Basic Study Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Aug 27, 2024; 16(8): 1167-1176
Published online Aug 27, 2024. doi: 10.4254/wjh.v16.i8.1167
Gadoxetic acid-enhanced magnetic resonance imaging in the assessment of hepatic sinusoidal obstruction syndrome in a mouse model
Yuan-Yuan Chen, Li Yang, Jun Li, Sheng-Xiang Rao, Ying Ding, Meng-Su Zeng, Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
ORCID number: Yuan-Yuan Chen (0000-0003-2342-145X); Sheng-Xiang Rao (0000-0001-7722-8327); Ying Ding (0000-0002-4478-3436).
Author contributions: Chen YY contributed to the conception, design, acquisition, analysis, interpretation of data, wrote the manuscript; Yang L and Li J collected and preprocessed the data; Rao SX provided supervision and methodology; Ding Y revised the manuscript and gave the final approval of the version to be published; Zeng MS was responsible for conceptualization and supervision.
Supported by the National Science Foundation for Young Scientists of China, No. 81701682.
Institutional animal care and use committee statement: Animal experimental procedures were reviewed and approved by Zhongshan Hospital Institutional Ethics and Animal Care on February 4, 2017.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: The data that support the findings of this study are available from the corresponding author upon reasonable request at ding.ying@zs-hospital.sh.cn.
ARRIVE guidelines statement: The authors have read the ARRIVE Guidelines, and the manuscript was prepared and revised according to the ARRIVE Guidelines.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ying Ding, MD, Associate Chief Physician, Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China. ding.ying@zs-hospital.sh.cn
Received: May 15, 2024
Revised: July 13, 2024
Accepted: August 2, 2024
Published online: August 27, 2024
Processing time: 97 Days and 23.7 Hours

Abstract
BACKGROUND

Neoadjuvant chemotherapy can cause hepatic sinusoidal obstruction syndrome (SOS) in patients with colorectal cancer liver metastases and increases postoperative morbidity and mortality.

AIM

To evaluate T1 mapping based on gadoxetic acid-enhanced magnetic resonance imaging (MRI) for diagnosis of hepatic SOS induced by monocrotaline.

METHODS

Twenty-four mice were divided into control (n = 10) and experimental (n = 14) groups. The experimental groups were injected with monocrotaline 2 or 6 days before MRI. MRI parameters were: T1 relaxation time before enhancement; T1 relaxation time 20 minutes after enhancement (T1post); a reduction in T1 relaxation time (△T1%); and first enhancement slope percentage of the liver parenchyma (ESP). Albumin and bilirubin score was determined. Histological results served as a reference. Liver parenchyma samples from the control and experimental groups were analyzed by western blotting, and organic anion transporter polypeptide 1 (OATP1) was measured.

RESULTS

T1post, △T1%, and ESP of the liver parenchyma were significantly different between two groups (all P < 0.001) and significantly correlated with the total histological score of hepatic SOS (r = -0.70, 0.68 and 0.79; P < 0.001). △T1% and ESP were positively correlated with OATP1 levels (r = 0.82, 0.85; P < 0.001), whereas T1post had a negative correlation with OATP1 levels (r = -0.83; P < 0.001).

CONCLUSION

T1 mapping based on gadoxetic acid-enhanced MRI may be useful for diagnosis of hepatic SOS, and MRI parameters were associated with OATP1 levels.

Key Words: T1 mapping; Gadoxetic acid; Sinusoidal obstruction syndrome; Organic anion transporter polypeptides; Magnetic resonance imaging

Core Tip: Neoadjuvant chemotherapy could cause hepatic sinusoidal obstruction syndrome (SOS) in colorectal liver metastases patients and increases postoperative morbidity and mortality. We used a mouse model of monocrotaline-induced hepatic SOS. We confirmed the relationship between magnetic resonance imaging (MRI) parameters T1 relaxation time 20 minutes after enhancement, a reduction in T1 relaxation time and first enhancement slope percentage of the liver parenchyma and hepatic SOS, suggesting a potential method for the assessment of hepatic SOS. Our results also demonstrated that MRI parameters were correlated with organic anion transporter polypeptide 1 levels.



INTRODUCTION

Hepatic sinusoidal obstruction syndrome (SOS) is also known as hepatic veno-occlusive disease of the liver[1]. The main pathological feature of hepatic SOS is damage to liver terminal vessels, and the clinical symptoms of it include ascites and abdominal pain[2]. It was first proposed in 1979 as an early complication of hematopoietic stem cell transplantation[3].The prevalence ranges from 5% to 60%, and hepatic SOS is a potentially severe complication and can even lead to death in severe cases[4]. Recently, systemic neoadjuvant chemotherapy became widely regarded as one of the causes hepatic SOS in the patients with advanced metastatic colorectal cancer[5,6], especially those were treated with oxaliplatin[7,8].

