Skip to main content
Erschienen in: Abdominal Radiology 11/2020

Open Access 09.10.2020 | Special Section : Diffuse Liver Disease

Quantification of liver function using gadoxetic acid-enhanced MRI

verfasst von: Sarah Poetter-Lang, Nina Bastati, Alina Messner, Antonia Kristic, Alexander Herold, Jacqueline C. Hodge, Ahmed Ba-Ssalamah

Erschienen in: Abdominal Radiology | Ausgabe 11/2020

Abstract

The introduction of hepatobiliary contrast agents, most notably gadoxetic acid (GA), has expanded the role of MRI, allowing not only a morphologic but also a functional evaluation of the hepatobiliary system. The mechanism of uptake and excretion of gadoxetic acid via transporters, such as organic anion transporting polypeptides (OATP1,3), multidrug resistance-associated protein 2 (MRP2) and MRP3, has been elucidated in the literature. Furthermore, GA uptake can be estimated on either static images or on dynamic imaging, for example, the hepatic extraction fraction (HEF) and liver perfusion. GA-enhanced MRI has achieved an important role in evaluating morphology and function in chronic liver diseases (CLD), allowing to distinguish between the two subgroups of nonalcoholic fatty liver diseases (NAFLD), simple steatosis and nonalcoholic steatohepatitis (NASH), and help to stage fibrosis and cirrhosis, predict liver transplant graft survival, and preoperatively evaluate the risk of liver failure if major resection is planned. Finally, because of its noninvasive nature, GA-enhanced MRI can be used for long-term follow-up and post-treatment monitoring. This review article aims to describe the current role of GA-enhanced MRI in quantifying liver function in a variety of hepatobiliary disorders.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALBI
Albumin-Bilirubin score
AUROC
Area under the receiver operator characteristic curve
CACLD
Compensated advanced chronic liver diseases
CASH
Chemotherapy-associated steatohepatitis
CEI
Contrast enhancement index
CES
Contrast enhancement spleen index
CLD
Chronic liver diseases
CM
Contrast media
CPS
Child–Pugh score
CRC
Colorectal cancer
CT
Computed tomography
CTP
Child–Turcotte–Pugh score
CUI
Contrast uptake index
DACLD
Decompensated advanced chronic liver diseases
DWI
Diffusion-weighted imaging
ER
Enhancement ratio
FLIS
Functional liver imaging score
functFLR
Functional future liver remnant
GA
Gadoxetic acid
HBP
Hepatobiliary phase
HBV
Hepatitis B virus
HCC
Hepatocellular carcinoma
HCV
Hepatitis C virus
HE
Hepatic encephalopathy
HEF
Hepatic extraction fraction
HUI
Hepatic uptake index
ICG
Indocyanine green clearance
LSI
Liver-to-spleen contrast index
MDCT
Multidetector computed tomography
MELD
Model of End-Stage Liver Disease score
MRCP
Magnetic resonance cholangiopancraticography
MRE
MR elastography
MRI
Magnetic resonance imaging
MRP
Multidrug resistance-associated protein
NAFLD
Nonalcoholic fatty liver diseases
NASH
Nonalcoholic steatohepatitis
OATP
Organic anion transporting polypeptides
PDFF
Proton density fat fraction
PHLF
Post-hepatectomy liver failure
PHLI
Post-hepatectomy liver insufficiency
RLE
Relative liver enhancement
SOS
Sinusoidal obstruction syndrome
SPIO
Superparamagnetic iron oxide
SV
Splenic volume
TSM
Transient severe motion
US
Ultrasound
VB
Variceal bleeding

Introduction

Imaging modalities such as ultrasound (US), computed tomography (CT), and conventional MRI are limited in their ability to detect chronic liver diseases (CLD) in their early stage, as structural changes often occur relatively late in the course of these diseases [1]. In recent years, the value of MRI in the diagnostic workup of hepatobiliary disorders has been significantly enhanced by three groups of contrast agents that increase the conspicuity of either focal or diffuse pathologic alterations and may be able to assess liver function [2].
(i)
Conventional gadolinium chelates (nontissue-specific extracellular contrast agents) similar to iodinated contrast on CT, are typically given as a rapid intravenous bolus followed by dynamic imaging. Timing is critical to capture the arterial, portal venous, and equilibrium phases. Generalized liver enhancement reflects tissue perfusion, allowing the distinction between healthy and diseased liver, and thus helping to detect and characterize focal liver lesions. Likewise, conventional extracellular MR contrast agents are largely excreted via the kidneys [3, 4].
 
(ii)
Gadolinium-based hepatobiliary contrast agents have dual uptake and excretion. Hepatocyte uptake, accounting for bile duct excretion, varies depending upon the specific hepatobiliary contrast agent, e.g., only 2–4% of gadobenate versus 50% of gadoxetic acid. The remaining injected dose behaves like extracellular contrast agents. Image acquisition after rapid bolus injection of gadobenate occurs much later (> 60 min rather than 20 min for gadoxetic acid), since slower hepatocyte uptake means increased time to accumulate adequate hepatobiliary contrast agent within the liver [2, 3, 5].
 
(iii)
Superparamagnetic iron oxide (SPIO) contrast agents that are taken up by the Kupffer cells of the reticuloendothelial system [2].
 
In this article, we will focus on hepatobiliary contrast agents, and, in particular, gadoxetic acid (Gd-EOB-DTPA), also commercially available under the brand name Primovist® in the EU or Eovist® in the US. The multiparametric ability of gadoxetic acid-enhanced MR imaging provides morphologic and functional information about the hepatobiliary system, simultaneously. Multiparametric MRI is a comprehensive MRI that includes T1-, T2-weighted, and proton density fat fraction (PDFF) images, as well as magnetic resonance cholangiopancraticography (MRCP), diffusion-weighted imaging (DWI), and contrast-enhanced dynamic imaging, with a 20-min hepatobiliary phase and, eventually, additional MR elastography (MRE) depending on the radiologic center. The compilation of all these signal characteristics enables the determination of the composition and properties of focal or diffuse liver pathologies [6]. As a gadolinium-based paramagnetic MR contrast agent with dual elimination, approximately 50% of gadoxetic acid is excreted by the kidneys through glomerular filtration, while the other 50% is taken up by functional hepatocytes and excreted into the biliary system [7, 8]. After intravenous injection, GA is dispersed into the intra- and extravascular compartments during the arterial and portal venous phases, similar to conventional gadolinium chelates. But, subsequently, it is actively taken up by the hepatocytes during the transitional (3–5 min) and hepatobiliary phases (HBPs), 20 min after injection. Therefore, GA-enhanced MRI allows the synchronous evaluation of the hepatic vessels, biliary tree, and focal liver lesions [913] in addition to regional and total excretory liver function [14].
Evidence from basic research indicates that the uptake and excretion of GA is mediated by transmembrane transporters on hepatocytes [15, 16]. Gadoxetic acid enters the hepatocytes via active transport by organic anion transporting polypeptides (OATP1B1/3) and is excreted into the biliary tree via MRP2 [17]. Organic acid efflux from hepatocytes may also occur through the sinusoidal membrane because of bidirectional transport with OATP1B1/3 and MRP3 [17]. These insights into the cellular mechanism of transportation have led to a better understanding of how this unique contrast agent acts, as well as enabling the correlation of radiologic with histologic features of certain focal liver lesions, and also aiding in the staging of diffuse CLD [16, 18]. There are two main factors that determine the cellular uptake and excretion of gadoxetic acid (Fig. 1). First, alterations in OATPs (OATP1B1 and OATP1B3) expression, affect gadoxetic acid uptake [16]. Second, it also reflects the total number of hepatocytes. For instance, the decreased relative liver enhancement (RLE) in fibrosis and cirrhosis results from the fibrotic replacement of some normal hepatocytes of the liver parenchyma [19]. In particular, gadoxetic acid enhancement in the hepatobiliary phase (HBP) reflects the net difference between uptake and excretory transporter activity [20]. Based upon a variety of pharmacokinetic properties, including liver perfusion, vascular permeability, extracellular diffusion, and hepatocyte transporter expression, GA-enhanced MRI enables us to obtain a combination of morphologic and functional data simultaneously in normal and diseased hepatobiliary systems [2125]. Therefore, gadoxetic acid is now considered a noninvasive biomarker of hepatobiliary disorders [6, 16].
In this article, we will describe the role of gadoxetic acid in the assessment of liver function in patients with diffuse CLDs using quantitative and semi-quantitative (i.e., qualitative features) scoring systems.

