Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2020 Jul 2;40(9):2050–2063. doi: 10.1111/liv.14555

Radiomics in liver diseases: Current progress and future opportunities

Jingwei Wei 1,2, Hanyu Jiang 3, Dongsheng Gu 1,2, Meng Niu 4, Fangfang Fu 5,6, Yuqi Han 1,2, Bin Song 3, Jie Tian 1,2,7,8,
PMCID: PMC7496410  PMID: 32515148

Abstract

Liver diseases, a wide spectrum of pathologies from inflammation to neoplasm, have become an increasingly significant health problem worldwide. Noninvasive imaging plays a critical role in the clinical workflow of liver diseases, but conventional imaging assessment may provide limited information. Accurate detection, characterization and monitoring remain challenging. With progress in quantitative imaging analysis techniques, radiomics emerged as an efficient tool that shows promise to aid in personalized diagnosis and treatment decision‐making. Radiomics could reflect the heterogeneity of liver lesions via extracting high‐throughput and high‐dimensional features from multi‐modality imaging. Machine learning algorithms are then used to construct clinical target‐oriented imaging biomarkers to assist disease management. Here, we review the methodological process in liver disease radiomics studies in a stepwise fashion from data acquisition and curation, region of interest segmentation, liver‐specific feature extraction, to task‐oriented modelling. Furthermore, the applications of radiomics in liver diseases are outlined in aspects of diagnosis and staging, evaluation of liver tumour biological behaviours, and prognosis according to different disease type. Finally, we discuss the current limitations of radiomics in liver disease studies and explore its future opportunities.

Keywords: data science, liver diseases, machine learning, precision medicine, radiologic technology


Abbreviations

AFP

α‐fetoprotein

ALB

serum albumin

ALT

serum alanine aminotransferase

AST

aspartate aminotransferase

AUC

area under the curve

CA 19‐9

carbohydrate antigen 19‐9

CB

conjugated bilirubin

CNN

convolution neural network

CT

computed tomography

DL

deep learning

HBsAg

hepatitis B virus surface antigen

HCC

hepatocellular carcinoma

ICC

intrahepatic cholangiocarcinoma

MRI

magnetic resonance imaging

NASH

nonalcoholic steatohepatitis

PD‐1

anti‐programmed cell death protein

PD‐L1

anti‐programmed cell death ligand 1

PIVKA‐II

prothrombin induced by vitamin K absence‐II

PLT

platelet count

PT

prothrombin time

ROI

region of interest

SWE

shear wave elastography

TACE

transcatheter arterial chemoembolization

TB

Serum total bilirubin

Key points.

  • Radiomics as an emerging technique based on medical imaging analysis is more commonly used in liver disease studies.

  • Inter‐personal heterogeneity could be revealed via extracting high‐dimensional quantitative imaging features and analysed by artificial intelligence algorithms.

  • Radiomics can be applied in the diagnosis, treatment effect evaluation and prognosis prediction in liver diseases.

1. INTRODUCTION

Liver diseases, a wide spectrum of pathologies from inflammation to neoplasm, have become a major health problem worldwide. Noninvasive imaging plays a critical role in the characterization and monitoring of liver diseases. Conventional ultrasound, computed tomography (CT) and magnetic resonance imaging (MRI) are widely used for qualitative evaluation of liver morphology and blood supply. 1 , 2 , 3 Tremendous progress is still being made in liver imaging with introduction of advanced techniques, including metabolic imaging, molecular imaging, and multi‐parametric functional MRI, etc, allowing improved evaluation of liver diseases and assisting personalized medical decision making. 4 , 5 , 6

With accumulation of scalable liver imaging data, radiomics emerges as a novel radiological technique that comprehensively utilizes large‐scale medical imaging into the process of liver disease management via artificial intelligence techniques. 7 , 8 It enables extraction of high‐throughput quantitative imaging features beyond inspections of naked human eyes and converting encrypted medical imaging into minable numerical data. 8 Combined with clinical, pathological, or genetic information, radiomics would assist in lesion characterization, preoperative diagnosis, treatment efficacy evaluation, as well as prognosis prediction in various clinical settings. 9 , 10 , 11

Quantitative imaging traits were proved to be associated with global gene expression programmes, and could reconstruct 78% of the global gene expression profiles in liver cancer. 12 This groundbreaking result laid a foundation and greatly encouraged researchers to explore the potential of quantitative imaging tool in preoperative genetic/pathological outcome prediction. Hence, a great deal of radiomics studies have been conducted using multi‐parametric and multi‐modality imaging in terms of liver disease diagnosis and treatment decision making. 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 In certain scenarios, this artificial intelligence‐based technique could even compete pathological gold standard, providing new ways for unsolved clinical problems in the paradigm of liver disease management. 16 Nevertheless, it still requires further multi‐centre and prospective validation for the validity of radiomics. The interpretability and the correlation with biological/pathological underpinnings also represent substantial obstacles for the translation of artificial intelligence into real clinical practice.

Here, we review the basic concepts of radiomics methodologies specific for liver studies from data acquisition, liver/lesion segmentation, feature design, to model construction (Figure 1). Meanwhile, representative clinical applications of radiomics in liver diseases regarding diagnosis, staging, evaluation of liver tumour biological behaviours, and prognosis are also within the scope of this study. Finally, we summarize the current challenges and limitation of radiomics, and explore its future directions in liver diseases.

FIGURE 1.

FIGURE 1

Workflow of radiomics methodological process

2. METHODOLOGY OF RADIOMICS IN LIVER DISEASES

2.1. Data acquisition and curation

Data used in radiomics studies can be single‐centre or multi‐centre, and retrospective or prospective. Here, we searched PubMed (8 October 2019) for radiomics studies on liver diseases using terms (liver diseases AND radiomics), and found 36 clinical target‐oriented published work. 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 Most (33 out of 36) studies were performed on single‐centre with retrospective cohort, while only two studies were performed on multi‐centre and prospective cohort (Table 1). And the most commonly used imaging modality was CT (18 studies), followed by MRI (12 studies), positron emission tomography (PET) (two studies) and ultrasonography (US) (four studies) (Table 1).

TABLE 1.