Oxaliplatin-based preoperative chemotherapy is used for patients with colorectal liver metastases as the standard regimen[8,9], because it could improve tumor resection outcome by shrinking the metastatic sites and reducing recurrence rate[10]. Nevertheless, chemotherapy-induced hepatic SOS has been associated with a higher risk of postresection morbidity[11], such as intraoperative bleeding, intraoperative transfusions, and postoperative liver failure[12]. Therefore, it is important to detect and diagnose of hepatic SOS timely. Currently, the gold standard is still based on liver biopsy[13], but it is an invasive procedure and has several limitations and complications, such as hemorrhage[14]. A noninvasive diagnostic modality is needed for the assessment of hepatic SOS.

Some noninvasive tools have been used for diagnosis of hepatic SOS. Researchers have utilized a preoperative platelet count and aspartate aminotransferase to platelet ratio index[15]. In addition, some imaging methods such as shear wave ultrasonography, computed tomography, and gadoxetic acid-enhanced magnetic resonance imaging (MRI) have been promoted as useful methods for evaluation of hepatic SOS[16-18]. Recent studies with monocrotaline (MCT)-treated rats were conducted to investigate diagnosis and prediction of severity of SOS. For example, intravoxel incoherent motion diffusion-weighted imaging, non-Gaussian diffusion models, and T1 rho quantification[19,20]. The MCT-induced hepatic SOS animal model was reproducible, with a detailed pathological scoring criteria[21].

Gadoxetic acid is a hepatocyte-specific contrast substance, which can provide parenchymal contrast in the hepatobiliary phase. It is reported that gadoxetic acid is absorbed into the liver parenchyma via organic anion transporter polypeptide 1 (OATP1) on the hepatocyte membranes[22-24]. Recently, several authors have described the feasibility of gadoxetic acid-enhanced MRI for the diagnosis of oxaliplatin-induced hepatic SOS[25]. They mainly diagnosed hepatic SOS based on the signal intensity of the hepatobiliary specific phase. However, there were several limitations due to the inconsistency between signal intensity of the liver parenchyma and the concentration of contrast agent for evaluation of the degree of hepatic SOS[26]. Therefore, we measured T1 relaxation time on parametric mapping because it is linearly related to the concentration of the contrast agent and is not affected by other factors[27]. Yang et al[28] demonstrated T1mapping on gadoxetic acid-enhanced MRI for the assessment of oxaliplatin-induced liver injury in a C57BL/6 mouse model. However, the main pathological changes in their model were hepatocyte degeneration and fibrosis.

Therefore, we aimed to explore the effectiveness of T1 mapping based on gadoxetic acid-enhanced MRI for the diagnosis of hepatic SOS in a C57BL/6 mouse model, as well as a possible relation between OATP1 Levels and MRI parameters.

MATERIALS AND METHODS
Animal models

Male 6-week-old C57BL/6 mice (17-22 g) were purchased from Shanghai Jiesijie Laboratory Animal Institution (Shanghai, China) and maintained according to the institutional animal care guidelines. Animal experimental procedures were reviewed and approved by our hospital Institutional Ethics and Animal Care. Twenty-four mice were randomly distributed into control (n = 10) and two experimental (n = 7) groups. SOS was induced by MCT as described previously[19]. The experimental mice were injected intraperitoneally (ip) with 360 mg/kg MCT, while control mice were treated with tap water. To assess experimental mice with different severity of hepatic SOS, MRI was randomly performed on 2 and 6 days after administration of MCT.

MRI protocols

All examinations were carried out on a 3.0 T MRI scanner (Verio; Siemens Medical Solutions, Erlangen, Germany). During the examination, each mouse was placed in a prone position with a mouse coil and the abdomen of the mouse was restrained with a belt to minimize the effects of respiratory movement: (1) T1 mapping were obtained before and 20 minutes after the tail vein injection of 0.1 mL/kg gadoxetic acid (Primovist; Bayer Schering Pharma AG, Berlin, Germany), by using a dual flip angle 3D gradient echo sequence with volumetric interpolation breath-holding examination sequence (VIBE): Repetition time (TR), 5.32 millisecond; echo time (TE), 1.73 millisecond; flip angle, 2° and 12°; field of view (FOV), 108 mm; matrix, 160 × 160; slice thickness, 1.5 mm; and R factor, 2; and (2) Dynamic contrast-enhanced (DCE) MRI was performed as the same time as the start of contrast injection, using T1-weighted fast dynamic VIBE: TR, 6.64 millisecond; TE, 1.53 millisecond; flip angle, 9°; FOV, 108 mm; slice thickness, 1.5 mm; R factor, 2; 48 consecutive acquisitions over a period of 6:04 minutes.

