zurück zum Zitat Cesaro S, Pillon M, Talenti E, Toffolutti T, Calore E, Tridello G, Strugo L, Destro R, Gazzola MV, Varotto S, Errigo G, Carli M, Zanesco L, Messina C (2005) A prospective survey on incidence, risk factors and therapy of hepatic veno-occlusive disease in children after hematopoietic stem cell transplantation. Haematologica 90(10):1396–1404PubMed Cesaro S, Pillon M, Talenti E, Toffolutti T, Calore E, Tridello G, Strugo L, Destro R, Gazzola MV, Varotto S, Errigo G, Carli M, Zanesco L, Messina C (2005) A prospective survey on incidence, risk factors and therapy of hepatic veno-occlusive disease in children after hematopoietic stem cell transplantation. Haematologica 90(10):1396–1404PubMed
Zurück zum Zitat Coppell JA, Richardson PG, Soiffer R, Martin PL, Kernan NA, Chen A, Guinan E, Vogelsang G, Krishnan A, Giralt S, Revta C, Carreau NA, Iacobelli M, Carreras E, Ruutu T, Barbui T, Antin JH, Niederwieser D (2010) Hepatic veno-occlusive disease following stem cell transplantation: incidence, clinical course, and outcome. Biol Blood Marrow Transplant 16(2):157–168. https://doi.org/10.1016/j.bbmt.2009.08.024CrossRefPubMed Coppell JA, Richardson PG, Soiffer R, Martin PL, Kernan NA, Chen A, Guinan E, Vogelsang G, Krishnan A, Giralt S, Revta C, Carreau NA, Iacobelli M, Carreras E, Ruutu T, Barbui T, Antin JH, Niederwieser D (2010) Hepatic veno-occlusive disease following stem cell transplantation: incidence, clinical course, and outcome. Biol Blood Marrow Transplant 16(2):157–168. https://​doi.​org/​10.​1016/​j.​bbmt.​2009.​08.​024CrossRefPubMed
Zurück zum Zitat Corbacioglu S, Greil J, Peters C, Wulffraat N, Laws HJ, Dilloo D, Straham B, Gross-Wieltsch U, Sykora KW, Ridolfi-Lüthy A, Basu O, Gruhn B, Güngör T, Mihatsch W, Schulz AS (2004) Defibrotide in the treatment of children with veno-occlusive disease (VOD): a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant 33(2):189–195. https://doi.org/10.1038/sj.bmt.1704329CrossRefPubMed Corbacioglu S, Greil J, Peters C, Wulffraat N, Laws HJ, Dilloo D, Straham B, Gross-Wieltsch U, Sykora KW, Ridolfi-Lüthy A, Basu O, Gruhn B, Güngör T, Mihatsch W, Schulz AS (2004) Defibrotide in the treatment of children with veno-occlusive disease (VOD): a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant 33(2):189–195. https://​doi.​org/​10.​1038/​sj.​bmt.​1704329CrossRefPubMed
Zurück zum Zitat Corbacioglu S, Cesaro S, Faraci M, Valteau-Couanet D, Gruhn B, Rovelli A, Boelens JJ, Hewitt A, Schrum J, Schulz AS (2012) Defibrotide for prophylaxis of hepatic veno-occlusive disease in paediatric haemopoietic stem-cell transplantation: an open-label, phase 3, randomised controlled trial. Lancet 379(9823):1301–1309. https://doi.org/10.1016/S0140-6736(11)61938-7CrossRefPubMed Corbacioglu S, Cesaro S, Faraci M, Valteau-Couanet D, Gruhn B, Rovelli A, Boelens JJ, Hewitt A, Schrum J, Schulz AS (2012) Defibrotide for prophylaxis of hepatic veno-occlusive disease in paediatric haemopoietic stem-cell transplantation: an open-label, phase 3, randomised controlled trial. Lancet 379(9823):1301–1309. https://​doi.​org/​10.​1016/​S0140-6736(11)61938-7CrossRefPubMed