Summary of published radiomics studies on liver diseases

Number Reference Study design (retrospective/prospective, single or multi‐centre study) No. of patients No. and type of radiomic features Statistical analysis (feature selection and modelling) Imaging Modality Clinical Characteristics
1 Zhou et al 13 Retrospective, single‐centre study 215 300 (histogram and GLCM) LASSO CT Prediction of early recurrence in HCC
2 Cozzi et al 14 Retrospective, single‐centre study 138 35 (histogram and texture) Cox CT Predict local control and survival of HCC
3 Naganawa et al 15 Retrospective, single‐centre study 88 6 (histogram) Logistic CT Prediction of nonalcoholic steatohepatitis
4 Wang et al 16 Prospective, multi‐centre study 398 Deep learning features DLRE Ultrasound Assessing liver fibrosis
5 Peng et al 17 Retrospective, single‐centre study 304 980 (histogram, shape and texture) LASSO CT Prediction of microvascular invasion
6 Reimer et al 18 Retrospective, single‐centre study 37 6 (histogram) Logistic MRI Assessment of Therapy Response to TACE
7 Akai et al 19 Retrospective, single‐centre study 127 96 (histogram) RSF CT Predicting prognosis of resected HCC
8 Li et al 20 Retrospective, single‐centre study 144 472 (radiomics, ORF and CEMF features) RF, SVM, DT, NN, Logistic Ultrasound Assessing liver fibrosis
9 Hui et al 21 Retrospective, single‐centre study 50 290 1‐nearest neighbor MRI Prediction of early recurrence in HCC
10 Kim et al 22 Retrospective, single‐centre study 88 116 LASSO, COX CT Predicting survival after TACE
11 Liu et al 23 Prospective, multi‐centre study 385 20 648 (non‐texture and texture) LASSO CT Noninvasively detect CSPH in cirrhosis
12 Wu et al 24 Retrospective, single‐centre study 170 328 (non‐texture and texture) LASSO MRI Predicting the grade of HCC
13 Yao et al 25 Retrospective, single‐centre study 177 Deep learning features KSVD + SRT+SVM Ultrasound Preoperative diagnosis
14 Hu et al 26 Retrospective, single‐centre study 482 1044 histogram and texture LASSO Ultrasound Prediction of microvascular invasion
15 Klaassen et al 27 Retrospective, single‐centre study 69 370 (histogram, shape, texture) Random forest CT Prediction of esophagogastric Cancer Liver Metastasis
16 Zheng et al 28 Retrospective, single‐centre study 319 110 texture features LASSO CT Preoperative Prediction of survival
17 Park et al 29 Retrospective, single‐centre study 436 8 histogram and 35 textural features logistic regression with elastic net regularization MRI Preoperative prediction of staging liver fibrosis
18 Chen et al 30 Retrospective, single‐centre study 207 1044 radiomic features Extremely randomized tree MRI Preoperative prediction of immunoscore
19 Feng et al 31 Retrospective, single‐centre study 160 1044 radiomic features Lasso MRI Preoperative prediction of microvascular invasion
20 Ma et al 32 Retrospective, single‐centre study 157 647 (histogram, shape, texture, wavelet) SVM CT Prediction of microvascular invasion
21 Shan et al 33 Retrospective, single‐centre study 156 1044 (histogram, wavelet, texture) LASSO CT Prediction of early recurrence in HCC
22 Cai et al 34 Retrospective, single‐centre study 125 713 (intensity, texture, wavelet, shape and size) LASSO, Logistic CT Prediction of Posthepatectomy Liver Failure in HCC
23 Wu et al 35 Retrospective, single‐centre study 369 1029 (first‐order, shape, texture, high‐order) Variance threshold, LASSO, Decision tree, Random forest, K nearest neighbors, Logistic MR Prediction of hepatocellular carcinoma and hepatic haemangioma
24 Xu et al 36 Retrospective, single‐centre study 495 7260 radiomic features Multivariable logistic regression CT Prediction of microvascular invasion
25 Rahmim et al 37 Retrospective, single‐centre study 52 41 (histogram) Univariate and multivariate PET Prognostic model for colorectal Liver Metastasis
26 Yuan et al 38 Retrospective, single‐centre study 184 647 (intensity, texture, wavelet, shape and size) MRMR, LASSO, Cox CT Prediction of early recurrence in HCC
27 Zhang et al 39 Retrospective, single‐centre study 155 385 (histogram, texture) LASSO MR Prediction of early recurrence in HCC
28 Zhao et al 40 Retrospective, single‐centre study 47 396 (histogram, texture, Haralick, morphological) Wilcoxon signed‐rank test, Logistic MR Prediction of early recurrence in intrahepatic cholangiocarcinoma
29 Guo et al 41 Retrospective, single‐centre study 133 853 radiomic features Lasso CT Prediction of recurrence in hcc after liver transplantation
30 Tseng et al 42 Retrospective, single‐centre study 169 1474 radiomic features LASSO CT Prediction of portal pressure and patient outcome in hypertension
31 Hectors et al 43 Retrospective, single‐centre study 48 218 radiomic features Binary logistic regression analysis MRI Prediction of immune‐oncological characteristics
32 Ni et al 44 Retrospective, single‐centre study 206 1044 textural features LASSO + BPNet CT Prediction of microvascular invasion
33 Liao et al 45 Retrospective, single‐centre study 142 57 radiomic features linear elastic‐net model PET Evaluation of Tumour‐Infiltrating CD8 + T Cells
34 Huang et al 46 Retrospective, single‐centre study 100 First order statistical, shape, textural, and higher order statistical features LASSO MRI Diagnosis of dual‐phenotype HCC
35 Shur et al 47 Retrospective, single‐centre study 102 114 radiomic features Multivariate cox propotional hazard modelling CT Improved prognostication of surgical candidates with colorectal liver metastasis
36 Jiang et al 48 Prospective, single‐centre 211 396 radiomic features LASSO MRI Diagnosis of HCC

Considering the effect of inconsistent imaging acquisition protocol and reconstruction procedure in multi‐centres via multi brand manufactories, preprocessing of the collected imaging data is required. Currently, the most commonly used methods conclude resampling and intensity normalization. Image resampling is used to improve image quality and eliminate bias introduced by non‐uniform imaging resolution. 49 , 50 Image intensity normalization is utilized to correct inter‐subject intensity variation by transforming all images from original greyscale into a standard greyscale. 51 , 52 Park et al normalized liver signal intensity according to the spleen signal on hepatobiliary phase (HBP) images to extract high‐order textural features and revealed the improved diagnostic value as compared with non‐normalized data. 29

In addition to imaging data, clinical factors were also involved in radiomics analysis, including patient age, gender, Child‐Pugh stage, histologic grading, BCLC stage, cirrhosis and its cause, etc. 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 Laboratory examination indexes comprise serum α‐fetoprotein (AFP) level, prothrombin induced by vitamin K absence‐II (PIVKA‐II) level, carbohydrate antigen 19‐9 (CA 19‐9) level, hepatitis B virus surface antigen (HBsAg), serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), serum total bilirubin (TB), conjugated bilirubin (CB), serum albumin (ALB), prothrombin time (PT), platelet count (PLT), etc. 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48

2.2. Region of interest segmentation

Segmentation of region of interest (ROI) could be divided into manual segmentation and semiautomatic/automatic segmentation. Most radiomics studies on liver disease applied manual segmentation. Only six studies performed semiautomatic/automatic segmentation. 17 , 30 , 39 , 46 , 53 , 54

Manual segmentation is performed by radiologists to annotate the location and precise boundary of the lesion. Another way of manual segmentation is realized by placing a rectangular/circle box via deep learning analysis. Wang et al conducted a squared ROI segmentation as the input of convolution neural network (CNN) and achieved satisfying performance in liver fibrosis stage prediction. 16 Naganawa et al applied similar segmentation approach with a 2‐cm diameter circular ROI covering the lesion while excluding intrahepatic vessels. 15 Considering the discrepancy of subjective judgement in manual segmentation, segmentations by multi‐clinicians, of multi‐time point, and using computer perturbation are required to decrease the intra‐ and inter‐reader variability. 32 Feature reproducibility and robustness are generally evaluated through calculation of intra‐class correlation coefficient and concordance correlation coefficient. 36 , 56 , 57

Automatic segmentation aims to annotate ROIs by computer automatically, whereas semiautomatic segmentation still needs partial manual intervention to mark the centre of the lesion before automatic segmentation. Several classic segmentation algorithms showed good performance in liver lesion annotation. 58 , 59 , 60 , 61 These methods can be generally divided into three categories: (a) algorithms based on intensity thresholds and region (global thresholding, local thresholding, region growing, and region splitting and merging methods), (b) algorithms based on statistical approach (statistical parametric mapping and maximization segmentation algorithm), clustering (k‐means clustering and fuzzy clustering) and deformable model approach (Snake model and geometric active contour model), (c) algorithms incorporating empirical knowledge into the segmentation process (Atlas Guided Approach and Artificial Neural Network).