Image analysis

All MRI datasets were transmitted to a workstation (Leonardo; Siemens). For T1 mapping, three regions of interest (ROIs) were chosen at the same locations in the liver parenchyma of the pre- and post-T1 maps (Figure 1). Each ROI was excluded blood vessels and the margin of liver, and the mean value was taken as the representative of the T1 relaxation time. All MRIs were measured by a radiologist with 15 years’ abdominal imaging experience, who was double-blind to the animal groups and pathology results. The MRI analyses were as follows: (1) Reduction rate of T1 relaxation time (ΔT1%) was quantified as 100% × (T1pre - T1post)/T1pre, where T1pre and T1post respectively refers T1 relaxation time before and after enhancement; and (2) The first rapid enhancement slope percentage (ESP) in the liver parenchyma was quantified as [(SI1st - SIbase)/SIbase] × △T × 100%; where SIbase referred to the mean signal value obtained in the first two images after a dynamic enhanced scan; SI1st was the maximum value in the liver parenchyma reached in the first perfusion on the time-signal curve; and △T denoted the interval of time from SIbase to SI1st. These data were calculated in custom software on a postprocessing workstation (Mean Curve, Syngo MR B17; Siemens). The same ROIs were selected in the liver parenchyma as those in T1 mapping. The time-signal curve for the liver parenchyma was also constructed using this software (Figure 2).

Figure 1
Figure 1 Regions of interest in the same area in the liver parenchyma before and after enhancement in the same C57BL/6 mouse. A: Before enhancement in the same C57BL/6 mouse; B: After enhancement in the same C57BL/6 mouse. 1The region of interest in the middle hepatic lobe are delineated to avoid blood vessels within the liver parenchyma; 2The region of interest in the right hepatic lobe are delineated to avoid blood vessels within the liver parenchyma; 3The region of interest in the right hepatic lobe are delineated to avoid blood vessels within the liver parenchyma.
Figure 2
Figure 2 Liver parenchyma time–signal curve: The x axis represents the time axis, and the y axis represents T1 relaxation time, and the slope represents the first rapid enhancement slope percentage. The three lines represent the values of the three regions of interest (ROIs). We selected the same ROIs in the liver parenchyma for T1 mapping.
Laboratory and histological analysis

After MRI, the mice were anesthetized and humanely killed by cervical dislocation, and blood samples were collected. An upper portion of serum was collected after centrifugation, total bilirubin and albumin levels were determined, and an albumin and bilirubin score (ALBI) was calculated as log10 bilirubin (μmol/L)´ 0.66 + albumin (g/L)´ - 0.085. The liver samples were stained with hematoxylin-eosin (HE) and Masson’s dye to examine pathological changes in hepatic structure and to evaluate hepatic SOS. The hepatic SOS histological scores were assessed by a pathologist blinded to the group assignments and imaging results. Histological features of hepatic SOS included sinusoidal hemorrhage, lobular inflammation, endothelial damage to the central vein, subendothelial hemorrhage in the central vein, centrilobular fibrosis, and coagulation necrosis of hepatocytes. Each feature was rated on a four-point scale via consensus (0 = absent; 1 = mild; 2 = moderate; and 3 = severe). We calculated the final pathology score by summing the individual scores of the six features.

OATP1 detection

Western blotting was performed to quantify OATP levels: (1) Protein extraction: The liver specimen was washed with ice-cold phosphate buffer three times, and an appropriate amount of lysis buffer was supplemented with benzyl sulfonyl fluoride to a final concentration of 1 mmol/L. Lysis buffer (100 μL) was added per 10 mg tissue; the mixture was shaken for 2 minutes in a 4-gauge homogenizer until full disintegration; and the supernatant was collected after 5 minutes of centrifugation at 10000 g; (2) Protein quantification: A human BCA working solution was prepared by mixing reagents A and B of the BCA Protein Quantitative Kit (Beyotime, Shanghai) at a volume ratio of 50:1. An appropriate amount of each liver tissue sample was placed into a 1.5 mL centrifuge tube; isosmotic saline was added to obtain a total volume of 100 μL; and 1 mL human BCA working solution was added. The protein concentration was calculated from the standard curve; (3) PAGE: This assay was carried out on a Bole electrophoresis apparatus (Bio-Rad, Hercules, CA, United States). The protein samples (lysates) were electrophoresed until the dye reached the top of the separation gel at a constant voltage of 120 V, and when bromophenol blue was just out of the loading gel, they were electrophoresed at a constant voltage of 160 V; and (4) Immunoassay: After a fresh sealing solution was prepared, skimmed milk powder was added to Tris-buffered saline supplemented with 0.05% of Tween 20 (TBST) to a final concentration of 0.05 kg/L. A diluted primary antibody was incubated with the samples overnight at 4 °C. The type and concentration of the antibody were as follows: A rabbit anti-mouse OATP1 antibody and the dilution ratio of 1:1000.