Quantitative assessment of liver function

Visual analysis of the 20-min HBP GA-MR images provides the simplest estimate of liver function and has become an integral part of the radiology report [2629]. More precise but tedious methods include (Emax), time to peak (Tpeak), and elimination half-life (t1/2), which are semi-quantitative, and liver and vascular input concentration versus time curves, which are quantitative [8]. Additional further quantitative parameters include liver blood flow, blood volume, arterial and portal venous perfusion, and hepatocyte extraction fraction obtained by compartmental analysis or deconvolution [14, 24]. Despite their potential to become biomarkers of liver function, quantitative parameters remain limited by complex mathematical modeling, long examination times, and respiratory artifacts. Furthermore, with no standardization across MR scanners, no threshold values that could define normal and abnormal measures have been established. Therefore, these methods are still confined to research purposes.
Several practical and simple quantitative imaging biomarkers of liver function have been introduced, obviating the need for the complex methods used in the animal models and human trials mentioned above. The relative liver enhancement (RLE), hepatic uptake index (HUI), contrast uptake index (CUI), and liver-to-spleen contrast index (LSI) [18, 29, 30] are such methods, relying on only two sequences: noncontrast, and 20-min HBP, which are both part of the routine MRI liver protocol. A simple equation yields any of these biomarkers, each of which has been shown to correlate with liver function parameters (Fig. 2). Most importantly, Beer et al. [18] found that all four of these scores showed a strong positive correlation with each other, answering the fierce debate as to which of these methods was superior [18]. Furthermore, our group confirmed that all four MRI-derived HPB scores correlated moderately with liver disease severity, as assessed by the Albumin-Bilirubin (ALBI) score, Model of End-Stage Liver Disease (MELD), and Child–Turcotte–Pugh (CTP) scores. These parameters were highly reproducible, with high inter-reader and intra-reader agreement. Two additional observations from this study should be mentioned. All four MRI parameters were fairly accurate at separating patients with significantly impaired liver function (MELD score ≥ 15) versus less-impaired (MELD < 15) liver function. This is of the outmost clinical importance, since a MELD [31] score of 15 is the threshold for liver transplantation, where the risk of dying from liver cirrhosis is greater than the post-transplantation mortality. Next, what stands out when considering these MR parameters in detail is the relatively high positive predictive value that each achieved, ranging from 0.876 for the RLE to 0.911 for the HUI. However, all these scores suffered from low negative predictive values, ranging between 0.447 and 0.532, which highlights their ability to validate rather than rule out liver dysfunction.
The new development of T1 mapping has more research than clinical applications, at present. There is a linear relationship between T1 relaxation times and contrast agent concentration. Therefore, T1 relaxation times should be more accurate than SI measurements derived from conventional T1 images [32, 33]. Haimerl et al. [34] have shown that GA-enhanced T1 relaxometry can be useful in diagnosing patients with impaired liver function or chronic liver disease, which they correlated with the Child–Pugh classification [35], the MELD score [31], and the ICG clearance test [36]. However, it should be noted that the relationship between T1 and GA concentration is somewhat more complex because GA is present in at least two compartments during the HBP, i.e., within hepatocytes and bile ducts, and may have different relaxivity rates within each compartment.
Although up to nine ROIs may be drawn, one for each segment, if 4a and 4b are considered as separate segments, four ROIs provide accurate values and are sufficient to reduce the heterogeneity seen in CLDs [24]. On a single level, ROIs of 2.0–5.0 cm2 encircle random, as-large-as-possible homogenous areas, avoiding large vessels, bile ducts, and focal lesions of both liver lobes, in addition to the spleen and nonatrophied left erector spinae. The mean value of these four liver ROIs is calculated.
Principally, it is still a subject of debate whether magnetic field strength (e.g., 1.5 or 3.0 Tesla (T)) influences quantitative MRI assessment, especially if contrast agents have been administered. Recently, Theilig et al. [37] reported that the HBP-derived RLE is constant over serial examinations, different scanner brands, as well as various field strengths. It should be noted that SI standardized to air, i.e., the signal-to-noise ratio, rather than absolute SI, is what matters. However, researchers should proceed with caution since 3 T MRI is known to give higher contrast-to-noise ratios than 1.5 T MRI for the liver parenchyma on contrast-enhanced MRI.

Potential clinical uses for GA-enhanced MRI measures of liver function

Nonalcoholic fatty liver disease (NAFLD)

Chronic liver diseases (CLD) are a major worldwide health problem. Although the prevalence of CLD from most etiologies has been stable or has even substantially declined, particularly in the case of hepatitis C virus (HCV) or hepatitis B virus (HBV) due to eradication or vaccination therapy, respectively [38], the prevalence of NAFLD has steadily increased, and is now the leading cause of CLD worldwide, affecting 80 to 100 million individuals in the USA alone [39, 40]. Therefore the early diagnosis of NAFLD and accurate assessment of liver disease severity are the keys to optimal patient management, where early treatment and lifestyle modification can arrest, and even reverse, deranged hepatic function [38] and histopathology [41].
Bastati et al. [42] showed that the RLE, after GA administration, can differentiate simple steatosis patients from nonalcoholic steatohepatitis (NASH) patients. This method using cutoff value of 1.24 achieved an area under the receiver operator characteristic curve (AUROC) of 0.85 (95% CI 0.75–0.91). The authors suggested that this method may be a useful screening tool in selecting potential NAFLD patients for liver biopsy. However, despite its high sensitivity (97%), the specificity of RLE was only 63%, likely due to the liver fibrosis that was present in 68% of the cohort. As in many CLDs, fibrosis is a well-known confounder that decreases the RLE [43] (Fig. 3a–e). Similar, previous, retrospective studies [44, 45] and unpublished prospective data from our group confirmed the above-mentioned results.

Liver fibrosis

Liver fibrosis is a key determinant in the natural history of chronic liver diseases (CLDs) [46]. Per definition, liver fibrosis is a healing response to myriad injuries that result in CLD. Common insults include viral (A, B, C and D, E) and nonviral infections, alcohol, and NAFLD, while congenital hepatic fibrosis, Wilson’s disease, and a host of cholestatic liver diseases [47] are among the rare etiologies. Progressive liver fibrosis leads to end-stage liver cirrhosis as a result of fibrogenesis [48]. Moreover, because fibrosis may be reversible, early diagnosis is imperative so that early therapy can be instituted. Therefore, knowing the extent and degree of liver fibrosis in CLD patients is of clinical importance, as it can influence patient´s prognosis, surveillance, and treatment [48]. Liver biopsy currently remains the reference standard for the detection and staging of liver fibrosis, despite the risks associated with its invasiveness, inter-observer variability, potential for sampling errors, and poor patient acceptance, which makes it inappropriate for long-term monitoring or follow-up [4951]. Routine biochemical and hematologic tests fail to help quantify liver fibrosis in approximately 50% of patients [51, 52]. There is a large body of literature describing the role of gadoxetic acid-enhanced MRI in the staging of liver fibrosis, using easily obtainable quantitative measures [53]. Feier et al. [54] found that RLE is a good discriminator for the presence of hepatic fibrosis, and Watanabe et al. have found that the contrast enhancement index (CEI) is an efficient biomarker for staging hepatic fibrosis [55]. Further studies have shown that RLE is an independent predictor of fibrosis and is highly accurate for staging F2 fibrosis and cirrhosis (stage F4), with AUROCs of 0.82 and 0.83, respectively [53]. Other groups have found that the hepatobiliary phase (HBP) T1 relaxation time significantly correlated with the fibrosis stage and necroinflammatory activity grade. However, although the HBP T1 relaxation time had a high diagnostic accuracy for stage 3 fibrosis (AUROC of 0.82), its diagnostic accuracy for grade 3 necroinflammatory activity was relatively low (AUROC of 0.68) [56, 57]. The authors found that HBP T1 relaxation times were better at differentiating stage 2 from stage 3 fibrosis than at distinguishing grade 2 from grade 3 necroinflammatory activity (Fig. 4a–c).

Liver cirrhosis

Cirrhosis, the end-stage of hepatic fibrosis, is equivalent to stage 4 fibrosis in the Meta-analysis of Histological Data in Viral Hepatitis (METAVIR) system, or stages 5–6 in the Ishak scoring system [58], as well as in the newly introduced systems, including the Yale (Jain-Garcia), Laennec [59], and PIR systems [60].
Today, we know that cirrhosis is a dynamic process that may or may not progress. Indeed, early cirrhosis may be reversible. Thus, the former one-stage clinical and histologic description of “cirrhosis” is simplistic, and misrepresents current dogma [61]. Therefore, in the clinical setting, the term cirrhosis has been increasingly replaced by the term chronic liver disease (CLD), and thus, it is more important to distinguish between compensated CLD versus decompensated CLD, since, once patients cross this clinical boundary into decompensation, the odds are, by comparison, extremely low that they can avoid liver failure without transplantation or retransplantation [28, 62].
As a cure for chronic hepatitis C in responders where eradication of the viral load exceeds 95% post-treatment [63, 64], the focus of attention has now shifted to the regression of HCV-induced liver fibrosis, cirrhosis, and portal hypertension in these patients. Our group [65] hypothesized that the RLE might identify patients who have persistent hepatic inflammation and fibrosis, as well as portal hypertension despite HCV eradication. This is clinically relevant, as surveillance for portal hypertension and hepatocellular carcinoma are particularly important in patients with CLD. We evaluated changes in relative liver enhancement (RLE) obtained by GA-MRI in the hepatobiliary phase and changes in splenic volume (SV) after hepatitis C virus (HCV) eradication, as well as their predictive value for the development of (further) hepatic decompensation during follow-up. We observed an inverse correlation between the changes in RLE and SV (P < 0.001). In the non-responder patients, there was a decrease in RLE of − 11% (− 25% to − 3%; P = 0.019) and an increase in SV of 23% (7–43%; P = 0.004) (both P < 0.001 versus the responder patients). Interestingly, GA-MRI-nonresponse was associated with a substantially increased risk of (further) hepatic decompensation two years after the end of treatment: 80% versus 8%; P < 0.001. We concluded that GA-MRI might distinguish between individuals at low- and high-risk for (further) hepatic decompensation (GA-MRI-nonresponse) after HCV eradication. This could allow for individualized surveillance strategies.
The degree of parenchymal uptake and biliary excretion in GA-MRI may be useful for the evaluation of liver function as cirrhosis evolves [28, 66]. Tamada et al. found that hepatic parenchymal enhancement, i.e., RLE on GA-MRI, is affected by the severity of cirrhosis [67]. Tsuda and others, using RLE, also found that the liver’s signal intensity on the HBP of GA-MRI was lower in cirrhotic than in normal livers [68]. Conversely, Kanki et al. reported that the RLE derived from GA-MRI did not necessarily decrease with increased morphologic features of cirrhosis [69]. Other groups reported the value of T1 relaxometry in cirrhosis [70, 71], either unenhanced, or, after contrast media (CM) injection [72]. More recently, Sandrasegaran K. et al. [73] conducted a retrospective study to determine the value of quantitative parameters of GA-MRI in predicting prognosis in cirrhotics. Examining a cohort of 63 patients who had GA-MRI and a two-year clinical follow-up, they calculated the enhancement ratio (ER), contrast enhancement index (CEI), and contrast enhancement spleen index (CES). Comparing the usefulness of these parameters to clinical scores, such as the Child–Pugh score (CPS) and the model for end-stage liver disease (MELD) score, for predicting adverse outcomes, such as variceal bleeding (VB), hepatic encephalopathy (HE), and mortality at two years, they concluded that an ER of 15 or a CES of 20 were equivalent or better predictors of major morbidity and mortality than these commonly used clinical scores in cirrhotic patients.