2.3. Feature extraction

Radiomic features are divided into manual engineered features and deep learning (DL) features. Manual engineered features include shape/histogram/texture‐based features. Shape‐based features describe the geometric attributes of the ROIs. Histogram features capture the first‐order statistic characteristics of liver parenchyma or liver lesion. Textural features, extracted from a series of high‐order textural matrixes, describe the granular textural pattern of the ROIs. In addition, filtered features are extracted from ROI preprocessed by wavelet, Laplacian and Gaussian filters from multiple dimensions. 62 Commonly used manual engineered features are shown in Table 2. Another type of engineered features is defined as empirical features or semantic features that are designed by experience and knowledge of radiologists. Fu et al designed “peer‐off” features with hypothesis that tumour grows from inside to outside. 63 By splitting the tumour into 10 peel‐off layers and extracting corresponding statistical features and its ratio, it can reflect tumour growth pattern and spatial heterogeneity. They found the feature ‐ POF_entropy showed satisfactory value for predicting the progress‐free survival following liver resection and transarterial chemoembolization. This feature exactly represented the texture randomness or irregularity of the innermost layer.

TABLE 2.

Radiomic features used in radiomics studies on liver diseases

Shape‐based 3D features

(n = 16)

Shape‐based 2D features

(n = 16)

Histogram features

(n = 19)

Textural features (n = 75)

Gray Level Co‐occurrence Matrix (GLCM) Features

(n = 24)

Gray Level Run Length Matrix (GLRLM) Features

(n = 16)

Gray Level Size Zone Matrix (GLSZM) Features

(n = 16)

Neighbouring Gray Tone Difference Matrix (NGTDM) Features

(n = 5)

Gray Level Dependence Matrix (GLDM) Features

(n = 14)

1 Mesh Volume Mesh Surface Energy Autocorrelation Short Run Emphasis (SRE) Small Area Emphasis (SAE) Coarseness Small Dependence Emphasis (SDE)
2 Voxel Volume Pixel Surface Total Energy Joint Average Long Run Emphasis (LRE) Large Area Emphasis (LAE) Contrast Large Dependence Emphasis (LDE)
3 Surface Area Perimeter Entropy Cluster Prominence Gray Level Non‐Uniformity (GLN) Gray Level Non‐Uniformity (GLN) Busyness Gray Level Non‐Uniformity (GLN)
4 Surface Area to Volume ratio Perimeter to Surface ratio Minimum Cluster Shade Gray Level Non‐Uniformity Normalized (GLNN) Gray Level Non‐Uniformity Normalized (GLNN) Complexity Dependence Non‐Uniformity (DN)
5 Sphericity Sphericity 10th percentile Cluster Tendency Run Length Non‐Uniformity (RLN) Size‐Zone Non‐Uniformity (SZN) Strength Dependence Non‐Uniformity Normalized (DNN)
6 Compactness Spherical Disproportion 90th percentile Contrast Run Length Non‐Uniformity Normalized (RLNN) Size‐Zone Non‐Uniformity Normalized (SZNN) Gray Level Variance (GLV)
7 Spherical Disproportion Maximum 2D diameter Maximum Correlation Run Percentage (RP) Zone Percentage (ZP) Dependence Variance (DV)
8 Maximum 3D diameter Major Axis Length Mean Difference Average Gray Level Variance (GLV) Gray Level Variance (GLV) Dependence Entropy (DE)
9 Maximum 2D diameter (Slice) Minor Axis Length Median Difference Entropy Run Variance (RV) Zone Variance (ZV) Low Gray Level Emphasis (LGLE)
10 Maximum 2D diameter (Column) Elongation Interquartile Range Difference Variance Run Entropy (RE) Zone Entropy (ZE) High Gray Level Emphasis (HGLE)
11 Maximum 2D diameter (Row) Range Joint Energy Low Gray Level Run Emphasis (LGLRE) Low Gray Level Zone Emphasis (LGLZE) Small Dependence Low Gray Level Emphasis (SDLGLE)
12 Major Axis Length Mean Absolute Deviation (MAD) Joint Entropy High Gray Level Run Emphasis (HGLRE) High Gray Level Zone Emphasis (HGLZE) Small Dependence High Gray Level Emphasis (SDHGLE)
13 Minor Axis Length Robust Mean Absolute Deviation (rMAD) Informational Measure of Correlation (IMC) 1 Short Run Low Gray Level Emphasis (SRLGLE) Small Area Low Gray Level Emphasis (SALGLE) Large Dependence Low Gray Level Emphasis (LDLGLE)
14 Least Axis Length Root Mean Squared (RMS) Informational Measure of Correlation (IMC) 2 Short Run High Gray Level Emphasis (SRHGLE) Small Area High Gray Level Emphasis (SAHGLE) Large Dependence High Gray Level Emphasis (LDHGLE)
15 Elongation Standard Deviation Inverse Difference Moment (IDM) Long Run Low Gray Level Emphasis (LRLGLE) Large Area Low Gray Level Emphasis (LALGLE)
16 Flatness Skewness Maximal Correlation Coefficient (MCC) Long Run High Gray Level Emphasis (LRHGLE) Large Area High Gray Level Emphasis (LAHGLE)
17 Kurtosis Inverse Difference Moment Normalized (IDMN)
18 Variance Inverse Difference (ID)
19 Uniformity Inverse Difference Normalized (IDN)
20 Inverse Variance
21 Maximum Probability
22 Sum Average
23 Sum Entropy
24 Sum of Squares

Filtered features extracted from images preprocessed by wavelet filter, Laplacian of Gaussian filter, etc, including the shape/histogram/texture‐based radiomic features. 63

Compared with manual engineered features, DL network could extract supplementary high‐dimensional features that are hard to depict by observers. 55 , 64 , 65 , 66 The DL network encodes medical image into shape information and abstract textural information via shallow and deep layers respectively. Wang et al proposed a novel method to automatically extract DL features from MR imaging using CNN. 64 They found that DL features outperformed textural features in predicting the malignancy of HCC. Chaudhary et al used unsupervised auto‐encoder framework to extract DL features. 66 Features extracted from the bottleneck layer showed predictive ability for the survival risk of liver cancer.

2.4. Task‐oriented modelling

Generally, the methods for feature selection conclude filter‐based, wrapper‐based, and model‐embedded methods. 67 Filter‐based methods produce a selected feature set according to the correlation between features and the classifying labels. Commonly used filter‐based methods include calculation of mutual information, correlation coefficient and uni‐variable analysis (ie Mann‐Whitney U test and Chi‐squared test), etc. 68 , 69 , 70 Wrapper‐based methods take into account the weighing of feature subsets, and are combined with an appointed classifier. It selects features that could improve the accuracy of the prediction to the maximum extend and removes the features that contribute less to the prediction until the specified feature number is reached. Model‐embedded methods perform feature selection in the process of model construction. An example of this method is the least absolute shrinkage and selection operator (LASSO) algorithm. 71 LASSO aims to minimize the residual sum of squares, subjected to the sum of the absolute value of the coefficients being less than a tuning parameter. It forces specified coefficients to zero and thus effectively produce a simpler model. Among the aforementioned methods, filter‐based methods require less computation time than the other two methods but with lower prediction accuracy. Thus, they are most commonly used as a primary selection method to initially reduce features. 23 , 55

Regarding modelling strategy, radiomics studies on liver disease mostly utilized supervised learning modelling. LASSO logistic regressing modelling was commonly used, demonstrating satisfying performance particularly in small sample size based studies. 22 , 31 , 72 Support vector machine and random forest were also used in published liver disease radiomics studies. 19 , 23 , 27 , 32 Notably, Li et al compared six types of machine‐learning algorithms in predicting liver fibrosis, including adaptive boosting, decision tree, logistic regression, neural network, random forest and support vector machine. 20 Their result indicated that adaptive boosting, random forest and support vector machine stood out as superior modelling methods with improved accuracy for fibrosis prediction.