Statistical analysis

Descriptive statistics were calculated for MRI parameters, ALBI and OATP1 levels, and were presented as mean ± SD. Statistical significance was performed by using the student’s t test between groups. Spearman’s rank correlation coefficient was calculated to evaluate the correlation between histological scores and individual MRI parameters, ALBI. The correlation between individual MRI parameters, ALBI and OATP1 Levels was assessed using Pearson’s correlation. P < 0.05 was considered statistically significant. All data analyses were performed SPSS 26 (IBM Corp., Armonk, NY, United States).

RESULTS
Animal models

One mouse died during the experiment period; therefore, there were 13 mice in the experimental group. In the HE and Masson staining findings, sinusoidal hemorrhage and endothelial damage to the central vein were noted in each mouse in the experimental group, whereas there was no endothelial damage in the control group. Table 1 summarizes the results of the histopathological examination. Typical samples from the experimental groups are shown in Figure 3. Table 2 presents the correlations between MRI parameters and the total histological scores. T1pre showed no correlation with the total histological score (r = 0.37, P = 0.095), T1post had a negative correlation with the total histological score of hepatic SOS (r = -0.70,P < 0.001), whereas ΔT1% and ESP (r = 0.68, 0.79, P < 0.001) significantly increased with the total histological of hepatic SOS. ALBI was also decreased with the total histological of hepatic SOS (r = -0.76, P < 0.001).

Figure 3
Figure 3 Hematoxylin and eosin staining (400 ×). Histological findings of monocrotaline mouse models, the liver specimen demonstrates sinusoidal hemorrhage and hepatocytes necrosis, which are characteristic findings of sinusoidal obstruction syndrome. The triangle represents the central vein, the white arrow indicates dilated hepatic sinusoids hemorrhage, and the black arrow indicates hepatocyte necrosis.
Table 1 Histological features in sinusoidal obstruction syndrome.
No.
Central vein endothelial damage
Central vein subendothelial hemorrhage
Sinusoidal hemorrhage
Lobular inflammation
Hepatocyte necrosis
Centrilobular fibrosis
All scores
12121006
20110002
31111004
41110003
50110022
61001002
71000001
81101115
91111015
102211118
111211016
121221118
1323330011
Table 2 Correlations between magnetic resonance imaging, albumin bilirubin, and total histological score.

T1pre
T1post
△T1%
ESP
ALBI
r0.37-0.700.680.79-0.76
P value0.095< 0.001< 0.001< 0.001< 0.001
Comparisons of MRI parameters and ALBI between the control and experimental groups

Table 3 and Figure 4 show the comparisons of MRI parameters and ALBI between the control and experimental groups. Among the MRI parameters, T1pre showed no significant difference between the control and experimental groups, whereas T1post, ΔT1%, and ESP did. The SOS-mice had a significantly lower T1post than the normal mice had (P < 0.001), and the difference in mean T1post between the control and experimental groups was 288 milliseconds; ΔT1% and ESP were significantly higher in the SOS than in the control group (P < 0.001, P < 0.001), and the difference between the groups was 46.56% and 15.53%, respectively. ALBI also significantly differed between the control and experimental groups.

Figure 4
Figure 4 Box plots in control and experimental groups. A: Box plots of T1 relaxation time before enhancement (T1pre); B: T1 relaxation times 20 minutes after enhancement (T1post); C: The reduction of T1 relaxation time (△T1%); D: The first enhancement slope percentage of the liver parenchyma in control and experimental groups. T1pre: T1 relaxation time before enhancement; T1post: T1 relaxation time 20 minutes after enhancement; △T1%: Reduction rate of T1 relaxation time; ESP: The first enhancement slope percentage in liver parenchyma.
Table 3 Magnetic resonance imaging and albumin/bilirubin in control and experimental groups.
Parameters
Control group (n = 10)
Experimental group (n = 13)
P value
T1pre (millisecond)659.13 ± 24.07681.57 ± 40.580.137
T1post (millisecond)408.87 ± 27.21120.48 ± 53.71< 0.001
△T1%37.84 ± 5.4682.40 ± 7.62< 0.001
ESP4.14 ± 0.9519.67 ± 12.12< 0.001
ALBI0.95 ± 0.040.12 ± 0.89< 0.001
OATP1 Levels between the control and experimental groups

The levels of OATP1 in the control and experimental groups were 29952 ± 11475 and 422060 ± 178634, which was a significant difference (P < 0.001).