Preoperative evaluation of remnant liver function

Several studies have addressed the value of preoperative GA-MRI in predicting the risk of liver failure should major liver resection be performed [7476]. This estimate of hepatic function reserve is crucial to avoid post-hepatectomy liver insufficiency (PHLI) [36, 74].
It is well known that, in the setting of metastatic liver disease, chemotherapy can adversely affect liver function. In particular, metastatic colorectal cancer (CRC) patients who receive specific drugs, such as oxaliplatin and or irinotecan, are predisposed to develop chemotherapy-associated steatohepatitis (CASH) and sinusoidal obstruction syndrome (SOS) [77, 78].
In those patients with less than 25–30% remnant liver function, PHLI is the major cause of postoperative morbidity and mortality. Because morphologic volume may overestimate functional volume [30, 74], volumetry alone (based on CT or conventional MRI) may not deliver an accurate prediction of PHLI in the presence of CLDs, where steatosis and/or fibrosis likely exist.
GA-MRI has proven superior for the estimation of both global and regional function, and studies have shown it to be a more reliable prognosticator than indocyanine green clearance (ICG) [30, 36].
In a retrospective study, our group [74] looked at how well functional future liver remnant (functFLR), as calculated from the RLE on GA-MRI and volumetry on multidetector computed tomography (MDCT) done within 10 weeks of a planned major resection, predicted post-hepatectomy liver failure (PHLF) following major liver resection compared to well-established clinical tests. In a multivariate analysis, we found that a decreased functFLR was independently associated with the probability of PHLF (0.561; P = 0.002). MRI software development is needed to eliminate the use of CT for volumetry, which is the major drawback of this technique.
Furthermore, Cho et al. showed that the RLE is an accurate discriminator for predicting which patients will develop PHLI after major liver resection [79]. Wibmer et al. found that decreased RLE in liver surgery candidates was associated with a higher risk of PHLI and perioperative mortality [30].
Kim DK et al. concluded that GA-MRI-derived measurements predicted PHLF better than the ICG clearance test in hepatocellular carcinoma (HCC) patients who underwent hepatectomy [36].

Orthotopic liver transplantation (OLT)

Currently, GA-MRI and MR cholangiography have been increasingly used to assess liver graft dysfunction, since, unlike conventional gadolinium chelates, these techniques can provide both anatomic and functional information [80, 81]. Our group reported that, among the above-mentioned quantitative parameters derived from GA-MRI, the RLE is one of the most promising for graft evaluation [27]. Global and regional contrast media uptake and excretion of the graft can be calculated, providing the probability of graft survival. We could show that the RLE 20 min after contrast injection was directly related to the probability of one-year retransplantation-free survival in proportional hazard regression analysis (P = 0.005) [27].

Qualitative or semi-quantitative assessment of liver function

Qualitative assessment of OLT graft function

Quantitative assessment of liver function using the calculation of the above-mentioned scores, such as RLE, is more tedious and time-consuming and normally cannot be performed as part of a routine MR examination. Furthermore, radiologists are used to assessing morphologic MR features and are less familiar with calculating lengthy and difficult equations. Therefore, Bastati et al. [27] introduced the functional liver imaging score (FLIS), derived from three hepatobiliary phase features on gadoxetic acid-enhanced MRI, each ranging from 0 to 2 on an ordinal scale. Following visual assessment of the liver parenchymal enhancement quality (EnQS), the rate of biliary contrast excretion (ExQS), and the persistence of contrast within the portal vein (PVsQS), the three scores are summed, and can range from 0 to 6, yielding the FLIS. Hence, the use of the term ‘semi-quantitative’ to describe the subjective and objective components that contribute to the FLIS (Fig. 5). Because the FLIS requires no signal intensity measurements, equations, or specific software, and is independent of MRI field strength and vendor, it can easily be incorporated into routine clinical practice.

Qualitative assessment of liver function on CLD

Imaging techniques, including US, CT, and conventional MRI, have been used to diagnose cirrhosis and its sequelae based on morphological features. However, the prototypical features of cirrhosis bear no relationship to objective (i.e., functional) parameters of cirrhosis severity [6, 82]. Just as the FLIS, derived from GA-MRI, allows estimation of liver function in OLT grafts, it can be used in patients with chronic liver disease (CLD).
Previously described methods with which to assess the hepatobiliary phase uptake include the relative liver enhancement, the hepatic uptake index, the contrast enhancement index, and T1 relaxation values [18, 22, 24]. These methods all require complex computations and have vendor, field strength, and sequence dependencies that complicate their clinical application. FLIS is more practical for routine use than quantitative parameters, as it requires no SI measurements or calculations. The use of such an easy and reproducible score in clinical practice may potentially lead to better management of patients with CLD.
In a retrospective study, 265 patients with CLD, who had undergone GA-MRI, were assigned a FLIS score ranging from 0 to 6, based on the sum of three hepatobiliary phase features, as described above [28]. Patients were stratified into the following three groups according to fibrosis stage and a presence or history of hepatic decompensation: nonadvanced CLD; compensated advanced CLD (CACLD); and decompensated advanced CLD (DACLD). The predictive value of FLIS for first and/or further hepatic decompensation and for transplant-free survival, i.e., mortality, was investigated using Kaplan–Meier analysis, log-rank tests, and Cox regression analysis. Intra-observer and inter-observer intraclass correlation coefficients for FLIS were excellent, demonstrating the robustness and reproducibility of this scoring system [28].
The results demonstrated that the FLIS is an independent predictor of liver-related events (e.g., first hepatic decompensation) and transplant-free survival (mortality) in patients with different causes of chronic liver disease (CLD). Both decompensated and compensated patients with CLD with a reduced FLIS showed a three- to seven-fold higher risk of mortality, respectively, even after adjusting for established prognostic factors (Fig. 6a–d).
GA has an excellent track record in focal liver lesion detection and characterization, as well as in liver function estimation. Transient severe motion (TSM) artifacts, which may degrade arterial-phase images in 18% of patients, on average [83], is the main limitation of GA. However, various techniques, including free-breathing, obtaining multiple arterial phases, saline dilution of GA, and/or slower injection rates, have successfully alleviated TSM [84, 85]. Furthermore, the 20-min delay between dynamic and HB phase imaging need not be wasted. By injecting GA at the start of the exam, dynamic imaging, T2 w-images, DWI, MR elastography, and other techniques, such as T2 mapping, can be performed while waiting. GA is preferable to gadobenate, not only because of the shorter interval between contrast injection and late-phase imaging but also because gadobenate’s negligible hepatocyte uptake limits its role in estimating liver function.
In conclusion, the estimation of liver function is a crucial clinical issue for identifying the broad spectrum of hepatobiliary disorders, for monitoring the progression of chronic liver disease (CLD), for determining the optimal therapeutic strategy, as well as for the prevention of post-interventional hepatic failure.
GA-MRI can be used to evaluate liver morphology, as well as calculate several quantitative parameters, including RLE, HUI, CEI, and the semi-quantitative FLIS parameter, which is independent of field strength, exam parameters, scanner type, or vendor.
Open AccessThis 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

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.