3. RADIOMICS IN THE DIAGNOSIS AND STAGING OF LIVER DISEASES

For clinical application, radiomics plays a pivotal role in the diagnosis, staging and grading of several liver diseases, of which most efforts focused on hepatic malignancies and liver diffuse diseases (Figure 2).

FIGURE 2.

FIGURE 2

Illustration of clinical application of radiomics on liver diseases

3.1. Hepatic malignancies

Hepatocellular carcinoma (HCC) is currently the most common primary liver cancer. 73 However, many non‐HCC malignancies (eg small duct type intrahepatic cholangiocarcinoma [ICC] and combined hepatocellular‐cholangiocarcinoma) and other atypical benign focal liver lesions (eg haemangioma and hepatic adenoma) can mimic HCC, making the diagnosis challenging via current imaging techniques. 74 , 75

Radiomics demonstrated great potential in differentiating focal liver lesions. 25 , 76 , 77 Li et al primarily investigated texture features of focal hepatic lesions on spectral attenuated inversion‐recovery T2 weighted MRI, and found that the radiomics signatures can help classify hepatic haemangioma, hepatic metastases and HCC with satisfying diagnostic performances (area under the curve [AUC]: 0.83‐0.91). 76 Trivizakis et al reported that the three‐dimensional convolutional neural network features on diffusion‐weighted MR images achieved an accuracy of 83% for discriminating primary and metastatic liver tumours. 77 In addition to MR imaging, radiomics analysis on multi‐modal ultrasound images also demonstrated diagnostic ability for benign and malignant focal liver lesion classification (AUC: 0.94, 95%CI: 0.88‐0.98) and malignant subtyping (AUC: 0.97, 95%CI: 0.93‐0.99). 25

3.2. Liver diffuse diseases

Besides hepatic malignancies, radiomics also showed potential in characterization of liver diffuse diseases including fatty liver diseases and liver fibrosis. The first study evaluating the performance of CT‐based texture features for predicting nonalcoholic steatohepatitis (NASH) was conducted by Naganawa et al, which included 88 retrospective suspected NASH patients. 15 They reported that the AUC reached up to 0.94 in patients without suspected fibrosis, but dropped significantly in patients with suspicion of fibrosis (AUC: 0.60). Tang et al further explored the relationship between a quantitative ultrasound‐based machine learning model and histopathology scoring in a rat model. 78 Their results demonstrated that combining quantitative ultrasound parameters with conventional shear wave elastography significantly improved the classification accuracy of steatohepatitis, liver steatosis, inflammation and fibrosis.

Other than fatty liver diseases, more studies focused on liver fibrosis staging and associated complications. A prospective multi‐centre study by Wang et al revealed that DL radiomics of shear wave elastography (SWE) significantly improved the accuracy of liver fibrosis staging, with AUCs of 0.97, 0.98 and 0.85 for cirrhosis (F4), advanced fibrosis (≥F3) and significant fibrosis (≥F2) respectively. 16 Similar results have been reported by another prospective study, in which the machine‐learning‐based multi‐parametric ultrasomics model achieved remarkably improved power for significant fibrosis (≥F2). 20

CT‐based radiomics was also utilized for noninvasive assessment of liver fibrosis. Choi et al retrospectively developed a DL system on portal venous phase CT images in 7461 patients and validated it in an independent data sets comprising 891 patients. 79 The accuracy was of 79.4% in the validation sets, with AUC of 0.96, 0.97 and 0.95 for ≥ F2, ≥F3 and F4 respectively. Regarding portal hypertension, Liu et al reported in their multi‐centre prospective study that the radiomics signature on portal venous phase CT images accurately detected portal hypertension with the C‐index of 0.889, 0.800, 0.917 and 0.827 in four external validation cohorts respectively. 23

4. RADIOMICS IN THE EVALUATION OF LIVER TUMOUR BIOLOGICAL BEHAVIOURS AND PROGNOSIS

Beyond diagnosis and staging, radiomics enables quantitative assessment of liver tumour biological behaviours, as well as prediction of prognosis and antitumoral treatment effect (Figure 2).

4.1. HCC

4.1.1. Measurement of tumour differentiation and proliferation

Histologic grade was one of the most important risk factors for postoperative recurrence in HCC. 80 , 81 , 82 , 83 Recently, two MRI‐based studies investigated radiomic features for HCC aggressiveness characterization, demonstrating the potential of radiomics as indicative biomarkers for HCC grade. 24 , 84 Regarding Ki‐67 level, Ye et al reported that radiomics analysis can evaluate the tumour Ki‐67 level preoperatively with good accuracy (C‐index: 0.936) in a prospective study. 85

4.1.2. Assessment of tumour vascular invasion

Preoperative discrimination between neoplastic and bland portal vein thrombosis and detection of microvascular invasion in HCC is critically important. 86 , 87 Canellas et al explored the role of CT texture features for differentiating neoplastic and bland portal vein thrombosis. They found that mean value of positive pixels and entropy can characterize portal vein thrombosis. 88 Recent studies have shown promising results of CT and ultrasound‐based radiomics signatures for preoperative microvascular invasion prediction, all with high diagnostic accuracy. 17 , 89

4.1.3. Prediction of treatment efficacy and prognosis

Radiomics analysis permits accurate prediction of prognosis and effective diverse therapy evaluation. 73 , 90 Several studies were conducted for hepatic resection evaluation, and one study was for liver transplantation evaluation. 13 , 19 , 21 , 28 , 91 , 92 , 93 Furthermore, Li et al found that texture analysis of CT images can be helpful not only in prognosis prediction, but also in treatment selection between liver resection and transcatheter arterial chemoembolization (TACE). 81 For HCC patients with prominent vascular invasion and/or extrahepatic spread (BCLC stage C), systematic treatment is the standard of care recommended by current guidelines from different geographical regions. 36 , 90 Mulé et al retrospectively investigated 92 advanced HCC patients from two centres and reported that the contrast‐enhanced CT texture feature entropy was correlated with tumour heterogeneity by manual visualization, and entropy on portal venous phase images was an independent predictor for OS. 94

Radiomics analysis also yielded promising results in predicting response for patients treated with immunotherapies. Sun et al retrospectively generated a contrast‐enhanced CT‐based radiomics signature of tumour‐infiltrating CD8 cells and investigated its performances in predicting tumour immune phenotype (immune‐inflamed vs immune‐desert) and response to anti‐programmed cell death protein (PD)‐1 or anti‐programmed cell death ligand 1 (PD‐L1) monotherapies. 95 Another study by Chen et al explored the capacity of radiomics analysis on gadoxetic acid‐enhanced MR imaging in predicting immunoscore, a new prognostic biomarker for immunotherapy revealing tumour infiltrating lymphocytes density. 96

4.2. ICC

ICC is an aggressive primary hepatic cancer arising from the bile duct epithelium. 97 However, unlike HCC, surgical resection is currently the only curative treatment for ICC patients. 98 A recent single‐centre retrospective study reported that the radiomics signature on preoperative arterial‐phase contrast‐enhanced MR images can be used to predict early recurrence of ICC after partial hepatectomy with the AUC of 0.82 and 0.77 in the training and validation cohort respectively. 55 Ji et al constructed a radiomics signature from portal venous CT to predict lymph node metastasis in biliary tract caners. 99 They found good discrimination of the signature in both training (AUC: 0.81) and validation cohort (AUC: 0.80). 99