Zurück zum Zitat Corbacioglu S, Carreras E, Ansari M, Balduzzi A, Cesaro S, Dalle JH, Dignan F, Gibson B, Guengoer T, Gruhn B, Lankester A, Locatelli F, Pagliuca A, Peters C, Richardson PG, Schulz AS, Sedlacek P, Stein J, Sykora KW, Toporski J, Trigoso E, Vetteranta K, Wachowiak J, Wallhult E, Wynn R, Yaniv I, Yesilipek A, Mohty M, Bader P (2018) Diagnosis and severity criteria for sinusoidal obstruction syndrome/veno-occlusive disease in pediatric patients: a new classification from the European society for blood and marrow transplantation. Bone Marrow Transplant 53(2):138–145. https://doi.org/10.1038/bmt.2017.161CrossRefPubMed Corbacioglu S, Carreras E, Ansari M, Balduzzi A, Cesaro S, Dalle JH, Dignan F, Gibson B, Guengoer T, Gruhn B, Lankester A, Locatelli F, Pagliuca A, Peters C, Richardson PG, Schulz AS, Sedlacek P, Stein J, Sykora KW, Toporski J, Trigoso E, Vetteranta K, Wachowiak J, Wallhult E, Wynn R, Yaniv I, Yesilipek A, Mohty M, Bader P (2018) Diagnosis and severity criteria for sinusoidal obstruction syndrome/veno-occlusive disease in pediatric patients: a new classification from the European society for blood and marrow transplantation. Bone Marrow Transplant 53(2):138–145. https://​doi.​org/​10.​1038/​bmt.​2017.​161CrossRefPubMed
Zurück zum Zitat Coutsouvelis J, Kirkpatrick CM, Dooley M, Spencer A, Kennedy G, Chau M, Huang G, Doocey R, Copeland TS, Do L, Bardy P, Kerridge I, Cole T, Fraser C, Perera T, Larsen SR, Mason K, O’Brien TA, Shaw PJ, Teague L, Butler A, Watson AM, Ramachandran S, Marsh J, Khan Z, Hamad N (2023) Incidence of sinusoidal obstruction syndrome/veno-occlusive disease and treatment with defibrotide in allogeneic transplant: a multicentre Australasian registry study. Transplant Cell Ther 29(6):383.e1-383.e10. https://doi.org/10.1016/j.jtct.2023.03.014CrossRefPubMed Coutsouvelis J, Kirkpatrick CM, Dooley M, Spencer A, Kennedy G, Chau M, Huang G, Doocey R, Copeland TS, Do L, Bardy P, Kerridge I, Cole T, Fraser C, Perera T, Larsen SR, Mason K, O’Brien TA, Shaw PJ, Teague L, Butler A, Watson AM, Ramachandran S, Marsh J, Khan Z, Hamad N (2023) Incidence of sinusoidal obstruction syndrome/veno-occlusive disease and treatment with defibrotide in allogeneic transplant: a multicentre Australasian registry study. Transplant Cell Ther 29(6):383.e1-383.e10. https://​doi.​org/​10.​1016/​j.​jtct.​2023.​03.​014CrossRefPubMed
Zurück zum Zitat Lee JH, Lee KH, Kim S, Lee JS, Kim WK, Park CJ, Chi HS, Kim SH (1998) Relevance of proteins C and S, antithrombin III, von Willebrand factor, and factor VIII for the development of hepatic veno-occlusive disease in patients undergoing allogeneic bone marrow transplantation: a prospective study. Bone Marrow Transplant 22(9):883–888. https://doi.org/10.1038/sj.bmt.1701445CrossRefPubMed Lee JH, Lee KH, Kim S, Lee JS, Kim WK, Park CJ, Chi HS, Kim SH (1998) Relevance of proteins C and S, antithrombin III, von Willebrand factor, and factor VIII for the development of hepatic veno-occlusive disease in patients undergoing allogeneic bone marrow transplantation: a prospective study. Bone Marrow Transplant 22(9):883–888. https://​doi.​org/​10.​1038/​sj.​bmt.​1701445CrossRefPubMed