Correlations of MRI parameters, ALBI and OATP1 levels

In Pearson’s correlation analysis, T1pre showed no correlation with OATP1 (r = 0.06, P = 0.457). ΔT1% and ESP were strongly positively correlated with OATP1 levels (r = 0.82, 0.85, respectively; P < 0.001), whereas T1post was negatively correlated with OATP1 levels (r = -0.83; P < 0.001; Table 4). In addition, ALBI exhibited a moderate negative correlation with OATP1 Levels (r = -0.56; P = 0.005).

Table 4 Correlation between magnetic resonance imaging, albumin/bilirubin and organic transporter polypeptide 1.

r
P value
T1pre-0.060.803
T1post-0.83< 0.001
△T1%0.82< 0.001
ESP0.85< 0.001
ALBI-0.560.005
DISCUSSION

In this study, we used a mouse model of MTC-induced hepatic SOS. We confirmed the relationship between MRI parameters T1post, △T1% and ESP and hepatic SOS, suggesting a potential method for the assessment of hepatic SOS. Our results also demonstrated that MRI parameters were correlated with OATP1 levels.

We showed that T1post and △T1% had significant differences between the control and experimental groups and were associated with total histological score. Several previous studies[29,30] have reported that T1 mapping on gadoxetic acid-enhanced MRI could reflect the liver function in rats with hepatic fibrosis and found that T1post and △T1% could be better than apparent diffusion coefficient for assessing the rate of liver necro-inflammatory activity and stage of liver fibrosis. Yang et al[28] showed that T1 relaxation time and △T1% might be a sensitive tool for diagnosis of oxaliplatin-induced liver injury by using gadoxetic acid-enhanced MRI. However, they did not explore the relationship between OATP1 Levels and MRI parameters. After injection, gadoxetic acid is gradually taken into the liver parenchyma through OATP1 on the hepatocyte membranes. Our experimental group also found that T1post and △T1% were correlated with expression of OATP1, which was consistent with the literature. Previous studies[31,32] have confirmed that OATP1a1 measurement had a negative correlation with gadoxetic acid in liver fibrosis rats caused by carbon tetrachloride. A recent case[32] reported a patient with hepatic SOS, indicating that changes in expression of OATP seem to be related to the appearance of a low signal in the hepatobiliary phase. These results indicate that T1post and △T1% are reliable methods for diagnosis of hepatic SOS. Gadoxetic acid-enhanced MRI could be applied clinically to examine hepatic SOS in colorectal cancer patients treated with oxaliplatin prior to liver surgery.

In addition to T1post and △T1%, our findings suggested that ESP was helpful for the evaluation of hepatic SOS. ESP exhibited a significant difference between the experimental and control mice. Both animal and human studies have demonstrated the potential of perfusion parameters acquired from DCE-MRI with gadoxetic acid in the assessment of liver function and fibrosis[33,34]. However, the analysis of hepatic DCE-MRI involves complex mathematical modeling, and no model has been generally accepted at present[35]. We calculated ESP to evaluate hemodynamic changes in the liver, which could be easily adapted to clinical practice. Previous research found that ESP of the liver could assess bile duct ligation caused by cholestatic liver injury and has diagnostic efficacy[36]. Our data showed that the ESP was significantly higher in SOS than in the control liver. These findings were consistent with several previous research[36,37]. Chen et al[37] found that arterial fraction was elevated with the severity of liver fibrosis by using DCE-MRI with gadoxetic acid-enhanced MRI in patients with chronic hepatitis. Wu et al[38] also showed that the fraction of hepatic arterial blood volume increased with severity of nonalcoholic fatty liver disease or in fibrotic liver. Based on the results of our study and previous literature, we hypothesized that hepatic SOS might increase blood flow in the damaged liver. Nevertheless, further studies are required to explain this phenomenon.

ALBI grade is a newly proposed, simple, and significant tool for assessment of liver function[39]. Our data revealed the ALBI scores of SOS were significantly different from the ALBI score of the normal liver. A recent study showed mathematical combinations with ALBI were able to identify chemotherapy-associated liver injury[40], which supports that ALBI might be a noninvasive tool for detecting hepatic SOS. Nevertheless, ALBI reflected the whole-liver function, while gadoxetic acid-enhanced MRI could provide information of regional liver function. Zhou et al[41] indicated that T1mapping based on gadoxetic acid-enhanced MRI could be used to assess the function of hepatic segments. This suggested that gadoxetic acid-enhanced MRI could better reflect liver function than laboratory examination index.