4.3. Metastatic hepatic malignancies

In addition to primary liver cancers, radiomics also showed promise in the evaluation of several metastatic hepatic malignancies. Lubner et al found that pretreatment portal venous phase CT texture features of the colorectal liver metastases were significantly associated with tumour grade, KRAS mutation and OS. 100 Another retrospective study investigated the ratio between the texture of colorectal liver metastases and the surrounding liver, and found that it may reflect tumour aggressiveness, chemotherapy response and OS. 101 However, Lee et al reported that texture features from liver parenchyma on portal venous phase CT cannot be used to predict the development of hepatic metastasis in colorectal cancer patients. 102 Apart from colorectal cancer, emerging evidence suggests that the CT‐based radiomics signature of esophagogastric liver metastases can help predict treatment response to chemotherapy. 27

5. FUTURE CHALLENGES AND OPPORTUNITIES

Current published studies revealed the potential of radiomics analysis in liver disease diagnosis, tumour biological property profiling, and prognosis estimation. However, although MR imaging can provide the multi‐parametric information regarding hepatic function and microenvironment with higher tissue resolution, most studies to date have focused on radiomics analyses of CT. 103 , 104 , 105 , 106 In addition, a large number of studies were retrospective in design and lack independent external validation across different geographical areas and races, which may limit the generalizability and applicability of the current findings. Different prevalence of disease may also influence the accuracy of the algorithm (eg positive and negative predictive values). Moreover radiomics results are extremely sensitive to the various technical acquisition parameters, especially among different vendors. Therefore, more large scale multi‐centre prospective studies with standardized acquisition, segmentation and imaging postprocessing are needed to ensure further development of radiomics in liver diseases.

6. CONCLUSIONS

Radiomics as a newly emerged quantitative technique is burgeoning in liver disease management with consistently developing methodology. Previous studies, although mainly retrospective in design and based on single imaging modality, have revealed its potential in diagnosis, treatment evaluation and prognosis prediction of several liver diseases. Nevertheless, further multi‐centre and prospective validation is still needed to valid its clinical usefulness, especially in prognosis‐related targets.

Current main obstacles for the application of radiomics in liver disease rely on high‐quality data collection and mechanism explanation on the biological basis. Multi‐institutional data sharing and intensive collaborations on data cleansing and labelling offer appeal in filling this gap. Artificial intelligence algorithms with improved accuracy and interpretability meanwhile need to be developed to facilitate broader translation and clinical adoption.

7. Financial information

This study has received funding by Ministry of Science and Technology of China under Grant No. 2017YFA0205200, National Natural Science Foundation of China under Grant No. 81227901 and 81527805, Chinese Academy of Sciences under Grant No. GJJSTD20170004 and QYZDJ‐SSW‐JSC005, Beijing Municipal Science & Technology Commission under Grant No. Z161100002616022 and 171100000117023.

CONFLICT OF INTEREST

None.

ACKNOWLEDGEMENTS

The authors appreciate the study participants, as well as researchers and staff.

Wei J, Jiang H, Gu D, et al. Radiomics in liver diseases: Current progress and future opportunities. Liver Int. 2020;40:2050–2063. 10.1111/liv.14555

Handling Editor: Luca Valenti

Jingwei Wei and Hanyu Jiang, and Dongsheng Gu contributed equally to this work.