e.Med Radiologie

Kombi-Abonnement

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

Literatur
1.
Zurück zum Zitat Van Beers BE, Daire JL, Garteiser P: New imaging techniques for liver diseases. J Hepatol 2015, 62(3):690-700.PubMedCrossRef Van Beers BE, Daire JL, Garteiser P: New imaging techniques for liver diseases. J Hepatol 2015, 62(3):690-700.PubMedCrossRef
2.
Zurück zum Zitat Ba-Ssalamah A, Uffmann M, Saini S, Bastati N, Herold C, Schima W: Clinical value of MRI liver-specific contrast agents: a tailored examination for a confident non-invasive diagnosis of focal liver lesions. European radiology 2009, 19(2):342-357.PubMedCrossRef Ba-Ssalamah A, Uffmann M, Saini S, Bastati N, Herold C, Schima W: Clinical value of MRI liver-specific contrast agents: a tailored examination for a confident non-invasive diagnosis of focal liver lesions. European radiology 2009, 19(2):342-357.PubMedCrossRef
3.
Zurück zum Zitat Gandhi SN, Brown MA, Wong JG, Aguirre DA, Sirlin CB: MR contrast agents for liver imaging: what, when, how. Radiographics 2006, 26(6):1621-1636.PubMedCrossRef Gandhi SN, Brown MA, Wong JG, Aguirre DA, Sirlin CB: MR contrast agents for liver imaging: what, when, how. Radiographics 2006, 26(6):1621-1636.PubMedCrossRef
4.
Zurück zum Zitat Semelka RC, Helmberger TK: Contrast agents for MR imaging of the liver. Radiology 2001, 218(1):27-38.PubMedCrossRef Semelka RC, Helmberger TK: Contrast agents for MR imaging of the liver. Radiology 2001, 218(1):27-38.PubMedCrossRef
5.
Zurück zum Zitat Seale MK, Catalano OA, Saini S, Hahn PF, Sahani DV: Hepatobiliary-specific MR contrast agents: role in imaging the liver and biliary tree. Radiographics : a review publication of the Radiological Society of North America, Inc 2009, 29(6):1725–1748. Seale MK, Catalano OA, Saini S, Hahn PF, Sahani DV: Hepatobiliary-specific MR contrast agents: role in imaging the liver and biliary tree. Radiographics : a review publication of the Radiological Society of North America, Inc 2009, 29(6):1725–1748.
6.
Zurück zum Zitat Ba-Ssalamah A, Qayyum A, Bastati N, Fakhrai N, Herold CJ, Caseiro Alves F: P4 radiology of hepatobiliary diseases with gadoxetic acid-enhanced MRI as a biomarker. Expert Rev Gastroenterol Hepatol 2014, 8(2):147-160.PubMedCrossRef Ba-Ssalamah A, Qayyum A, Bastati N, Fakhrai N, Herold CJ, Caseiro Alves F: P4 radiology of hepatobiliary diseases with gadoxetic acid-enhanced MRI as a biomarker. Expert Rev Gastroenterol Hepatol 2014, 8(2):147-160.PubMedCrossRef
7.
Zurück zum Zitat Schalkx HJ, van Stralen M, Coenegrachts K, van den Bosch MA, van Kessel CS, van Hillegersberg R, van Erpecum KJ, Verkooijen HM, Pluim JP, Veldhuis WB et al: Liver perfusion in dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI): comparison of enhancement in Gd-BT-DO3A and Gd-EOB-DTPA in normal liver parenchyma. European radiology 2014, 24(9):2146-2156.PubMedCrossRef Schalkx HJ, van Stralen M, Coenegrachts K, van den Bosch MA, van Kessel CS, van Hillegersberg R, van Erpecum KJ, Verkooijen HM, Pluim JP, Veldhuis WB et al: Liver perfusion in dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI): comparison of enhancement in Gd-BT-DO3A and Gd-EOB-DTPA in normal liver parenchyma. European radiology 2014, 24(9):2146-2156.PubMedCrossRef
8.
Zurück zum Zitat Zhang T, Runge JH, Lavini C, Stoker J, van Gulik T, Cieslak KP, van Vliet LJ, Vos FM: A pharmacokinetic model including arrival time for two inputs and compensating for varying applied flip-angle in dynamic gadoxetic acid-enhanced MR imaging. PLoS One 2019, 14(8):e0220835.PubMedPubMedCentralCrossRef Zhang T, Runge JH, Lavini C, Stoker J, van Gulik T, Cieslak KP, van Vliet LJ, Vos FM: A pharmacokinetic model including arrival time for two inputs and compensating for varying applied flip-angle in dynamic gadoxetic acid-enhanced MR imaging. PLoS One 2019, 14(8):e0220835.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Zech CJ, Vos B, Nordell A, Urich M, Blomqvist L, Breuer J, Reiser MF, Weinmann HJ: Vascular enhancement in early dynamic liver MR imaging in an animal model: comparison of two injection regimen and two different doses Gd-EOB-DTPA (gadoxetic acid) with standard Gd-DTPA. Invest Radiol 2009, 44(6):305-310.PubMedCrossRef Zech CJ, Vos B, Nordell A, Urich M, Blomqvist L, Breuer J, Reiser MF, Weinmann HJ: Vascular enhancement in early dynamic liver MR imaging in an animal model: comparison of two injection regimen and two different doses Gd-EOB-DTPA (gadoxetic acid) with standard Gd-DTPA. Invest Radiol 2009, 44(6):305-310.PubMedCrossRef
10.
Zurück zum Zitat Lee NK, Kim S, Lee JW, Lee SH, Kang DH, Kim GH, Seo HI: Biliary MR imaging with Gd-EOB-DTPA and its clinical applications. Radiographics : a review publication of the Radiological Society of North America, Inc 2009, 29(6):1707–1724. Lee NK, Kim S, Lee JW, Lee SH, Kang DH, Kim GH, Seo HI: Biliary MR imaging with Gd-EOB-DTPA and its clinical applications. Radiographics : a review publication of the Radiological Society of North America, Inc 2009, 29(6):1707–1724.
11.
Zurück zum Zitat Shimada K, Isoda H, Hirokawa Y, Arizono S, Shibata T, Togashi K: Comparison of gadolinium-EOB-DTPA-enhanced and diffusion-weighted liver MRI for detection of small hepatic metastases. European radiology 2010, 20(11):2690-2698.PubMedCrossRef Shimada K, Isoda H, Hirokawa Y, Arizono S, Shibata T, Togashi K: Comparison of gadolinium-EOB-DTPA-enhanced and diffusion-weighted liver MRI for detection of small hepatic metastases. European radiology 2010, 20(11):2690-2698.PubMedCrossRef
12.
Zurück zum Zitat Seo HJ, Kim MJ, Lee JD, Chung WS, Kim YE: Gadoxetate disodium-enhanced magnetic resonance imaging versus contrast-enhanced 18F-fluorodeoxyglucose positron emission tomography/computed tomography for the detection of colorectal liver metastases. Invest Radiol 2011, 46(9):548-555.PubMedCrossRef Seo HJ, Kim MJ, Lee JD, Chung WS, Kim YE: Gadoxetate disodium-enhanced magnetic resonance imaging versus contrast-enhanced 18F-fluorodeoxyglucose positron emission tomography/computed tomography for the detection of colorectal liver metastases. Invest Radiol 2011, 46(9):548-555.PubMedCrossRef
13.
Zurück zum Zitat Motosugi U, Ichikawa T, Sano K, Sou H, Onohara K, Muhi A, Kitamura T, Amemiya F, Enomoto N, Matsuda M et al: Double-dose gadoxetic Acid-enhanced magnetic resonance imaging in patients with chronic liver disease. Invest Radiol 2011, 46(2):141-145.PubMedCrossRef Motosugi U, Ichikawa T, Sano K, Sou H, Onohara K, Muhi A, Kitamura T, Amemiya F, Enomoto N, Matsuda M et al: Double-dose gadoxetic Acid-enhanced magnetic resonance imaging in patients with chronic liver disease. Invest Radiol 2011, 46(2):141-145.PubMedCrossRef
14.
Zurück zum Zitat Sourbron S, Sommer WH, Reiser MF, Zech CJ: Combined quantification of liver perfusion and function with dynamic gadoxetic acid-enhanced MR imaging. Radiology 2012, 263(3):874-883.PubMedCrossRef Sourbron S, Sommer WH, Reiser MF, Zech CJ: Combined quantification of liver perfusion and function with dynamic gadoxetic acid-enhanced MR imaging. Radiology 2012, 263(3):874-883.PubMedCrossRef
15.
Zurück zum Zitat Ba-Ssalamah A, Antunes C, Feier D, Bastati N, Hodge JC, Stift J, Cipriano MA, Wrba F, Trauner M, Herold CJ et al: Morphologic and Molecular Features of Hepatocellular Adenoma with Gadoxetic Acid-enhanced MR Imaging. Radiology 2015, 277(1):104-113.PubMedCrossRef Ba-Ssalamah A, Antunes C, Feier D, Bastati N, Hodge JC, Stift J, Cipriano MA, Wrba F, Trauner M, Herold CJ et al: Morphologic and Molecular Features of Hepatocellular Adenoma with Gadoxetic Acid-enhanced MR Imaging. Radiology 2015, 277(1):104-113.PubMedCrossRef
16.
Zurück zum Zitat Van Beers BE, Pastor CM, Hussain HK: Primovist, Eovist: what to expect?Journal of hepatology 2012, 57(2):421-429.PubMedCrossRef Van Beers BE, Pastor CM, Hussain HK: Primovist, Eovist: what to expect?Journal of hepatology 2012, 57(2):421-429.PubMedCrossRef
17.
Zurück zum Zitat Tsuboyama T, Onishi H, Kim T, Akita H, Hori M, Tatsumi M, Nakamoto A, Nagano H, Matsuura N, Wakasa K et al: Hepatocellular carcinoma: hepatocyte-selective enhancement at gadoxetic acid-enhanced MR imaging--correlation with expression of sinusoidal and canalicular transporters and bile accumulation. Radiology 2010, 255(3):824-833.PubMedCrossRef Tsuboyama T, Onishi H, Kim T, Akita H, Hori M, Tatsumi M, Nakamoto A, Nagano H, Matsuura N, Wakasa K et al: Hepatocellular carcinoma: hepatocyte-selective enhancement at gadoxetic acid-enhanced MR imaging--correlation with expression of sinusoidal and canalicular transporters and bile accumulation. Radiology 2010, 255(3):824-833.PubMedCrossRef
18.