There are other imaging techniques that have been suggested to be helpful for detecting hepatic SOS. For example, Park et al[42] confirmed the potential of acoustic-radiation-pulsed elastography for diagnosis, severity evaluation and follow-up of hepatic SOS in animal models. However, the application of ultrasound elastography has limitations and the results rely more on the subjective judgment of the examiners. Hong et al[19] and Lyu et al[20] respectively demonstrated the traditional double-exponential Gaussian model and non-Gaussian model to diagnose the severity of hepatic SOS. Compared with other tools, gadoxetic acid-enhanced MRI also can detect chemotherapy-related focal nodular hyperplasia-like lesions and accurately identify liver metastases, and it can allow “one-stop shopping” in one examination.

There were some limitations to our research. First, our study involved MCT-induced SOS, which was considered as a reproducible animal model, rather than clinical oxaliplatin-induced SOS. Second, the sample size was small; however, T1post and △T1% were significantly associated with the severity of hepatic SOS. A large sample size study on humans should be researched to confirm these results.

CONCLUSION

In conclusion, our findings suggest that T1 mapping on gadoxetic acid-enhanced MRI was suitable for detecting hepatic SOS. In addition, we conclude that T1 relaxation time correlated with OATP1.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade A

P-Reviewer: Sijens PE S-Editor: Li L L-Editor: A P-Editor: Zhang L

References
1.  Valla DC, Cazals-Hatem D. Sinusoidal obstruction syndrome. Clin Res Hepatol Gastroenterol. 2016;40:378-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
2.  Rubbia-Brandt L. Sinusoidal obstruction syndrome. Clin Liver Dis. 2010;14:651-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 75]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
3.  Jacobs P, Miller JL, Uys CJ, Dietrich BE. Fatal veno-occlusive disease of the liver after chemotherapy, whole-body irradiation and bone marrow transplantation for refractory acute leukaemia. S Afr Med J. 1979;55:5-10.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Mohty M, Malard F, Abecassis M, Aerts E, Alaskar AS, Aljurf M, Arat M, Bader P, Baron F, Bazarbachi A, Blaise D, Ciceri F, Corbacioglu S, Dalle JH, Duarte RF, Fukuda T, Huynh A, Masszi T, Michallet M, Nagler A, NiChonghaile M, Pagluica T, Peters C, Petersen FB, Richardson PG, Ruutu T, Savani BN, Wallhult E, Yakoub-Agha I, Carreras E. Sinusoidal obstruction syndrome/veno-occlusive disease: current situation and perspectives-a position statement from the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 2015;50:781-789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 251]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
5.  Han NY, Park BJ, Sung DJ, Kim MJ, Cho SB, Lee CH, Jang YJ, Kim SY, Kim DS, Um SH, Won NH, Yang KS. Chemotherapy-induced focal hepatopathy in patients with gastrointestinal malignancy: gadoxetic acid--enhanced and diffusion-weighted MR imaging with clinical-pathologic correlation. Radiology. 2014;271:416-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
6.  Zhao J, van Mierlo KMC, Gómez-Ramírez J, Kim H, Pilgrim CHC, Pessaux P, Rensen SS, van der Stok EP, Schaap FG, Soubrane O, Takamoto T, Viganò L, Winkens B, Dejong CHC, Olde Damink SWM; Chemotherapy-Associated Liver Injury (CALI) consortium. Systematic review of the influence of chemotherapy-associated liver injury on outcome after partial hepatectomy for colorectal liver metastases. Br J Surg. 2017;104:990-1002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 64]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
7.  Rubbia-Brandt L, Audard V, Sartoretti P, Roth AD, Brezault C, Le Charpentier M, Dousset B, Morel P, Soubrane O, Chaussade S, Mentha G, Terris B. Severe hepatic sinusoidal obstruction associated with oxaliplatin-based chemotherapy in patients with metastatic colorectal cancer. Ann Oncol. 2004;15:460-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 752]  [Cited by in F6Publishing: 736]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
8.  Zorzi D, Laurent A, Pawlik TM, Lauwers GY, Vauthey JN, Abdalla EK. Chemotherapy-associated hepatotoxicity and surgery for colorectal liver metastases. Br J Surg. 2007;94:274-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 369]  [Cited by in F6Publishing: 357]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
9.  Macedo FI, Makarawo T. Colorectal hepatic metastasis: Evolving therapies. World J Hepatol. 2014;6:453-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