REFERENCES

  • 1. Zhang YN, Fowler KJ, Hamilton G, et al. Liver fat imaging—a clinical overview of ultrasound, CT, and MR imaging[J]. Br J Radiol. 2018;91(1089):20170959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Ippolito D, Inchingolo R, Grazioli L, et al. Recent advances in non‐invasive magnetic resonance imaging assessment of hepatocellular carcinoma[J]. World J Gastroenterol. 2018;24(23):2413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Hope TA, Ohliger MA, Qayyum A. MR imaging of diffuse liver disease: from technique to diagnosis[J]. Radiologic Clinics. 2014;52(4):709‐724. [DOI] [PubMed] [Google Scholar]
  • 4. Ricke J, Seidensticker M. Molecular imaging and liver function assessment by hepatobiliary MRI[J]. J Hepatol. 2016;65(6):1081‐1082. [DOI] [PubMed] [Google Scholar]
  • 5. Pulli B, Wojtkiewicz G, Iwamoto Y, et al. Molecular MR imaging of myeloperoxidase distinguishes steatosis from steatohepatitis in nonalcoholic fatty liver disease[J]. Radiology. 2017;284(2):390‐400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Imajo K, Kessoku T, Honda Y, et al. Magnetic resonance imaging more accurately classifies steatosis and fibrosis in patients with nonalcoholic fatty liver disease than transient elastography[J]. Gastroenterology. 2016;150(3):626‐637.e7. [DOI] [PubMed] [Google Scholar]
  • 7. Limkin EJ, Sun R, Dercle L, et al. Promises and challenges for the implementation of computational medical imaging (radiomics) in oncology[J]. Ann Oncol. 2017;28(6):1191‐1206. [DOI] [PubMed] [Google Scholar]
  • 8. Lambin P, Leijenaar RTH, Deist TM, et al. Radiomics: the bridge between medical imaging and personalized medicine[J]. Nature Rev Clin Oncol. 2017;14(12):749. [DOI] [PubMed] [Google Scholar]
  • 9. Huang Y, Liang C, He L, et al. Development and validation of a radiomics nomogram for preoperative prediction of lymph node metastasis in colorectal cancer. J Clin Oncol. 2016;34(18):2157‐2164. [DOI] [PubMed] [Google Scholar]
  • 10. Liu Z, Li Z, Qu J, et al. Radiomics of multiparametric MRI for pretreatment prediction of pathologic complete response to neoadjuvant chemotherapy in breast cancer: a multicenter study[J]. Clin Cancer Res. 2019;25(12):3538‐3547. [DOI] [PubMed] [Google Scholar]
  • 11. Wang S, Shi J, Ye Z, et al. Predicting EGFR mutation status in lung adenocarcinoma on computed tomography image using deep learning[J]. Eur Respir J. 2019;53(3):1800986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Segal E, Sirlin CB, Ooi C, et al. Decoding global gene expression programs in liver cancer by noninvasive imaging[J]. Nat Biotechnol. 2007;25(6):675. [DOI] [PubMed] [Google Scholar]
  • 13. Zhou Y, He L, Huang Y, et al. CT‐based radiomics signature: a potential biomarker for preoperative prediction of early recurrence in hepatocellular carcinoma[J]. Abdominal Radiol. 2017;42(6):1695‐1704. [DOI] [PubMed] [Google Scholar]
  • 14. Cozzi L, Dinapoli N, Fogliata A, et al. Radiomics based analysis to predict local control and survival in hepatocellular carcinoma patients treated with volumetric modulated arc therapy[J]. BMC Cancer. 2017;17(1):829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Naganawa S, Enooku K, Tateishi R, et al. Imaging prediction of nonalcoholic steatohepatitis using computed tomography texture analysis[J]. Eur Radiol. 2018;28(7):3050‐3058. [DOI] [PubMed] [Google Scholar]
  • 16. Wang K, Lu X, Zhou H, et al. Deep learning Radiomics of shear wave elastography significantly improved diagnostic performance for assessing liver fibrosis in chronic hepatitis B: a prospective multicentre study[J]. Gut. 2019;68(4):729‐741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Peng J, Zhang J, Zhang Q, et al. A radiomics nomogram for preoperative prediction of microvascular invasion risk in hepatitis B virus‐related hepatocellular carcinoma[J]. Diagnostic Int Radiol. 2018;24(3):121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Reimer RP, Reimer P, Mahnken AH. Assessment of therapy response to transarterial radioembolization for liver metastases by means of post‐treatment MRI‐based texture analysis[J]. Cardiovasc Intervent Radiol. 2018;41(10):1545‐1556. [DOI] [PubMed] [Google Scholar]
  • 19. Akai H, Yasaka K, Kunimatsu A, et al. Predicting prognosis of resected hepatocellular carcinoma by radiomics analysis with random survival forest[J]. Diagnostic Interventional Imaging. 2018;99(10):643‐651. [DOI] [PubMed] [Google Scholar]
  • 20. Li W, Huang Y, Zhuang B‐W, et al. Multiparametric ultrasomics of significant liver fibrosis: a machine learning‐based analysis[J]. Eur Radiol. 2019;29(3):1496‐1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Hui T, Chuah TK, Low HM, et al. Predicting early recurrence of hepatocellular carcinoma with texture analysis of preoperative MRI: a radiomics study[J]. Clin Radiol. 2018;73(12):1056.e11‐1056.e16. [DOI] [PubMed] [Google Scholar]
  • 22. Kim J, Choi SJ, Lee S‐H, et al. Predicting survival using pretreatment CT for patients with hepatocellular carcinoma treated with transarterial chemoembolization: comparison of models using radiomics[J]. Am J Roentgenol. 2018;1026‐1034. [DOI] [PubMed] [Google Scholar]
  • 23. Liu F, Ning Z, Liu Y, et al. Development and validation of a radiomics signature for clinically significant portal hypertension in cirrhosis (CHESS1701): a prospective multicenter study[J]. EBioMedicine. 2018;36:151‐158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Wu M, Tan H, Gao F, et al. Predicting the grade of hepatocellular carcinoma based on non‐contrast‐enhanced MRI radiomics signature[J]. Eur Radiol. 2019;29(6):2802‐2811. [DOI] [PubMed] [Google Scholar]
  • 25. Yao Z, Dong YI, Wu G, et al. Preoperative diagnosis and prediction of hepatocellular carcinoma: radiomics analysis based on multi‐modal ultrasound images[J]. BMC Cancer. 2018;18(1):1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Hu H, Wang Z, Huang X, et al. Ultrasound‐based radiomics score: a potential biomarker for the prediction of microvascular invasion in hepatocellular carcinoma[J]. Eur Radiol. 2019, 29(6):2890‐2901. [DOI] [PubMed] [Google Scholar]
  • 27. Klaassen R, Larue RTHM, Mearadji B, et al. Feasibility of CT radiomics to predict treatment response of individual liver metastases in esophagogastric cancer patients[J]. PLoS One. 2018;13(11):e0207362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Zheng B‐H, Liu L‐Z, Zhang Z‐Z, et al. Radiomics score: a potential prognostic imaging feature for postoperative survival of solitary HCC patients[J]. BMC Cancer. 2018;18(1):1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Park HJ, Lee SS, Park B, et al. Radiomics Analysis of gadoxetic acid–enhanced MRI for staging liver fibrosis[J]. Radiology. 2018;290(2):380‐387. [DOI] [PubMed] [Google Scholar]
  • 30. Chen S, Feng S, Wei J, et al. Pretreatment prediction of immunoscore in hepatocellular cancer: a radiomics‐based clinical model based on Gd‐EOB‐DTPA‐enhanced MRI imaging[J]. Eur Radiol. 2019;29(8):4177‐4187. [DOI] [PubMed] [Google Scholar]
  • 31. Feng ST, Jia Y, Liao B, et al. Preoperative prediction of microvascular invasion in hepatocellular cancer: a radiomics model using Gd‐EOB‐DTPA‐enhanced MRI[J]. Eur Radiol. 2019;1‐12. [DOI] [PubMed] [Google Scholar]
  • 32. Ma X, Wei J, Gu D, et al. Preoperative radiomics nomogram for microvascular invasion prediction in hepatocellular carcinoma using contrast‐enhanced CT[J]. Eur Radiol. 2019;1‐11. [DOI] [PubMed] [Google Scholar]
  • 33. Shan Q‐Y, Hu H‐T, Feng S‐T, et al. CT‐based peritumoral radiomics signatures to predict early recurrence in hepatocellular carcinoma after curative tumor resection or ablation[J]. Cancer Imaging. 2019;19(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Cai W, He B, Hu M, et al. A radiomics‐based nomogram for the preoperative prediction of posthepatectomy liver failure in patients with hepatocellular carcinoma[J]. Surg Oncol. 2019;28:78‐85. [DOI] [PubMed] [Google Scholar]