Zurück zum Zitat Lee SH, Yoo KH, Sung KW, Koo HH, Kwon YJ, Kwon MM, Park HJ, Park BK, Kim YY, Park JA, Im HJ, Seo JJ, Kang HJ, Shin HY, Ahn HS (2010) Hepatic veno-occlusive disease in children after hematopoietic stem cell transplantation: incidence, risk factors, and outcome. Bone Marrow Transplant 45(8):1287–1293. https://doi.org/10.1038/bmt.2009.349CrossRefPubMed Lee SH, Yoo KH, Sung KW, Koo HH, Kwon YJ, Kwon MM, Park HJ, Park BK, Kim YY, Park JA, Im HJ, Seo JJ, Kang HJ, Shin HY, Ahn HS (2010) Hepatic veno-occlusive disease in children after hematopoietic stem cell transplantation: incidence, risk factors, and outcome. Bone Marrow Transplant 45(8):1287–1293. https://​doi.​org/​10.​1038/​bmt.​2009.​349CrossRefPubMed
Zurück zum Zitat Mohty M, Blaise D, Peffault de Latour R, Labopin M, Bourhis JH, Bruno B, Ceballos P, Detrait M, Gandemer V, Huynh A, Izadifar-Legrand F, Jubert C, Labussière-Wallet H, Lebon D, Maury S, Paillard C, Pochon C, Renard C, Rialland F, Schneider P, Sirvent A, Asubonteng K, Guindeuil G, Yakoub-Agha I, Dalle JH (2022) Real-world use of defibrotide for veno-occlusive disease/sinusoidal obstruction syndrome: the DEFIFrance registry study. Bone Marrow Transplant 58:367–376. https://doi.org/10.1038/s41409-022-01900-6CrossRefPubMedPubMedCentral Mohty M, Blaise D, Peffault de Latour R, Labopin M, Bourhis JH, Bruno B, Ceballos P, Detrait M, Gandemer V, Huynh A, Izadifar-Legrand F, Jubert C, Labussière-Wallet H, Lebon D, Maury S, Paillard C, Pochon C, Renard C, Rialland F, Schneider P, Sirvent A, Asubonteng K, Guindeuil G, Yakoub-Agha I, Dalle JH (2022) Real-world use of defibrotide for veno-occlusive disease/sinusoidal obstruction syndrome: the DEFIFrance registry study. Bone Marrow Transplant 58:367–376. https://​doi.​org/​10.​1038/​s41409-022-01900-6CrossRefPubMedPubMedCentral
Zurück zum Zitat Richardson PG, Riches ML, Kernan NA, Brochstein JA, Mineishi S, Termuhlen AM, Arai S, Grupp SA, Guinan EC, Martin PL, Steinbach G, Krishnan A, Nemecek ER, Giralt S, Rodriguez T, Duerst R, Doyle J, Antin JH, Smith A, Lehmann L, Champlin R, Gillio A, Bajwa R, D’Agostino RB Sr, Massaro J, Warren D, Miloslavsky M, Hume RL, Iacobelli M, Nejadnik B, Hannah AL, Soiffer RJ (2016) Phase 3 trial of defibrotide for the treatment of severe veno-occlusive disease and multi-organ failure. Blood 127(13):1656–1665. https://doi.org/10.1182/blood-2015-10-676924CrossRefPubMedPubMedCentral Richardson PG, Riches ML, Kernan NA, Brochstein JA, Mineishi S, Termuhlen AM, Arai S, Grupp SA, Guinan EC, Martin PL, Steinbach G, Krishnan A, Nemecek ER, Giralt S, Rodriguez T, Duerst R, Doyle J, Antin JH, Smith A, Lehmann L, Champlin R, Gillio A, Bajwa R, D’Agostino RB Sr, Massaro J, Warren D, Miloslavsky M, Hume RL, Iacobelli M, Nejadnik B, Hannah AL, Soiffer RJ (2016) Phase 3 trial of defibrotide for the treatment of severe veno-occlusive disease and multi-organ failure. Blood 127(13):1656–1665. https://​doi.​org/​10.​1182/​blood-2015-10-676924CrossRefPubMedPubMedCentral
Zurück zum Zitat Roeker LE, Kim HT, Glotzbecker B, Nageshwar P, Nikiforow S, Koreth J, Armand P, Cutler C, Alyea EP, Antin JH, Richardson PG, Soiffer RJ, Ho VT (2019) Early clinical predictors of hepatic veno-occlusive disease/sinusoidal obstruction syndrome after myeloablative stem cell transplantation. Biol Blood Marrow Transplant 25:137–144. https://doi.org/10.1016/j.bbmt.2018.07.039CrossRefPubMed Roeker LE, Kim HT, Glotzbecker B, Nageshwar P, Nikiforow S, Koreth J, Armand P, Cutler C, Alyea EP, Antin JH, Richardson PG, Soiffer RJ, Ho VT (2019) Early clinical predictors of hepatic veno-occlusive disease/sinusoidal obstruction syndrome after myeloablative stem cell transplantation. Biol Blood Marrow Transplant 25:137–144. https://​doi.​org/​10.​1016/​j.​bbmt.​2018.​07.​039CrossRefPubMed