10.  Bismuth H, Adam R. Reduction of nonresectable liver metastasis from colorectal cancer after oxaliplatin chemotherapy. Semin Oncol. 1998;25:40-46.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Nakano H, Oussoultzoglou E, Rosso E, Casnedi S, Chenard-Neu MP, Dufour P, Bachellier P, Jaeck D. Sinusoidal injury increases morbidity after major hepatectomy in patients with colorectal liver metastases receiving preoperative chemotherapy. Ann Surg. 2008;247:118-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 329]  [Cited by in F6Publishing: 339]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
12.  Aloia T, Sebagh M, Plasse M, Karam V, Lévi F, Giacchetti S, Azoulay D, Bismuth H, Castaing D, Adam R. Liver histology and surgical outcomes after preoperative chemotherapy with fluorouracil plus oxaliplatin in colorectal cancer liver metastases. J Clin Oncol. 2006;24:4983-4990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 395]  [Cited by in F6Publishing: 381]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
13.  Castera L. Invasive and non-invasive methods for the assessment of fibrosis and disease progression in chronic liver disease. Best Pract Res Clin Gastroenterol. 2011;25:291-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 103]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
14.  Piccinino F, Sagnelli E, Pasquale G, Giusti G. Complications following percutaneous liver biopsy. A multicentre retrospective study on 68,276 biopsies. J Hepatol. 1986;2:165-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 850]  [Cited by in F6Publishing: 779]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
15.  Soubrane O, Brouquet A, Zalinski S, Terris B, Brézault C, Mallet V, Goldwasser F, Scatton O. Predicting high grade lesions of sinusoidal obstruction syndrome related to oxaliplatin-based chemotherapy for colorectal liver metastases: correlation with post-hepatectomy outcome. Ann Surg. 2010;251:454-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 138]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
16.  Oki E, Kakeji Y, Taketomi A, Yamashita Y, Ohgaki K, Harada N, Iguchi T, Shibahara K, Sadanaga N, Morita M, Maehara Y. Transient elastography for the prediction of oxaliplatin-associated liver injury in colon cancer patients: a preliminary analysis. J Gastrointest Cancer. 2008;39:82-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
17.  Han NY, Park BJ, Kim MJ, Sung DJ, Cho SB. Hepatic Parenchymal Heterogeneity on Contrast-enhanced CT Scans Following Oxaliplatin-based Chemotherapy: Natural History and Association with Clinical Evidence of Sinusoidal Obstruction Syndrome. Radiology. 2015;276:766-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
18.  Ünal E, Karaosmanoğlu AD, Ozmen MN, Akata D, Karcaaltincaba M. Hepatobiliary phase liver MR imaging findings after Oxaliplatin-based chemotherapy in cancer patients. Abdom Radiol (NY). 2018;43:2321-2328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
19.  Hong EK, Joo I, Park J, Lee K. Assessment of hepatic sinusoidal obstruction syndrome with intravoxel incoherent motion diffusion-weighted imaging: An experimental study in a rat model. J Magn Reson Imaging. 2020;51:81-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
20.  Lyu J, Yang G, Mei Y, Guo L, Guo Y, Zhang X, Xu Y, Feng Y. Non-Gaussian Diffusion Models and T(1) rho Quantification in the Assessment of Hepatic Sinusoidal Obstruction Syndrome in Rats. J Magn Reson Imaging. 2020;52:1110-1121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
21.  DeLeve LD, Valla DC, Garcia-Tsao G; American Association for the Study Liver Diseases. Vascular disorders of the liver. Hepatology. 2009;49:1729-1764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 602]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
22.  Joo I, Lee JM. Recent Advances in the Imaging Diagnosis of Hepatocellular Carcinoma: Value of Gadoxetic Acid-Enhanced MRI. Liver Cancer. 2016;5:67-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
23.  Van Beers BE, Pastor CM, Hussain HK. Primovist, Eovist: what to expect? J Hepatol. 2012;57:421-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 298]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
24.  Kudo M. Will Gd-EOB-MRI change the diagnostic algorithm in hepatocellular carcinoma? Oncology. 2010;78 Suppl 1:87-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 56]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
25.  Shin NY, Kim MJ, Lim JS, Park MS, Chung YE, Choi JY, Kim KW, Park YN. Accuracy of gadoxetic acid-enhanced magnetic resonance imaging for the diagnosis of sinusoidal obstruction syndrome in patients with chemotherapy-treated colorectal liver metastases. Eur Radiol. 2012;22:864-871.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
26.  Bae KE, Kim SY, Lee SS, Kim KW, Won HJ, Shin YM, Kim PN, Lee MG. Assessment of hepatic function with Gd-EOB-DTPA-enhanced hepatic MRI. Dig Dis. 2012;30:617-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