  • 35. Wu J, Liu A, Cui J, et al. Radiomics‐based classification of hepatocellular carcinoma and hepatic haemangioma on precontrast magnetic resonance images[J]. BMC Med Imaging. 2019;19(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Xu X, Zhang H‐L, Liu Q‐P, et al. Radiomic analysis of contrast‐enhanced CT predicts microvascular invasion and outcome in hepatocellular carcinoma[J]. J Hepatol. 2019;70(6):1133‐1144. [DOI] [PubMed] [Google Scholar]
  • 37. Rahmim A, Bak‐Fredslund KP, Ashrafinia S, et al. Prognostic modeling for patients with colorectal liver metastases incorporating FDG PET radiomic features[J]. Eur J Radiol. 2019;113:101‐109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Yuan C, Wang Z, Gu D, et al. Prediction early recurrence of hepatocellular carcinoma eligible for curative ablation using a Radiomics nomogram[J]. Cancer Imaging. 2019;19(1):21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Zhang Z, Jiang H, Chen J, et al. Hepatocellular carcinoma: radiomics nomogram on gadoxetic acid‐enhanced MR imaging for early postoperative recurrence prediction[J]. Cancer Imaging. 2019;19(1):22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zhao LI, Ma X, Liang M, et al. Prediction for early recurrence of intrahepatic mass‐forming cholangiocarcinoma: quantitative magnetic resonance imaging combined with prognostic immunohistochemical markers[J]. Cancer Imaging. 2019;19(1):49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Guo D, Gu D, Wang H, et al. Radiomics analysis enables recurrence prediction for hepatocellular carcinoma after liver transplantation[J]. Eur J Radiol. 2019;117:33‐40. [DOI] [PubMed] [Google Scholar]
  • 42. Tseng Y, Ma L, Li S, et al. Application of CT‐based radiomics in predicting portal pressure and patient outcome in portal hypertension[J]. Eur J Radiol. 2020;108927. [DOI] [PubMed] [Google Scholar]
  • 43. Hectors SJ, Lewis S, Besa C, et al. MRI radiomics features predict immuno‐oncological characteristics of hepatocellular carcinoma[J]. Eur Radiol. 2020;1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Ni M, Zhou X, Lv Q, et al. Radiomics models for diagnosing microvascular invasion in hepatocellular carcinoma: which model is the best model?[J]. Cancer Imaging. 2019;19(1):60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Liao H, Zhang Z, Chen J, et al. Preoperative radiomic approach to evaluate tumor‐infiltrating CD8+ T cells in hepatocellular carcinoma patients using contrast‐enhanced computed tomography[J]. Ann Surg Oncol. 2019;26(13):4537‐4547. [DOI] [PubMed] [Google Scholar]
  • 46. Huang X, Long L, Wei J, et al. Radiomics for diagnosis of dual‐phenotype hepatocellular carcinoma using Gd‐EOB‐DTPA‐enhanced MRI and patient prognosis[J]. J Cancer Res Clin Oncol. 2019;145(12):2995‐3003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Shur J, Orton M, Connor A, et al. A clinical‐radiomic model for improved prognostication of surgical candidates with colorectal liver metastases[J]. J Surg Oncol. 2019;121(2):357‐364. [DOI] [PubMed] [Google Scholar]
  • 48. Jiang H, Liu X, Chen J, et al. Man or machine? Prospective comparison of the version 2018 EASL, LI‐RADS criteria and a radiomics model to diagnose hepatocellular carcinoma[J]. Cancer Imaging. 2019;19(1):84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Lehmann TM, Gonner C, Spitzer K. Survey: interpolation methods in medical image processing[J]. IEEE Trans Med Imaging. 1999;18(11):1049‐1075. [DOI] [PubMed] [Google Scholar]
  • 50. Lehmann T, Oberschelp W, Pelikan E, et al. Bildverarbeitung für die Medizin: Grundlagen, Modelle, Methoden, Anwendungen[M]. Berlin, Germany: Springer‐Verlag; 2013. [Google Scholar]
  • 51. Nyúl LG, Udupa JK, Zhang X. New variants of a method of MRI scale standardization[J]. IEEE Trans Med Imaging. 2000;19(2):143‐150. [DOI] [PubMed] [Google Scholar]
  • 52. Bağcı U, Udupa JK, Bai L. The role of intensity standardization in medical image registration[J]. Pattern Recognition Lett, 2010,31(4):315‐323. [Google Scholar]
  • 53. Shu Z, Fang S, Ding Z, et al. MRI‐based Radiomics nomogram to detect primary rectal cancer with synchronous liver metastases. Sci Rep. 2019;9(1):315‐323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Peng J, Qi X, Zhang Q, et al. A radiomics nomogram for preoperatively predicting prognosis of patients in hepatocellular carcinoma[J]. Translational Cancer Res. 2018;7(4):936‐946. [Google Scholar]
  • 55. Liang W, Xu L, Yang P, et al. Novel nomogram for preoperative prediction of early recurrence prediction in intrahepatic cholangiocarcinoma[J]. Front Oncol. 2018;8:360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Yang LI, Gu D, Wei J, et al. A Radiomics nomogram for preoperative prediction of microvascular invasion in hepatocellular carcinoma[J]. Liver Cancer. 2019;8(5):373‐386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Bakr S, Echegaray S, Shah R, et al. Noninvasive radiomics signature based on quantitative analysis of computed tomography images as a surrogate for microvascular invasion in hepatocellular carcinoma: a pilot study[J]. J Med Imaging. 2017;4(4):041303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Gao L, Heath DG, Kuszyk BS, et al. Automatic liver segmentation technique for three‐dimensional visualization of CT data[J]. Radiology. 1996;201(2):359‐364. [DOI] [PubMed] [Google Scholar]
  • 59. Massoptier L, Casciaro S. A new fully automatic and robust algorithm for fast segmentation of liver tissue and tumors from CT scans[J]. Eur Radiol. 2008;18(8):1658. [DOI] [PubMed] [Google Scholar]
  • 60. Moltz JH, Bornemann L, Kuhnigk J‐M, et al. Advanced segmentation techniques for lung nodules, liver metastases, and enlarged lymph nodes in CT scans[J]. IEEE J Selected Topics Signal Process. 2009;3(1):122‐134. [Google Scholar]
  • 61. Häme Y, Pollari M. Semi‐automatic liver tumor segmentation with hidden Markov measure field model and non‐parametric distribution estimation[J]. Med Image Anal. 2012;16(1):140‐149. [DOI] [PubMed] [Google Scholar]
  • 62. van Griethuysen JJM, Fedorov A, Parmar C, et al. Computational radiomics system to decode the radiographic phenotype[J]. Can Res. 2017;77(21):e104‐e107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Fu S, Wei J, Zhang J, et al. Selection between liver resection versus transarterial chemoembolization in hepatocellular carcinoma: a multicenter study[J]. Clin Trans Gastroenterol. 2019;10(8):e00070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Wang Q, Zhang L, Xie Y, et al. Malignancy characterization of hepatocellular carcinoma using hybrid texture and deep features[C]//2017. IEEE Int Conf Image Process (ICIP) IEEE. 2017;4162‐4166. [Google Scholar]
  • 65. Dou T, Zhou W. 2D and 3D convolutional neural network fusion for predicting the histological grade of hepatocellular carcinoma[C]//2018 24th International Conference on Pattern Recognition (ICPR). IEEE; 2018:3832‐3837. [Google Scholar]
  • 66. Chaudhary K, Poirion OB, Lu L, et al. Deep learning–based multi‐omics integration robustly predicts survival in liver cancer[J]. Clin Cancer Res. 2018;24(6):1248‐1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Guyon I, Elisseeff A. An introduction to variable and feature selection[J]. J Machine Learning Res. 2003;3(Mar):1157‐1182. [Google Scholar]
  • 68. Benesty J, Chen J, Huang Y, et al. Pearson correlation coefficient[M]//Noise reduction in speech processing. Berlin, Heidelberg: Springer; 2009:1‐4. [Google Scholar]
  • 69. Ruxton GD. The unequal variance t‐test is an underused alternative to Student's t‐test and the Mann‐Whitney U test[J]. Behav Ecol. 2006;17(4):688‐690. [Google Scholar]
  • 70. Greenwood PE, Nikulin MS. A guide to chi‐squared testing[M]. New Jersey: John Wiley & Sons; 1996. [Google Scholar]
  • 71. Tibshirani R. Regression shrinkage and selection via the lasso[J]. J Roy Stat Soc: Ser B (Methodol). 1996;58(1):267‐288. [Google Scholar]
  • 72. Ji GW, Zhu FP, Zhang YD, et al. A radiomics approach to predict lymph node metastasis and clinical outcome of intrahepatic cholangiocarcinoma[J]. Eur Radiol. 2019;29 3725‐3735. [DOI] [PubMed] [Google Scholar]