Zurück zum Zitat Beer L, Mandorfer M, Bastati N, Poetter-Lang S, Tamandl D, Stoyanova DP, Elmer MC, Semmler G, Simbrunner B, Hodge JC et al: Inter- and intra-reader agreement for gadoxetic acid-enhanced MRI parameter readings in patients with chronic liver diseases. Eur Radiol 2019, 29(12):6600-6610.PubMedPubMedCentralCrossRef Beer L, Mandorfer M, Bastati N, Poetter-Lang S, Tamandl D, Stoyanova DP, Elmer MC, Semmler G, Simbrunner B, Hodge JC et al: Inter- and intra-reader agreement for gadoxetic acid-enhanced MRI parameter readings in patients with chronic liver diseases. Eur Radiol 2019, 29(12):6600-6610.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Verloh N, Probst U, Utpatel K, Zeman F, Brennfleck F, Werner JM, Fellner C, Stroszczynski C, Evert M, Wiggermann P et al: Influence of hepatic fibrosis and inflammation: Correlation between histopathological changes and Gd-EOB-DTPA-enhanced MR imaging. PLoS One 2019, 14(5):e0215752.PubMedPubMedCentralCrossRef Verloh N, Probst U, Utpatel K, Zeman F, Brennfleck F, Werner JM, Fellner C, Stroszczynski C, Evert M, Wiggermann P et al: Influence of hepatic fibrosis and inflammation: Correlation between histopathological changes and Gd-EOB-DTPA-enhanced MR imaging. PLoS One 2019, 14(5):e0215752.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Feier D, Balassy C, Bastati N, Stift J, Badea R, Ba-Ssalamah A: Liver fibrosis: histopathologic and biochemical influences on diagnostic efficacy of hepatobiliary contrast-enhanced MR imaging in staging. Radiology 2013, 269(2):460-468.PubMedCrossRef Feier D, Balassy C, Bastati N, Stift J, Badea R, Ba-Ssalamah A: Liver fibrosis: histopathologic and biochemical influences on diagnostic efficacy of hepatobiliary contrast-enhanced MR imaging in staging. Radiology 2013, 269(2):460-468.PubMedCrossRef
21.
Zurück zum Zitat Kukuk GM, Schaefer SG, Fimmers R, Hadizadeh DR, Ezziddin S, Spengler U, Schild HH, Willinek WA: Hepatobiliary magnetic resonance imaging in patients with liver disease: correlation of liver enhancement with biochemical liver function tests. European radiology 2014, 24(10):2482-2490.PubMedCrossRef Kukuk GM, Schaefer SG, Fimmers R, Hadizadeh DR, Ezziddin S, Spengler U, Schild HH, Willinek WA: Hepatobiliary magnetic resonance imaging in patients with liver disease: correlation of liver enhancement with biochemical liver function tests. European radiology 2014, 24(10):2482-2490.PubMedCrossRef
22.
Zurück zum Zitat Verloh N, Haimerl M, Zeman F, Schlabeck M, Barreiros A, Loss M, Schreyer AG, Stroszczynski C, Fellner C, Wiggermann P: Assessing liver function by liver enhancement during the hepatobiliary phase with Gd-EOB-DTPA-enhanced MRI at 3 Tesla. European radiology 2014, 24(5):1013-1019.PubMedCrossRef Verloh N, Haimerl M, Zeman F, Schlabeck M, Barreiros A, Loss M, Schreyer AG, Stroszczynski C, Fellner C, Wiggermann P: Assessing liver function by liver enhancement during the hepatobiliary phase with Gd-EOB-DTPA-enhanced MRI at 3 Tesla. European radiology 2014, 24(5):1013-1019.PubMedCrossRef
23.
Zurück zum Zitat Yoon JH, Lee JM, Kang HJ, Ahn SJ, Yang H, Kim E, Okuaki T, Han JK: Quantitative Assessment of Liver Function by Using Gadoxetic Acid-enhanced MRI: Hepatocyte Uptake Ratio. Radiology 2019, 290(1):125-133.PubMedCrossRef Yoon JH, Lee JM, Kang HJ, Ahn SJ, Yang H, Kim E, Okuaki T, Han JK: Quantitative Assessment of Liver Function by Using Gadoxetic Acid-enhanced MRI: Hepatocyte Uptake Ratio. Radiology 2019, 290(1):125-133.PubMedCrossRef
24.
Zurück zum Zitat Ba-Ssalamah A, Bastati N, Wibmer A, Fragner R, Hodge JC, Trauner M, Herold CJ, Bashir MR, Van Beers BE: Hepatic gadoxetic acid uptake as a measure of diffuse liver disease: Where are we?J Magn Reson Imaging 2017, 45(3):646-659.PubMedCrossRef Ba-Ssalamah A, Bastati N, Wibmer A, Fragner R, Hodge JC, Trauner M, Herold CJ, Bashir MR, Van Beers BE: Hepatic gadoxetic acid uptake as a measure of diffuse liver disease: Where are we?J Magn Reson Imaging 2017, 45(3):646-659.PubMedCrossRef
25.
Zurück zum Zitat Zhou ZP, Long LL, Qiu WJ, Cheng G, Huang LJ, Yang TF, Huang ZK: Comparison of 10- and 20-min hepatobiliary phase images on Gd-EOB-DTPA-enhanced MRI T1 mapping for liver function assessment in clinic. Abdom Radiol (NY) 2017, 42(9):2272-2278.CrossRef Zhou ZP, Long LL, Qiu WJ, Cheng G, Huang LJ, Yang TF, Huang ZK: Comparison of 10- and 20-min hepatobiliary phase images on Gd-EOB-DTPA-enhanced MRI T1 mapping for liver function assessment in clinic. Abdom Radiol (NY) 2017, 42(9):2272-2278.CrossRef
26.
Zurück zum Zitat Zech CJ, Ba-Ssalamah A, Berg T, Chandarana H, Chau GY, Grazioli L, Kim MJ, Lee JM, Merkle EM, Murakami T et al: Consensus report from the 8th International Forum for Liver Magnetic Resonance Imaging. European radiology 2020, 30(1):370-382.PubMedCrossRef Zech CJ, Ba-Ssalamah A, Berg T, Chandarana H, Chau GY, Grazioli L, Kim MJ, Lee JM, Merkle EM, Murakami T et al: Consensus report from the 8th International Forum for Liver Magnetic Resonance Imaging. European radiology 2020, 30(1):370-382.PubMedCrossRef
27.
Zurück zum Zitat Bastati N, Wibmer A, Tamandl D, Einspieler H, Hodge JC, Poetter-Lang S, Rockenschaub S, Berlakovich GA, Trauner M, Herold C et al: Assessment of Orthotopic Liver Transplant Graft Survival on Gadoxetic Acid-Enhanced Magnetic Resonance Imaging Using Qualitative and Quantitative Parameters. Invest Radiol 2016, 51(11):728-734.PubMedCrossRef Bastati N, Wibmer A, Tamandl D, Einspieler H, Hodge JC, Poetter-Lang S, Rockenschaub S, Berlakovich GA, Trauner M, Herold C et al: Assessment of Orthotopic Liver Transplant Graft Survival on Gadoxetic Acid-Enhanced Magnetic Resonance Imaging Using Qualitative and Quantitative Parameters. Invest Radiol 2016, 51(11):728-734.PubMedCrossRef
28.
Zurück zum Zitat Bastati N, Beer L, Mandorfer M, Poetter-Lang S, Tamandl D, Bican Y, Elmer MC, Einspieler H, Semmler G, Simbrunner B et al: Does the Functional Liver Imaging Score Derived from Gadoxetic Acid-enhanced MRI Predict Outcomes in Chronic Liver Disease?Radiology 2020, 294(1):98-107.PubMedCrossRef Bastati N, Beer L, Mandorfer M, Poetter-Lang S, Tamandl D, Bican Y, Elmer MC, Einspieler H, Semmler G, Simbrunner B et al: Does the Functional Liver Imaging Score Derived from Gadoxetic Acid-enhanced MRI Predict Outcomes in Chronic Liver Disease?Radiology 2020, 294(1):98-107.PubMedCrossRef
29.
Zurück zum Zitat Wibmer A, Nolz R, Trauner M, Ba-Ssalamah A: [Functional MR imaging of the liver]. Der Radiologe 2015, 55(12):1057-1066.PubMedCrossRef Wibmer A, Nolz R, Trauner M, Ba-Ssalamah A: [Functional MR imaging of the liver]. Der Radiologe 2015, 55(12):1057-1066.PubMedCrossRef
30.
Zurück zum Zitat Wibmer A, Prusa AM, Nolz R, Gruenberger T, Schindl M, Ba-Ssalamah A: Liver Failure after Major Liver Resection: Risk Assessment by Using Preoperative Gadoxetic Acid-enhanced 3-T MR Imaging. Radiology 2013, 269(3):777-786.PubMedCrossRef Wibmer A, Prusa AM, Nolz R, Gruenberger T, Schindl M, Ba-Ssalamah A: Liver Failure after Major Liver Resection: Risk Assessment by Using Preoperative Gadoxetic Acid-enhanced 3-T MR Imaging. Radiology 2013, 269(3):777-786.PubMedCrossRef
31.
Zurück zum Zitat Wiesner R, Edwards E, Freeman R, Harper A, Kim R, Kamath P, Kremers W, Lake J, Howard T, Merion RM et al: Model for end-stage liver disease (MELD) and allocation of donor livers. Gastroenterology 2003, 124(1):91-96.PubMedCrossRef Wiesner R, Edwards E, Freeman R, Harper A, Kim R, Kamath P, Kremers W, Lake J, Howard T, Merion RM et al: Model for end-stage liver disease (MELD) and allocation of donor livers. Gastroenterology 2003, 124(1):91-96.PubMedCrossRef
32.
Zurück zum Zitat Katsube T, Okada M, Kumano S, Hori M, Imaoka I, Ishii K, Kudo M, Kitagaki H, Murakami T: Estimation of liver function using T1 mapping on Gd-EOB-DTPA-enhanced magnetic resonance imaging. Invest Radiol 2011, 46(4):277-283.PubMedCrossRef Katsube T, Okada M, Kumano S, Hori M, Imaoka I, Ishii K, Kudo M, Kitagaki H, Murakami T: Estimation of liver function using T1 mapping on Gd-EOB-DTPA-enhanced magnetic resonance imaging. Invest Radiol 2011, 46(4):277-283.PubMedCrossRef
33.
Zurück zum Zitat Kim JE, Kim HO, Bae K, Choi DS, Nickel D: T1 mapping for liver function evaluation in gadoxetic acid-enhanced MR imaging: comparison of look-locker inversion recovery and B1 inhomogeneity-corrected variable flip angle method. European radiology 2019, 29(7):3584-3594.PubMedCrossRef Kim JE, Kim HO, Bae K, Choi DS, Nickel D: T1 mapping for liver function evaluation in gadoxetic acid-enhanced MR imaging: comparison of look-locker inversion recovery and B1 inhomogeneity-corrected variable flip angle method. European radiology 2019, 29(7):3584-3594.PubMedCrossRef
34.
Zurück zum Zitat Haimerl M, Fuhrmann I, Poelsterl S, Fellner C, Nickel MD, Weigand K, Dahlke MH, Verloh N, Stroszczynski C, Wiggermann P: Gd-EOB-DTPA-enhanced T1 relaxometry for assessment of liver function determined by real-time (13)C-methacetin breath test. European radiology 2018, 28(9):3591-3600.PubMedCrossRef Haimerl M, Fuhrmann I, Poelsterl S, Fellner C, Nickel MD, Weigand K, Dahlke MH, Verloh N, Stroszczynski C, Wiggermann P: Gd-EOB-DTPA-enhanced T1 relaxometry for assessment of liver function determined by real-time (13)C-methacetin breath test. European radiology 2018, 28(9):3591-3600.PubMedCrossRef
35.