Zurück zum Zitat Sartori MT, Spiezia L, Cesaro S, Messina C, Paris M, Pillon M, Saggiorato G, Pagnan A, Girolami A, Zanesco L, Cella G (2005) Role of fibrinolytic and clotting parameters in the diagnosis of liver veno-occlusive disease after hematopoietic stem cell transplantation in a pediatric population. Thromb Haemost 93(4):682–689. https://doi.org/10.1160/th04-09-0621CrossRefPubMed Sartori MT, Spiezia L, Cesaro S, Messina C, Paris M, Pillon M, Saggiorato G, Pagnan A, Girolami A, Zanesco L, Cella G (2005) Role of fibrinolytic and clotting parameters in the diagnosis of liver veno-occlusive disease after hematopoietic stem cell transplantation in a pediatric population. Thromb Haemost 93(4):682–689. https://​doi.​org/​10.​1160/​th04-09-0621CrossRefPubMed
Zurück zum Zitat Sartori MT, Cesaro S, Peruzzo M, Messina C, Saggiorato G, Calore E, Pillon M, Varotto S, Spiezia L, Cella G (2012) Contribution of fibrinolytic tests to the differential diagnosis of veno-occlusive disease complicating pediatric hematopoietic stem cell transplantation. Pediatr Blood Cancer 58(5):791–797. https://doi.org/10.1002/pbc.23213CrossRefPubMed Sartori MT, Cesaro S, Peruzzo M, Messina C, Saggiorato G, Calore E, Pillon M, Varotto S, Spiezia L, Cella G (2012) Contribution of fibrinolytic tests to the differential diagnosis of veno-occlusive disease complicating pediatric hematopoietic stem cell transplantation. Pediatr Blood Cancer 58(5):791–797. https://​doi.​org/​10.​1002/​pbc.​23213CrossRefPubMed
Zurück zum Zitat Shulman HM, Gown AM, Nugent DJ (1987) Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol 127(3):549–558PubMedPubMedCentral Shulman HM, Gown AM, Nugent DJ (1987) Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol 127(3):549–558PubMedPubMedCentral
Zurück zum Zitat Yoon JH, Yoo KH, Sung KW, Jung CW, Kim JS, Hahn SM, Kang HJ, Lee JH, Im HJ, Ahn JS, Kook H, Cho B, Lee JW (2019) Validation of treatment outcomes according to revised severity criteria from European Society for Blood and Marrow Transplantation (EBMT) for sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD). Bone Marrow Transplant 54(8):1361–1368. https://doi.org/10.1038/s41409-019-0492-6CrossRefPubMed Yoon JH, Yoo KH, Sung KW, Jung CW, Kim JS, Hahn SM, Kang HJ, Lee JH, Im HJ, Ahn JS, Kook H, Cho B, Lee JW (2019) Validation of treatment outcomes according to revised severity criteria from European Society for Blood and Marrow Transplantation (EBMT) for sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD). Bone Marrow Transplant 54(8):1361–1368. https://​doi.​org/​10.​1038/​s41409-019-0492-6CrossRefPubMed
Metadaten
Titel
Analysis of laboratory parameters before the occurrence of hepatic sinusoidal obstruction syndrome in children, adolescents, and young adults after hematopoietic stem cell transplantation
verfasst von
Lorena Johann
Bernd Gruhn
Publikationsdatum
01.01.2025
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Cancer Research and Clinical Oncology / Ausgabe 1/2024
Print ISSN: 0171-5216
Elektronische ISSN: 1432-1335
DOI
https://doi.org/10.1007/s00432-023-05561-w

Weitere Artikel der Ausgabe 1/2024

Journal of Cancer Research and Clinical Oncology 1/2024 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.