  • 73. Marrero JA, Kulik LM, Sirlin CB, et al. Diagnosis, staging, and management of hepatocellular carcinoma: 2018 practice guidance by the American Association for the Study of Liver Diseases[J]. Hepatology. 2018;68(2):723‐750. [DOI] [PubMed] [Google Scholar]
  • 74. Joo I, Lee JM, Yoon JH. Imaging diagnosis of intrahepatic and perihilar cholangiocarcinoma: recent advances and challenges[J]. Radiology. 2018;288(1):7‐13. [DOI] [PubMed] [Google Scholar]
  • 75. Potretzke TA, Tan BR, Doyle MB, et al. Imaging features of biphenotypic primary liver carcinoma (hepatocholangiocarcinoma) and the potential to mimic hepatocellular carcinoma: LI‐RADS analysis of CT and MRI features in 61 cases[J]. Am J Roentgenol. 2016;207(1):25‐31. [DOI] [PubMed] [Google Scholar]
  • 76. Li Z, Mao YU, Huang W, et al. Texture‐based classification of different single liver lesion based on SPAIR T2W MRI images[J]. BMC Med Imaging. 2017;17(1):42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Trivizakis E, Manikis GC, Nikiforaki K, et al. Extending 2‐D convolutional neural networks to 3‐D for advancing deep learning cancer classification with application to MRI liver tumor differentiation[J]. IEEE J Biomed Health Informatics. 2018;23(3):923‐930. [DOI] [PubMed] [Google Scholar]
  • 78. Tang AN, Destrempes F, Kazemirad S, et al. Quantitative ultrasound and machine learning for assessment of steatohepatitis in a rat model[J]. Eur Radiol. 2019;29(5):2175‐2184. [DOI] [PubMed] [Google Scholar]
  • 79. Choi KJ, Jang JK, Lee SS, et al. Development and validation of a deep learning system for staging liver fibrosis by using contrast agent–enhanced CT images in the liver[J]. Radiology. 2018;289(3):688‐697. [DOI] [PubMed] [Google Scholar]
  • 80. Ng IOL, Lai ECS, Fan ST, et al. Prognostic significance of pathologic features of hepatocellular carcinoma a multivariate analysis of 278 patients[J]. Cancer. 1995;76(12):2443‐2448. [DOI] [PubMed] [Google Scholar]
  • 81. Suh S‐W, Lee K‐W, Lee J‐M, et al. Prediction of aggressiveness in early‐stage hepatocellular carcinoma for selection of surgical resection[J]. J Hepatol. 2014;60(6):1219‐1224. [DOI] [PubMed] [Google Scholar]
  • 82. Okusaka T, Okada S, Ueno H, et al. Satellite lesions in patients with small hepatocellular carcinoma with reference to clinicopathologic features[J]. Cancer: Interdisciplinary Int J Am Cancer Soc. 2002;95(9):1931‐1937. [DOI] [PubMed] [Google Scholar]
  • 83. Witjes CDM, Willemssen FEJA, Verheij J, et al. Histological differentiation grade and microvascular invasion of hepatocellular carcinoma predicted by dynamic contrast‐enhanced MRI[J]. J Magn Reson Imaging. 2012;36(3):641‐647. [DOI] [PubMed] [Google Scholar]
  • 84. Zhou WU, Zhang L, Wang K, et al. Malignancy characterization of hepatocellular carcinomas based on texture analysis of contrast‐enhanced MR images[J]. J Magn Reson Imaging. 2017;45(5):1476‐1484. [DOI] [PubMed] [Google Scholar]
  • 85. Ye Z, Jiang H, Chen J, et al. Texture analysis on gadoxetic acid enhanced‐MRI for predicting Ki‐67 status in hepatocellular carcinoma: A prospective study[J]. Chinese J Cancer Res. 2019;31(5):806‐817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Schöniger‐Hekele M, Müller C, Kutilek M, et al. Hepatocellular carcinoma in Central Europe: prognostic features and survival[J]. Gut. 2001;48(1):103‐109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Jiang H‐Y, Chen J, Xia C‐C, et al. Noninvasive imaging of hepatocellular carcinoma: From diagnosis to prognosis[J]. World J Gastroenterol. 2018;24(22):2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Canellas R, Mehrkhani F, Patino M, et al. Characterization of portal vein thrombosis (neoplastic versus bland) on CT images using software‐based texture analysis and thrombus density (Hounsfield units)[J]. Am J Roentgenol. 2016;207(5):W81‐W87. [DOI] [PubMed] [Google Scholar]
  • 89. Bakr SH, Echegaray S, Shah RP, et al. Noninvasive radiomics signature based on quantitative analysis of computed tomography images as a surrogate for microvascular invasion in hepatocellular carcinoma: a pilot study[J]. J Med I. 2017;4(4):041303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Galle PR, Forner A, Llovet JM, et al. EASL clinical practice guidelines: management of hepatocellular carcinoma[J]. J Hepatol. 2018;69(1):182‐236. [DOI] [PubMed] [Google Scholar]
  • 91. Li M, Fu S, Zhu Y, et al. Computed tomography texture analysis to facilitate therapeutic decision making in hepatocellular carcinoma[J]. Oncotarget. 2016;7(11):13248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Chen S, Zhu Y, Liu Z, et al. Texture analysis of baseline multiphasic hepatic computed tomography images for the prognosis of single hepatocellular carcinoma after hepatectomy: a retrospective pilot study[J]. Eur J Radiol. 2017;90:198‐204. [DOI] [PubMed] [Google Scholar]
  • 93. Kiryu S, Akai H, Nojima M, et al. Impact of hepatocellular carcinoma heterogeneity on computed tomography as a prognostic indicator[J]. Sci Rep. 2017;7(1):12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Mulé S, Thiefin G, Costentin C, et al. Advanced hepatocellular carcinoma: pretreatment contrast‐enhanced CT texture parameters as predictive biomarkers of survival in patients treated with sorafenib[J]. Radiology. 2018;288(2):445‐455. [DOI] [PubMed] [Google Scholar]
  • 95. Sun R, Limkin EJ, Vakalopoulou M, et al. A radiomics approach to assess tumour‐infiltrating CD8 cells and response to anti‐PD‐1 or anti‐PD‐L1 immunotherapy: an imaging biomarker, retrospective multicohort study[J]. Lancet Oncol. 2018;19(9):1180‐1191. [DOI] [PubMed] [Google Scholar]
  • 96. Chen S, Feng S, Xiao H, et al. Pretreatment prediction of Immunoscore in hepatocellular cancer: a radiomics‐based clinical nomogram based on Gd‐EOB‐DTPA enhanced‐MRI[J]. Cancer Biol Med. 2018;15(Suppl 1):2. [DOI] [PubMed] [Google Scholar]
  • 97. Tyson GL, El‐Serag HB. Risk factors for cholangiocarcinoma[J]. Hepatology. 2011;54(1):173‐184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Guglielmi A, Ruzzenente A, Campagnaro T, et al. Intrahepatic cholangiocarcinoma: prognostic factors after surgical resection[J]. World J Surg. 2009;33(6):1247‐1254. [DOI] [PubMed] [Google Scholar]
  • 99. Ji G‐W, Zhang Y‐D, Zhang H, et al. Biliary tract Cancer at CT: a Radiomics‐based model to predict lymph node metastasis and survival outcomes[J]. Radiology. 2018;290(1):90‐98. [DOI] [PubMed] [Google Scholar]
  • 100. Lubner MG, Stabo N, Lubner SJ, et al. CT textural analysis of hepatic metastatic colorectal cancer: pre‐treatment tumor heterogeneity correlates with pathology and clinical outcomes[J]. Abdom Imaging. 2015;40(7):2331‐2337. [DOI] [PubMed] [Google Scholar]
  • 101. Beckers R, Trebeschi S, Maas M, et al. CT texture analysis in colorectal liver metastases and the surrounding liver parenchyma and its potential as an imaging biomarker of disease aggressiveness, response and survival[J]. Eur J Radiol. 2018;102:15‐21. [DOI] [PubMed] [Google Scholar]
  • 102. Lee SJ, Zea R, Kim DH, et al. CT texture features of liver parenchyma for predicting development of metastatic disease and overall survival in patients with colorectal cancer[J]. Eur Radiol. 2018;28(4):1520‐1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Chernyak V, Fowler KJ, Kamaya A, et al. Liver Imaging Reporting and Data System (LI‐RADS) version 2018: imaging of hepatocellular carcinoma in at‐risk patients[J]. Radiology. 2018;289(3):816‐830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Taouli B, Koh DM. Diffusion‐weighted MR imaging of the liver[J]. Radiology. 2009;254(1):47‐66. [DOI] [PubMed] [Google Scholar]
  • 105. Motosugi U, Bannas P, Sano K, et al. Hepatobiliary MR contrast agents in hypovascular hepatocellular carcinoma[J]. J Magn Reson Imaging. 2015;41(2):251‐265. [DOI] [PubMed] [Google Scholar]
  • 106. Choi JY, Lee JM, Sirlin CB. CT and MR imaging diagnosis and staging of hepatocellular carcinoma: part II. Extracellular agents, hepatobiliary agents, and ancillary imaging features[J]. Radiology. 2014;273(1):30‐50. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Liver International are provided here courtesy of Wiley

RESOURCES