Zurück zum Zitat Peng Y, Qi X, Guo X: Child-Pugh Versus MELD Score for the Assessment of Prognosis in Liver Cirrhosis: A Systematic Review and Meta-Analysis of Observational Studies. Medicine (Baltimore) 2016, 95(8):e2877.CrossRef Peng Y, Qi X, Guo X: Child-Pugh Versus MELD Score for the Assessment of Prognosis in Liver Cirrhosis: A Systematic Review and Meta-Analysis of Observational Studies. Medicine (Baltimore) 2016, 95(8):e2877.CrossRef
36.
Zurück zum Zitat Kim DK, Choi JI, Choi MH, Park MY, Lee YJ, Rha SE, Jung SE: Prediction of Posthepatectomy Liver Failure: MRI With Hepatocyte-Specific Contrast Agent Versus Indocyanine Green Clearance Test. AJR Am J Roentgenol 2018, 211(3):580-587.PubMedCrossRef Kim DK, Choi JI, Choi MH, Park MY, Lee YJ, Rha SE, Jung SE: Prediction of Posthepatectomy Liver Failure: MRI With Hepatocyte-Specific Contrast Agent Versus Indocyanine Green Clearance Test. AJR Am J Roentgenol 2018, 211(3):580-587.PubMedCrossRef
37.
Zurück zum Zitat Theilig D, Elkilany A, Schmelzle M, Muller T, Hamm B, Denecke T, Geisel D: Consistency of hepatocellular gadoxetic acid uptake in serial MRI examinations for evaluation of liver function. Abdom Radiol (NY) 2019, 44(8):2759-2768.CrossRef Theilig D, Elkilany A, Schmelzle M, Muller T, Hamm B, Denecke T, Geisel D: Consistency of hepatocellular gadoxetic acid uptake in serial MRI examinations for evaluation of liver function. Abdom Radiol (NY) 2019, 44(8):2759-2768.CrossRef
38.
Zurück zum Zitat Marcellin P, Gane E, Buti M, Afdhal N, Sievert W, Jacobson IM, Washington MK, Germanidis G, Flaherty JF, Aguilar Schall R et al: Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: a 5-year open-label follow-up study. Lancet 2013, 381(9865):468-475.PubMedCrossRef Marcellin P, Gane E, Buti M, Afdhal N, Sievert W, Jacobson IM, Washington MK, Germanidis G, Flaherty JF, Aguilar Schall R et al: Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: a 5-year open-label follow-up study. Lancet 2013, 381(9865):468-475.PubMedCrossRef
39.
Zurück zum Zitat Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M: Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64(1):73-84.PubMedCrossRef Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M: Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64(1):73-84.PubMedCrossRef
40.
Zurück zum Zitat Ahmed A, Wong RJ, Harrison SA: Nonalcoholic Fatty Liver Disease Review: Diagnosis, Treatment, and Outcomes. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 2015, 13(12):2062-2070.CrossRef Ahmed A, Wong RJ, Harrison SA: Nonalcoholic Fatty Liver Disease Review: Diagnosis, Treatment, and Outcomes. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 2015, 13(12):2062-2070.CrossRef
41.
Zurück zum Zitat Lewis JR, Mohanty SR: Nonalcoholic fatty liver disease: a review and update. Dig Dis Sci 2010, 55(3):560-578.PubMedCrossRef Lewis JR, Mohanty SR: Nonalcoholic fatty liver disease: a review and update. Dig Dis Sci 2010, 55(3):560-578.PubMedCrossRef
42.
Zurück zum Zitat Bastati N, Feier D, Wibmer A, Traussnigg S, Balassy C, Tamandl D, Einspieler H, Wrba F, Trauner M, Herold C et al: Noninvasive differentiation of simple steatosis and steatohepatitis by using gadoxetic acid-enhanced MR imaging in patients with nonalcoholic fatty liver disease: a proof-of-concept study. Radiology 2014, 271(3):739-747.PubMedCrossRef Bastati N, Feier D, Wibmer A, Traussnigg S, Balassy C, Tamandl D, Einspieler H, Wrba F, Trauner M, Herold C et al: Noninvasive differentiation of simple steatosis and steatohepatitis by using gadoxetic acid-enhanced MR imaging in patients with nonalcoholic fatty liver disease: a proof-of-concept study. Radiology 2014, 271(3):739-747.PubMedCrossRef
43.
Zurück zum Zitat Taylor RS, Taylor RJ, Bayliss S, Hagstrom H, Nasr P, Schattenberg JM, Ishigami M, Toyoda H, Wai-Sun Wong V, Peleg N et al: Association Between Fibrosis Stage and Outcomes of Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Gastroenterology 2020. Taylor RS, Taylor RJ, Bayliss S, Hagstrom H, Nasr P, Schattenberg JM, Ishigami M, Toyoda H, Wai-Sun Wong V, Peleg N et al: Association Between Fibrosis Stage and Outcomes of Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Gastroenterology 2020.
44.
Zurück zum Zitat Ding Y, Rao SX, Meng T, Chen C, Li R, Zeng MS: Usefulness of T1 mapping on Gd-EOB-DTPA-enhanced MR imaging in assessment of non-alcoholic fatty liver disease. European radiology 2014, 24(4):959-966.PubMedCrossRef Ding Y, Rao SX, Meng T, Chen C, Li R, Zeng MS: Usefulness of T1 mapping on Gd-EOB-DTPA-enhanced MR imaging in assessment of non-alcoholic fatty liver disease. European radiology 2014, 24(4):959-966.PubMedCrossRef
45.
Zurück zum Zitat Wu Z, Matsui O, Kitao A, Kozaka K, Koda W, Kobayashi S, Ryu Y, Minami T, Sanada J, Gabata T: Usefulness of Gd-EOB-DTPA-enhanced MR imaging in the evaluation of simple steatosis and nonalcoholic steatohepatitis. J Magn Reson Imaging 2013, 37(5):1137-1143.PubMedCrossRef Wu Z, Matsui O, Kitao A, Kozaka K, Koda W, Kobayashi S, Ryu Y, Minami T, Sanada J, Gabata T: Usefulness of Gd-EOB-DTPA-enhanced MR imaging in the evaluation of simple steatosis and nonalcoholic steatohepatitis. J Magn Reson Imaging 2013, 37(5):1137-1143.PubMedCrossRef
46.
Zurück zum Zitat Blachier M, Leleu H, Peck-Radosavljevic M, Valla DC, Roudot-Thoraval F: The burden of liver disease in Europe: a review of available epidemiological data. Journal of hepatology 2013, 58(3):593-608.PubMedCrossRef Blachier M, Leleu H, Peck-Radosavljevic M, Valla DC, Roudot-Thoraval F: The burden of liver disease in Europe: a review of available epidemiological data. Journal of hepatology 2013, 58(3):593-608.PubMedCrossRef
47.
Zurück zum Zitat Kovac JD, Dakovic M, Stanisavljevic D, Alempijevic T, Jesic R, Seferovic P, Maksimovic R: Diffusion-weighted MRI versus transient elastography in quantification of liver fibrosis in patients with chronic cholestatic liver diseases. European journal of radiology 2012, 81(10):2500-2506.PubMedCrossRef Kovac JD, Dakovic M, Stanisavljevic D, Alempijevic T, Jesic R, Seferovic P, Maksimovic R: Diffusion-weighted MRI versus transient elastography in quantification of liver fibrosis in patients with chronic cholestatic liver diseases. European journal of radiology 2012, 81(10):2500-2506.PubMedCrossRef
48.
Zurück zum Zitat Van Beers BE, Garteiser P, Leporq B, Rautou PE, Valla D: Quantitative Imaging in Diffuse Liver Diseases. Semin Liver Dis 2017, 37(3):243-258.PubMedCrossRef Van Beers BE, Garteiser P, Leporq B, Rautou PE, Valla D: Quantitative Imaging in Diffuse Liver Diseases. Semin Liver Dis 2017, 37(3):243-258.PubMedCrossRef
49.
Zurück zum Zitat Kwok R, Tse YK, Wong GL, Ha Y, Lee AU, Ngu MC, Chan HL, Wong VW: Systematic review with meta-analysis: non-invasive assessment of non-alcoholic fatty liver disease--the role of transient elastography and plasma cytokeratin-18 fragments. Aliment Pharmacol Ther 2014, 39(3):254-269.PubMedCrossRef Kwok R, Tse YK, Wong GL, Ha Y, Lee AU, Ngu MC, Chan HL, Wong VW: Systematic review with meta-analysis: non-invasive assessment of non-alcoholic fatty liver disease--the role of transient elastography and plasma cytokeratin-18 fragments. Aliment Pharmacol Ther 2014, 39(3):254-269.PubMedCrossRef
50.
Zurück zum Zitat Younossi ZM, Loomba R, Anstee QM, Rinella ME, Bugianesi E, Marchesini G, Neuschwander-Tetri BA, Serfaty L, Negro F, Caldwell SH et al: Diagnostic modalities for nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, and associated fibrosis. Hepatology 2018, 68(1):349-360.PubMedPubMedCentralCrossRef Younossi ZM, Loomba R, Anstee QM, Rinella ME, Bugianesi E, Marchesini G, Neuschwander-Tetri BA, Serfaty L, Negro F, Caldwell SH et al: Diagnostic modalities for nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, and associated fibrosis. Hepatology 2018, 68(1):349-360.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Vilar-Gomez E, Chalasani N: Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. Journal of hepatology 2018, 68(2):305-315.PubMedCrossRef Vilar-Gomez E, Chalasani N: Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. Journal of hepatology 2018, 68(2):305-315.PubMedCrossRef
52.
Zurück zum Zitat Li S, Sun X, Chen M, Ying Z, Wan Y, Pi L, Ren B, Cao Q: Liver Fibrosis Conventional and Molecular Imaging Diagnosis Update. J Liver 2019, 8(1). Li S, Sun X, Chen M, Ying Z, Wan Y, Pi L, Ren B, Cao Q: Liver Fibrosis Conventional and Molecular Imaging Diagnosis Update. J Liver 2019, 8(1).
53.
Zurück zum Zitat Yang D, Li D, Li J, Yang Z, Wang Z: Systematic review: The diagnostic efficacy of gadoxetic acid-enhanced MRI for liver fibrosis staging. European journal of radiology 2020, 125:108857.PubMedCrossRef Yang D, Li D, Li J, Yang Z, Wang Z: Systematic review: The diagnostic efficacy of gadoxetic acid-enhanced MRI for liver fibrosis staging. European journal of radiology 2020, 125:108857.PubMedCrossRef
54.