27.  Kamimura K, Fukukura Y, Yoneyama T, Takumi K, Tateyama A, Umanodan A, Shindo T, Kumagae Y, Ueno S, Koriyama C, Nakajo M. Quantitative evaluation of liver function with T1 relaxation time index on Gd-EOB-DTPA-enhanced MRI: comparison with signal intensity-based indices. J Magn Reson Imaging. 2014;40:884-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 57]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
28.  Yang L, Ding Y, Rao S, Chen C, Zeng M. T(1) Mapping on Gd-EOB-DTPA-Enhanced MRI for the Prediction of Oxaliplatin-Induced Liver Injury in a Mouse Model. J Magn Reson Imaging. 2021;53:896-902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
29.  Ding Y, Rao SX, Zhu T, Chen CZ, Li RC, Zeng MS. Liver fibrosis staging using T1 mapping on gadoxetic acid-enhanced MRI compared with DW imaging. Clin Radiol. 2015;70:1096-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
30.  Giraudeau C, Leporq B, Doblas S, Lagadec M, Pastor CM, Daire JL, Van Beers BE. Gadoxetate-enhanced MR imaging and compartmental modelling to assess hepatocyte bidirectional transport function in rats with advanced liver fibrosis. Eur Radiol. 2017;27:1804-1811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
31.  Sheng RF, Wang HQ, Yang L, Jin KP, Xie YH, Fu CX, Zeng MS. Assessment of liver fibrosis using T1 mapping on Gd-EOB-DTPA-enhanced magnetic resonance. Dig Liver Dis. 2017;49:789-795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
32.  Yoneda N, Matsui O, Ikeno H, Inoue D, Yoshida K, Kitao A, Kozaka K, Kobayashi S, Gabata T, Ikeda H, Nakamura K, Ohta T. Correlation between Gd-EOB-DTPA-enhanced MR imaging findings and OATP1B3 expression in chemotherapy-associated sinusoidal obstruction syndrome. Abdom Imaging. 2015;40:3099-3103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
33.  Lagadec M, Doblas S, Giraudeau C, Ronot M, Lambert SA, Fasseu M, Paradis V, Moreau R, Pastor CM, Vilgrain V, Daire JL, Van Beers BE. Advanced fibrosis: Correlation between pharmacokinetic parameters at dynamic gadoxetate-enhanced MR imaging and hepatocyte organic anion transporter expression in rat liver. Radiology. 2015;274:379-386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
34.  Dyvorne HA, Jajamovich GH, Bane O, Fiel MI, Chou H, Schiano TD, Dieterich D, Babb JS, Friedman SL, Taouli B. Prospective comparison of magnetic resonance imaging to transient elastography and serum markers for liver fibrosis detection. Liver Int. 2016;36:659-666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
35.  Liao YS, Lee LW, Yang PH, Kuo LM, Kuan LY, Tseng WI, Hwang DW. Assessment of liver cirrhosis for patients with Child's A classification before hepatectomy using dynamic contrast-enhanced MRI. Clin Radiol. 2019;74:407.e11-407.e17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
36.  Ding Y, Rao SX, Chen C, Li R, Zeng MS. Potential of Gd-EOB-DTPA-enhanced MR imaging for evaluation of bile duct ligation-induced liver injury in rabbits. Hepatol Int. 2015;9:303-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
37.  Chen BB, Hsu CY, Yu CW, Wei SY, Kao JH, Lee HS, Shih TT. Dynamic contrast-enhanced magnetic resonance imaging with Gd-EOB-DTPA for the evaluation of liver fibrosis in chronic hepatitis patients. Eur Radiol. 2012;22:171-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 83]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
38.  Wu Z, Cheng ZL, Yi ZL, Xie MW, Zeng H, Lu LJ, Xu X, Shen J. Assessment of Nonalcoholic Fatty Liver Disease in Rats Using Quantitative Dynamic Contrast-Enhanced MRI. J Magn Reson Imaging. 2017;45:1485-1493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
39.  Hiraoka A, Kumada T, Michitaka K, Kudo M. Newly Proposed ALBI Grade and ALBI-T Score as Tools for Assessment of Hepatic Function and Prognosis in Hepatocellular Carcinoma Patients. Liver Cancer. 2019;8:312-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
40.  Pereyra D, Rumpf B, Ammann M, Perrodin SF, Tamandl D, Haselmann C, Stift J, Brostjan C, Laengle F, Beldi G, Gruenberger T, Starlinger P. The Combination of APRI and ALBI Facilitates Preoperative Risk Stratification for Patients Undergoing Liver Surgery After Neoadjuvant Chemotherapy. Ann Surg Oncol. 2019;26:791-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 23]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
41.  Zhou ZP, Long LL, Qiu WJ, Cheng G, Huang LJ, Yang TF, Huang ZK. Evaluating segmental liver function using T1 mapping on Gd-EOB-DTPA-enhanced MRI with a 3.0 Tesla. BMC Med Imaging. 2017;17:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
42.  Park SH, Lee SS, Sung JY, Na K, Kim HJ, Kim SY, Park BJ, Byun JH. Noninvasive assessment of hepatic sinusoidal obstructive syndrome using acoustic radiation force impulse elastography imaging: A proof-of-concept study in rat models. Eur Radiol. 2018;28:2096-2106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]