Zurück zum Zitat Feier D, Balassy C, Bastati N, Fragner R, Wrba F, Ba-Ssalamah A: The diagnostic efficacy of quantitative liver MR imaging with diffusion-weighted, SWI, and hepato-specific contrast-enhanced sequences in staging liver fibrosis--a multiparametric approach. European radiology 2016, 26(2):539-546.PubMedCrossRef Feier D, Balassy C, Bastati N, Fragner R, Wrba F, Ba-Ssalamah A: The diagnostic efficacy of quantitative liver MR imaging with diffusion-weighted, SWI, and hepato-specific contrast-enhanced sequences in staging liver fibrosis--a multiparametric approach. European radiology 2016, 26(2):539-546.PubMedCrossRef
55.
Zurück zum Zitat Watanabe H, Kanematsu M, Goshima S, Kondo H, Onozuka M, Moriyama N, Bae KT: Staging hepatic fibrosis: comparison of gadoxetate disodium-enhanced and diffusion-weighted MR imaging--preliminary observations. Radiology 2011, 259(1):142-150.PubMedCrossRef Watanabe H, Kanematsu M, Goshima S, Kondo H, Onozuka M, Moriyama N, Bae KT: Staging hepatic fibrosis: comparison of gadoxetate disodium-enhanced and diffusion-weighted MR imaging--preliminary observations. Radiology 2011, 259(1):142-150.PubMedCrossRef
56.
Zurück zum Zitat Hoffman DH, Ayoola A, Nickel D, Han F, Chandarana H, Shanbhogue KP: T1 mapping, T2 mapping and MR elastography of the liver for detection and staging of liver fibrosis. Abdom Radiol (NY) 2020, 45(3):692-700.CrossRef Hoffman DH, Ayoola A, Nickel D, Han F, Chandarana H, Shanbhogue KP: T1 mapping, T2 mapping and MR elastography of the liver for detection and staging of liver fibrosis. Abdom Radiol (NY) 2020, 45(3):692-700.CrossRef
57.
Zurück zum Zitat 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. Clinical radiology 2015, 70(10):1096-1103.PubMedCrossRef 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. Clinical radiology 2015, 70(10):1096-1103.PubMedCrossRef
58.
Zurück zum Zitat Mohamadnejad M, Tavangar SM, Sotoudeh M, Kosari F, Khosravi M, Geramizadeh B, Montazeri G, Estakhri A, Mirnasseri MM, Fazlollahi A et al: Histopathological Study of Chronic Hepatitis B: A Comparative Study of Ishak and METAVIR Scoring Systems. Int J Organ Transplant Med 2010, 1(4):171-176.PubMedPubMedCentral Mohamadnejad M, Tavangar SM, Sotoudeh M, Kosari F, Khosravi M, Geramizadeh B, Montazeri G, Estakhri A, Mirnasseri MM, Fazlollahi A et al: Histopathological Study of Chronic Hepatitis B: A Comparative Study of Ishak and METAVIR Scoring Systems. Int J Organ Transplant Med 2010, 1(4):171-176.PubMedPubMedCentral
59.
Zurück zum Zitat Kim SU, Oh HJ, Wanless IR, Lee S, Han KH, Park YN: The Laennec staging system for histological sub-classification of cirrhosis is useful for stratification of prognosis in patients with liver cirrhosis. J Hepatol 2012, 57(3):556-563.PubMedCrossRef Kim SU, Oh HJ, Wanless IR, Lee S, Han KH, Park YN: The Laennec staging system for histological sub-classification of cirrhosis is useful for stratification of prognosis in patients with liver cirrhosis. J Hepatol 2012, 57(3):556-563.PubMedCrossRef
60.
Zurück zum Zitat Theise ND, Jia J, Sun Y, Wee A, You H: Progression and regression of fibrosis in viral hepatitis in the treatment era: the Beijing classification. Mod Pathol 2018, 31(8):1191-1200.PubMedCrossRef Theise ND, Jia J, Sun Y, Wee A, You H: Progression and regression of fibrosis in viral hepatitis in the treatment era: the Beijing classification. Mod Pathol 2018, 31(8):1191-1200.PubMedCrossRef
61.
Zurück zum Zitat Garcia-Tsao G, Friedman S, Iredale J, Pinzani M: Now there are many (stages) where before there was one: In search of a pathophysiological classification of cirrhosis. Hepatology 2010, 51(4):1445-1449.PubMedPubMedCentralCrossRef Garcia-Tsao G, Friedman S, Iredale J, Pinzani M: Now there are many (stages) where before there was one: In search of a pathophysiological classification of cirrhosis. Hepatology 2010, 51(4):1445-1449.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Singh S, Fujii LL, Murad MH, Wang Z, Asrani SK, Ehman RL, Kamath PS, Talwalkar JA: Liver stiffness is associated with risk of decompensation, liver cancer, and death in patients with chronic liver diseases: a systematic review and meta-analysis. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 2013, 11(12):1573–1584 e1571–1572; quiz e1588–1579. Singh S, Fujii LL, Murad MH, Wang Z, Asrani SK, Ehman RL, Kamath PS, Talwalkar JA: Liver stiffness is associated with risk of decompensation, liver cancer, and death in patients with chronic liver diseases: a systematic review and meta-analysis. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 2013, 11(12):1573–1584 e1571–1572; quiz e1588–1579.
63.
Zurück zum Zitat Mandorfer M, Kozbial K, Schwabl P, Freissmuth C, Schwarzer R, Stern R, Chromy D, Stattermayer AF, Reiberger T, Beinhardt S et al: Sustained virologic response to interferon-free therapies ameliorates HCV-induced portal hypertension. Journal of hepatology 2016, 65(4):692-699.PubMedCrossRef Mandorfer M, Kozbial K, Schwabl P, Freissmuth C, Schwarzer R, Stern R, Chromy D, Stattermayer AF, Reiberger T, Beinhardt S et al: Sustained virologic response to interferon-free therapies ameliorates HCV-induced portal hypertension. Journal of hepatology 2016, 65(4):692-699.PubMedCrossRef
64.
Zurück zum Zitat Mandorfer M, Kozbial K, Freissmuth C, Schwabl P, Stattermayer AF, Reiberger T, Beinhardt S, Schwarzer R, Trauner M, Ferlitsch A et al: Interferon-free regimens for chronic hepatitis C overcome the effects of portal hypertension on virological responses. Aliment Pharmacol Ther 2015, 42(6):707-718.PubMedCrossRef Mandorfer M, Kozbial K, Freissmuth C, Schwabl P, Stattermayer AF, Reiberger T, Beinhardt S, Schwarzer R, Trauner M, Ferlitsch A et al: Interferon-free regimens for chronic hepatitis C overcome the effects of portal hypertension on virological responses. Aliment Pharmacol Ther 2015, 42(6):707-718.PubMedCrossRef
65.
Zurück zum Zitat Haider L, Mandorfer M, Gungoren Z, Reiberger T, Bastati N, Hodge JC, Chromy D, Trauner M, Herold C, Peck-Radosavljevic M et al: Noninvasive Monitoring of Liver Disease Regression after Hepatitis C Eradication Using Gadoxetic Acid-Enhanced MRI. Contrast Media Mol Imaging 2018, 2018:8489709.PubMedPubMedCentralCrossRef Haider L, Mandorfer M, Gungoren Z, Reiberger T, Bastati N, Hodge JC, Chromy D, Trauner M, Herold C, Peck-Radosavljevic M et al: Noninvasive Monitoring of Liver Disease Regression after Hepatitis C Eradication Using Gadoxetic Acid-Enhanced MRI. Contrast Media Mol Imaging 2018, 2018:8489709.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Truhn D, Kuhl CK, Ciritsis A, Barabasch A, Kraemer NA: A New Model for MR Evaluation of Liver Function with Gadoxetic Acid, Including Both Uptake and Excretion. European radiology 2019, 29(1):383-391.PubMedCrossRef Truhn D, Kuhl CK, Ciritsis A, Barabasch A, Kraemer NA: A New Model for MR Evaluation of Liver Function with Gadoxetic Acid, Including Both Uptake and Excretion. European radiology 2019, 29(1):383-391.PubMedCrossRef
67.
Zurück zum Zitat Tamada T, Ito K, Higaki A, Yoshida K, Kanki A, Sato T, Higashi H, Sone T: Gd-EOB-DTPA-enhanced MR imaging: evaluation of hepatic enhancement effects in normal and cirrhotic livers. European journal of radiology 2011, 80(3):e311-316.PubMedCrossRef Tamada T, Ito K, Higaki A, Yoshida K, Kanki A, Sato T, Higashi H, Sone T: Gd-EOB-DTPA-enhanced MR imaging: evaluation of hepatic enhancement effects in normal and cirrhotic livers. European journal of radiology 2011, 80(3):e311-316.PubMedCrossRef
68.
Zurück zum Zitat Tsuda N, Okada M, Murakami T: New proposal for the staging of nonalcoholic steatohepatitis: evaluation of liver fibrosis on Gd-EOB-DTPA-enhanced MRI. European journal of radiology 2010, 73(1):137-142.PubMedCrossRef Tsuda N, Okada M, Murakami T: New proposal for the staging of nonalcoholic steatohepatitis: evaluation of liver fibrosis on Gd-EOB-DTPA-enhanced MRI. European journal of radiology 2010, 73(1):137-142.PubMedCrossRef
69.
Zurück zum Zitat Kanki A, Tamada T, Higaki A, Noda Y, Tanimoto D, Sato T, Higashi H, Ito K: Hepatic parenchymal enhancement at Gd-EOB-DTPA-enhanced MR imaging: correlation with morphological grading of severity in cirrhosis and chronic hepatitis. Magn Reson Imaging 2012, 30(3):356-360.PubMedCrossRef Kanki A, Tamada T, Higaki A, Noda Y, Tanimoto D, Sato T, Higashi H, Ito K: Hepatic parenchymal enhancement at Gd-EOB-DTPA-enhanced MR imaging: correlation with morphological grading of severity in cirrhosis and chronic hepatitis. Magn Reson Imaging 2012, 30(3):356-360.PubMedCrossRef
70.
Zurück zum Zitat Besa C, Bane O, Jajamovich G, Marchione J, Taouli B: 3D T1 relaxometry pre and post gadoxetic acid injection for the assessment of liver cirrhosis and liver function. Magn Reson Imaging 2015, 33(9):1075-1082.PubMedCrossRef Besa C, Bane O, Jajamovich G, Marchione J, Taouli B: 3D T1 relaxometry pre and post gadoxetic acid injection for the assessment of liver cirrhosis and liver function. Magn Reson Imaging 2015, 33(9):1075-1082.PubMedCrossRef
71.
Zurück zum Zitat Cassinotto C, Feldis M, Vergniol J, Mouries A, Cochet H, Lapuyade B, Hocquelet A, Juanola E, Foucher J, Laurent F et al: MR relaxometry in chronic liver diseases: Comparison of T1 mapping, T2 mapping, and diffusion-weighted imaging for assessing cirrhosis diagnosis and severity. European journal of radiology 2015, 84(8):1459-1465.PubMedCrossRef Cassinotto C, Feldis M, Vergniol J, Mouries A, Cochet H, Lapuyade B, Hocquelet A, Juanola E, Foucher J, Laurent F et al: MR relaxometry in chronic liver diseases: Comparison of T1 mapping, T2 mapping, and diffusion-weighted imaging for assessing cirrhosis diagnosis and severity. European journal of radiology 2015, 84(8):1459-1465.PubMedCrossRef
72.
Zurück zum Zitat Heye T, Yang SR, Bock M, Brost S, Weigand K, Longerich T, Kauczor HU, Hosch W: MR relaxometry of the liver: significant elevation of T1 relaxation time in patients with liver cirrhosis. European radiology 2012, 22(6):1224-1232.PubMedCrossRef Heye T, Yang SR, Bock M, Brost S, Weigand K, Longerich T, Kauczor HU, Hosch W: MR relaxometry of the liver: significant elevation of T1 relaxation time in patients with liver cirrhosis. European radiology 2012, 22(6):1224-1232.PubMedCrossRef
73.
Zurück zum Zitat Sandrasegaran K, Cui E, Elkady R, Gasparis P, Borthakur G, Tann M, Liangpunsakul S: Can functional parameters from hepatobiliary phase of gadoxetate MRI predict clinical outcomes in patients with cirrhosis?European radiology 2018, 28(10):4215-4224.PubMedCrossRef Sandrasegaran K, Cui E, Elkady R, Gasparis P, Borthakur G, Tann M, Liangpunsakul S: Can functional parameters from hepatobiliary phase of gadoxetate MRI predict clinical outcomes in patients with cirrhosis?European radiology 2018, 28(10):4215-4224.PubMedCrossRef
74.
Zurück zum Zitat Asenbaum U, Kaczirek K, Ba-Ssalamah A, Ringl H, Schwarz C, Waneck F, Fitschek F, Loewe C, Nolz R: Post-hepatectomy liver failure after major hepatic surgery: not only size matters. European radiology 2018, 28(11):4748-4756.PubMedPubMedCentralCrossRef Asenbaum U, Kaczirek K, Ba-Ssalamah A, Ringl H, Schwarz C, Waneck F, Fitschek F, Loewe C, Nolz R: Post-hepatectomy liver failure after major hepatic surgery: not only size matters. European radiology 2018, 28(11):4748-4756.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Costa AF, Tremblay St-Germain A, Abdolell M, Smoot RL, Cleary S, Jhaveri KS: Can contrast-enhanced MRI with gadoxetic acid predict liver failure and other complications after major hepatic resection?Clinical radiology 2017, 72(7):598-605.PubMedCrossRef Costa AF, Tremblay St-Germain A, Abdolell M, Smoot RL, Cleary S, Jhaveri KS: Can contrast-enhanced MRI with gadoxetic acid predict liver failure and other complications after major hepatic resection?Clinical radiology 2017, 72(7):598-605.PubMedCrossRef
76.
Zurück zum Zitat Theilig D, Steffen I, Malinowski M, Stockmann M, Seehofer D, Pratschke J, Hamm B, Denecke T, Geisel D: Predicting liver failure after extended right hepatectomy following right portal vein embolization with gadoxetic acid-enhanced MRI. European radiology 2019, 29(11):5861-5872.PubMedCrossRef Theilig D, Steffen I, Malinowski M, Stockmann M, Seehofer D, Pratschke J, Hamm B, Denecke T, Geisel D: Predicting liver failure after extended right hepatectomy following right portal vein embolization with gadoxetic acid-enhanced MRI. European radiology 2019, 29(11):5861-5872.PubMedCrossRef
77.
Zurück zum Zitat O'Rourke TR, Welsh FK, Tekkis PP, Lyle N, Mustajab A, John TG, Peppercorn D, Rees M: Accuracy of liver-specific magnetic resonance imaging as a predictor of chemotherapy-associated hepatic cellular injury prior to liver resection. Eur J Surg Oncol 2009, 35(10):1085-1091.PubMedCrossRef O'Rourke TR, Welsh FK, Tekkis PP, Lyle N, Mustajab A, John TG, Peppercorn D, Rees M: Accuracy of liver-specific magnetic resonance imaging as a predictor of chemotherapy-associated hepatic cellular injury prior to liver resection. Eur J Surg Oncol 2009, 35(10):1085-1091.PubMedCrossRef
78.
Zurück zum Zitat Shindoh J, Tzeng CW, Aloia TA, Curley SA, Zimmitti G, Wei SH, Huang SY, Mahvash A, Gupta S, Wallace MJ et al: Optimal future liver remnant in patients treated with extensive preoperative chemotherapy for colorectal liver metastases. Ann Surg Oncol 2013, 20(8):2493-2500.PubMedCrossRef Shindoh J, Tzeng CW, Aloia TA, Curley SA, Zimmitti G, Wei SH, Huang SY, Mahvash A, Gupta S, Wallace MJ et al: Optimal future liver remnant in patients treated with extensive preoperative chemotherapy for colorectal liver metastases. Ann Surg Oncol 2013, 20(8):2493-2500.PubMedCrossRef
79.
Zurück zum Zitat Cho SH, Kang UR, Kim JD, Han YS, Choi DL: The value of gadoxetate disodium-enhanced MR imaging for predicting posthepatectomy liver failure after major hepatic resection: a preliminary study. European journal of radiology 2011, 80(2):e195-200.PubMedCrossRef Cho SH, Kang UR, Kim JD, Han YS, Choi DL: The value of gadoxetate disodium-enhanced MR imaging for predicting posthepatectomy liver failure after major hepatic resection: a preliminary study. European journal of radiology 2011, 80(2):e195-200.PubMedCrossRef
80.
Zurück zum Zitat Boraschi P, Della Pina MC, Donati F: Graft complications following orthotopic liver transplantation: Role of non-invasive cross-sectional imaging techniques. European journal of radiology 2016, 85(7):1271-1283.PubMedCrossRef Boraschi P, Della Pina MC, Donati F: Graft complications following orthotopic liver transplantation: Role of non-invasive cross-sectional imaging techniques. European journal of radiology 2016, 85(7):1271-1283.PubMedCrossRef
81.
Zurück zum Zitat Nolz R, Asenbaum U, Schoder M, Wibmer A, Einspieler H, Prusa AM, Peck-Radosavljevic M, Ba-Ssalamah A: Diagnostic workup of primary sclerosing cholangitis: the benefit of adding gadoxetic acid-enhanced T1-weighted magnetic resonance cholangiography to conventional T2-weighted magnetic resonance cholangiography. Clinical radiology 2014, 69(5):499-508.PubMedCrossRef Nolz R, Asenbaum U, Schoder M, Wibmer A, Einspieler H, Prusa AM, Peck-Radosavljevic M, Ba-Ssalamah A: Diagnostic workup of primary sclerosing cholangitis: the benefit of adding gadoxetic acid-enhanced T1-weighted magnetic resonance cholangiography to conventional T2-weighted magnetic resonance cholangiography. Clinical radiology 2014, 69(5):499-508.PubMedCrossRef
82.
Zurück zum Zitat Pandey P, Pandey A, Dillhoff M, Schmidt C, Kamel IR, Pawlik TM: Assessing the Non-tumorous Liver: Implications for Patient Management and Surgical Therapy. J Gastrointest Surg 2018, 22(2):344-360.PubMedCrossRef Pandey P, Pandey A, Dillhoff M, Schmidt C, Kamel IR, Pawlik TM: Assessing the Non-tumorous Liver: Implications for Patient Management and Surgical Therapy. J Gastrointest Surg 2018, 22(2):344-360.PubMedCrossRef
83.
Zurück zum Zitat Davenport MS, Viglianti BL, Al-Hawary MM, Caoili EM, Kaza RK, Liu PS, Maturen KE, Chenevert TL, Hussain HK: Comparison of acute transient dyspnea after intravenous administration of gadoxetate disodium and gadobenate dimeglumine: effect on arterial phase image quality. Radiology 2013, 266(2):452-461.PubMedCrossRef Davenport MS, Viglianti BL, Al-Hawary MM, Caoili EM, Kaza RK, Liu PS, Maturen KE, Chenevert TL, Hussain HK: Comparison of acute transient dyspnea after intravenous administration of gadoxetate disodium and gadobenate dimeglumine: effect on arterial phase image quality. Radiology 2013, 266(2):452-461.PubMedCrossRef
84.
Zurück zum Zitat Ringe KI, von Falck C, Raatschen HJ, Wacker F, Hinrichs J: Evaluation of transient respiratory motion artifact at gadoxetate disodium-enhanced MRI-Influence of different contrast agent application protocols. PLoS One 2018, 13(7):e0200887.PubMedPubMedCentralCrossRef Ringe KI, von Falck C, Raatschen HJ, Wacker F, Hinrichs J: Evaluation of transient respiratory motion artifact at gadoxetate disodium-enhanced MRI-Influence of different contrast agent application protocols. PLoS One 2018, 13(7):e0200887.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Polanec SH, Bickel H, Baltzer PAT, Thurner P, Gittler F, Hodge JC, Bashir MR, Ba-Ssalamah A: Respiratory motion artifacts during arterial phase imaging with gadoxetic acid: Can the injection protocol minimize this drawback?J Magn Reson Imaging 2017, 46(4):1107-1114.PubMedCrossRef Polanec SH, Bickel H, Baltzer PAT, Thurner P, Gittler F, Hodge JC, Bashir MR, Ba-Ssalamah A: Respiratory motion artifacts during arterial phase imaging with gadoxetic acid: Can the injection protocol minimize this drawback?J Magn Reson Imaging 2017, 46(4):1107-1114.PubMedCrossRef
Metadaten
Titel
Quantification of liver function using gadoxetic acid-enhanced MRI
verfasst von
Sarah Poetter-Lang
Nina Bastati
Alina Messner
Antonia Kristic
Alexander Herold
Jacqueline C. Hodge
Ahmed Ba-Ssalamah
Publikationsdatum
09.10.2020
Verlag
Springer US
Erschienen in
Abdominal Radiology / Ausgabe 11/2020
Print ISSN: 2366-004X
Elektronische ISSN: 2366-0058
DOI
https://doi.org/10.1007/s00261-020-02779-x

Weitere Artikel der Ausgabe 11/2020

Abdominal Radiology 11/2020 Zur Ausgabe

Update Radiologie

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